1
|
Karakaya C, van Turnhout M, van den Hurk EAN, Bouten CVC, Sahlgren CM, Loerakker S. Equibiaxial and uniaxial cyclic strain similarly affect Notch signaling and vascular smooth muscle cell phenotype in 2D. Integr Biol (Camb) 2025; 17:zyaf007. [PMID: 40434193 DOI: 10.1093/intbio/zyaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/16/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
Vascular smooth muscle cells (VSMCs) play a crucial role in vascular growth and remodeling by adapting their phenotype in response to biomechanical cues. The Notch signaling pathway, known for its sensitivity to mechanical forces, is a regulator of strain-induced phenotypic plasticity of VSMCs. However, the impact of the intricate mechanical environment within the vessel wall on Notch signaling and VSMCs is not completely elucidated. In this study, we investigated the influence of strain anisotropy, which is important for understanding (patho)physiological mechanical conditions, on mechanosensitive Notch signaling and subsequent changes in VSMC phenotype. Using varying amplitudes of cyclic strain in the physiological range, we examined the effects of equibiaxial and uniaxial strain on Notch signaling and phenotypic transitions in synthetic and contractile VSMCs. Additionally, we compared cell responses between equibiaxial and uniaxial loading conditions by analyzing three different deformation characteristics to determine the primary strain measure governing Notch signaling and VSMC phenotype. Our findings reveal that both cyclic equibiaxial and uniaxial strain downregulate Notch signaling and contractile characteristics of VSMCs. Notably, these reductions are most similar for both loading conditions when the maximum principal strain values were compared. Overall, our results suggest that VSMCs respond in a comparable manner to equibiaxial and uniaxial strain, indicating that strain anisotropy may not significantly influence Notch signaling or phenotypic switching of VSMCs. Insight Box: Vascular smooth muscle cells (VSMCs) adapt their phenotype during vascular growth and remodeling in response to mechanical cues. The Notch signaling pathway, sensitive to mechanical stimuli, regulates this phenotypic plasticity. However, the effect of strain anisotropy, which is important for understanding (patho)physiological mechanical conditions, on Notch signaling and subsequent changes in VSMC phenotype is not clear. Understanding this relationship is crucial to determine how VSMC phenotype, contributing to vascular growth and remodeling, is regulated in physiological and pathological hemodynamic environments. Here, we showed that both equibiaxial and uniaxial strain downregulate Notch signaling components and the contractile properties of VSMCs. Our findings further highlighted the maximum principal strain as the dominant mechanical parameter influencing Notch signaling and VSMC phenotypic changes.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Receptors, Notch/metabolism
- Stress, Mechanical
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Signal Transduction
- Phenotype
- Animals
- Cells, Cultured
- Mechanotransduction, Cellular/physiology
- Anisotropy
- Rats
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
| | - Mark van Turnhout
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
| | - Eva A N van den Hurk
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, 20500, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, the Netherlands
| |
Collapse
|
2
|
Yang J, Wang P, Zhang Y, Zhang M, Sun Q, Chen H, Dong L, Chu Z, Xue B, Hoff WD, Zhao C, Wang W, Wei Q, Cao Y. Photo-tunable hydrogels reveal cellular sensing of rapid rigidity changes through the accumulation of mechanical signaling molecules. Cell Stem Cell 2025; 32:121-136.e6. [PMID: 39437791 DOI: 10.1016/j.stem.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Cells use traction forces to sense mechanical cues in their environment. While the molecular clutch model effectively explains how cells exert more forces on stiffer substrates, it falls short in addressing their adaptation to dynamic mechanical fluctuations prevalent in tissues and organs. Here, using hydrogel with photo-responsive rigidity, we show that cells' response to rigidity changes is frequency dependent. Strikingly, at certain frequencies, cellular traction forces exceed those on static substrates 4-fold stiffer, challenging the established molecular clutch model. We discover that the discrepancy between the rapid adaptation of traction forces and the slower deactivation of mechanotransduction signaling proteins results in their accumulation, thereby enhancing long-term cellular traction in dynamic settings. Consequently, we propose a new model that melds immediate mechanosensing with extended mechanical signaling. Our study underscores the significance of dynamic rigidity in the development of synthetic biomaterials, emphasizing the importance of considering both immediate and prolonged cellular responses.
Collapse
Affiliation(s)
- Jiapeng Yang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Yu Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Man Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Huiyan Chen
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Liang Dong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 999077, China; Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Bin Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Wouter David Hoff
- Department of Physics, Oklahoma State University, Stillwater, OK 74078, USA
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Wei Wang
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China.
| | - Yi Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| |
Collapse
|
3
|
Khanmohammadi M, Danish H, Sekar NC, Suarez SA, Chheang C, Peter K, Khoshmanesh K, Baratchi S. Cyclic stretch enhances neutrophil extracellular trap formation. BMC Biol 2024; 22:209. [PMID: 39289752 PMCID: PMC11409804 DOI: 10.1186/s12915-024-02009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Neutrophils, the most abundant leukocytes circulating in blood, contribute to host defense and play a significant role in chronic inflammatory disorders. They can release their DNA in the form of extracellular traps (NETs), which serve as scaffolds for capturing bacteria and various blood cells. However, uncontrolled formation of NETs (NETosis) can lead to excessive activation of coagulation pathways and thrombosis. Once neutrophils are migrated to infected or injured tissues, they become exposed to mechanical forces from their surrounding environment. However, the impact of transient changes in tissue mechanics due to the natural process of aging, infection, tissue injury, and cancer on neutrophils remains unknown. To address this gap, we explored the interactive effects of changes in substrate stiffness and cyclic stretch on NETosis. Primary neutrophils were cultured on a silicon-based substrate with stiffness levels of 30 and 300 kPa for at least 3 h under static conditions or cyclic stretch levels of 5% and 10%, mirroring the biomechanics of aged and young arteries. RESULTS Using this approach, we found that neutrophils are sensitive to cyclic stretch and that increases in stretch intensity and substrate stiffness enhance nuclei decondensation and histone H3 citrullination (CitH3). In addition, stretch intensity and substrate stiffness promote the response of neutrophils to the NET-inducing agents phorbol 12-myristate 13-acetate (PMA), adenosine triphosphate (ATP), and lipopolysaccharides (LPS). Stretch-induced activation of neutrophils was dependent on calpain activity, the phosphatidylinositol 3-kinase (PI3K)/focal adhesion kinase (FAK) signalling and actin polymerization. CONCLUSIONS In summary, these results demonstrate that the mechanical forces originating from the surrounding tissue influence NETosis, an important neutrophil function, and thus identify a potential novel therapeutic target.
Collapse
Affiliation(s)
- Manijeh Khanmohammadi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Habiba Danish
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Chanly Chheang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Karlheinz Peter
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Sala S, Caillier A, Oakes PW. Principles and regulation of mechanosensing. J Cell Sci 2024; 137:jcs261338. [PMID: 39297391 PMCID: PMC11423818 DOI: 10.1242/jcs.261338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
Research over the past two decades has highlighted that mechanical signaling is a crucial component in regulating biological processes. Although many processes and proteins are termed 'mechanosensitive', the underlying mechanisms involved in mechanosensing can vary greatly. Recent studies have also identified mechanosensing behaviors that can be regulated independently of applied force. This important finding has major implications for our understanding of downstream mechanotransduction, the process by which mechanical signals are converted into biochemical signals, as it offers another layer of biochemical regulatory control for these crucial signaling pathways. In this Review, we discuss the different molecular and cellular mechanisms of mechanosensing, how these processes are regulated and their effects on downstream mechanotransduction. Together, these discussions provide an important perspective on how cells and tissues control the ways in which they sense and interpret mechanical signals.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Alexia Caillier
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| |
Collapse
|
5
|
Carrara E, Soliveri L, Poloni S, Bozzetto M, Campiglio CE. Effects of high-frequency mechanical stimuli on flow related vascular cell biology. Int J Artif Organs 2024; 47:590-601. [PMID: 39166431 PMCID: PMC11487902 DOI: 10.1177/03913988241268105] [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: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
Mechanical forces related to blood pressure and flow patterns play a crucial role in vascular homeostasis. Perturbations in vascular stresses and strain resulting from changes in hemodynamic may occur in pathological conditions, leading to vascular dysfunction as well as in vascular prosthesis, arteriovenous shunt for hemodialysis and in mechanical circulation support. Turbulent-like blood flows can induce high-frequency vibrations of the vessel wall, and this stimulus has recently gained attention as potential contributors to vascular pathologies, such as development of intimal hyperplasia in arteriovenous fistula for hemodialysis. However, the biological response of vascular cells to this stimulus remains incompletely understood. This review provides an analysis of the existing literature concerning the impact of high-frequency stimuli on vascular cell morphology, function, and gene expression. Morphological and functional investigations reveal that vascular cells stimulated at frequencies higher than the normal heart rate exhibit alterations in cell shape, alignment, and proliferation, potentially leading to vessel remodeling. Furthermore, vibrations modulate endothelial and smooth muscle cells gene expression, affecting pathways related to inflammation, oxidative stress, and muscle hypertrophy. Understanding the effects of high-frequency vibrations on vascular cells is essential for unraveling the mechanisms underlying vascular diseases and identifying potential therapeutic targets. Nevertheless, there are still gaps in our understanding of the molecular pathways governing these cellular responses. Further research is necessary to elucidate these mechanisms and their therapeutic implications for vascular diseases.
Collapse
Affiliation(s)
- Elena Carrara
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Luca Soliveri
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Sofia Poloni
- Department of Engineering and Applied Sciences, University of Bergamo, Dalmine, Italy
| | - Michela Bozzetto
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Chiara Emma Campiglio
- Department of Management, Information and Production Engineering, University of Bergamo, Dalmine, Italy
| |
Collapse
|
6
|
Ren C, Chang Z, Li K, Wang X, Wang D, Xu Y, Li X, Li Q. Impact of uniaxial cyclic stretching on matrix-associated endothelial cell responses. Mater Today Bio 2024; 27:101152. [PMID: 39104901 PMCID: PMC11298614 DOI: 10.1016/j.mtbio.2024.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/07/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Uniaxial cyclic stretching plays a pivotal role in the fields of tissue engineering and regenerative medicine, influencing cell behaviors and functionality based on physical properties, including matrix morphology and mechanical stimuli. This study delves into the response of endothelial cells to uniaxial cyclic strain within the geometric constraints of micro-nano fibers. Various structural scaffold forms of poly(l-lactide-co-caprolactone) (PLCL), such as flat membranes, randomly oriented fiber membranes, and aligned fiber membranes, were fabricated through solvent casting and electrospinning methods. Our investigation focuses on the morphological variation of endothelial cells under diverse geometric constraints and the mechanical-dependent release of nitric oxide (NO) on oriented fibrous membranes. Our results indicate that while uniaxial cyclic stretching promotes endothelial cell spreading, the anisotropy of the matrix morphology remains the primary driving factor for cell alignment. Additionally, uniaxial cyclic stretching significantly enhances NO release, with a notably stronger effect correlated to the increasing strain amplitude. Importantly, this study reveals that uniaxial cyclic stretching enhances the mRNA expression of key proteins, including talin, vinculin, rac, and nitric oxide synthase (eNOS).
Collapse
Affiliation(s)
- Cuihong Ren
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Zhonghua Chang
- Institute of Laser Manufacturing, Henan Academy of Sciences, Zhengzhou, 450046, PR China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Dongfang Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yiyang Xu
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, PR China
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou, 450001, PR China
| |
Collapse
|
7
|
Nietupski CA, Moset Zupan A, Schutte SC. Impact of Cyclic Strain on Elastin Synthesis in a 3D Human Myometrial Culture Model. Tissue Eng Part C Methods 2024; 30:279-288. [PMID: 38943281 DOI: 10.1089/ten.tec.2024.0038] [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: 07/01/2024] Open
Abstract
The synthesis and assembly of mature, organized elastic fibers remains a limitation to the clinical use of many engineered tissue replacements. There is a critical need for a more in-depth understanding of elastogenesis regulation for the advancement of methods to induce and guide production of elastic matrix structures in engineered tissues that meet the structural and functional requirements of native tissue. The dramatic increase in elastic fibers through normal pregnancy has led us to explore the potential role of mechanical stretch in combination with pregnancy levels of the steroid hormones 17β-estradiol and progesterone on elastic fiber production by human uterine myometrial smooth muscle cells in a three-dimensional (3D) culture model. Opposed to a single strain regimen, we sought to better understand how the amplitude and frequency parameters of cyclic strain influence elastic fiber production in these myometrial tissue constructs (MTC). Mechanical stretch was applied to MTC at a range of strain amplitudes (5%, 10%, and 15% at 0.5 Hz frequency) and frequencies (0.1 Hz, 0.5 Hz, 1 Hz, and constant 0 Hz at 10% amplitude), with and without pregnancy-level hormones, for 6 days. MTC were assessed for cell proliferation, matrix elastin protein content, and expression of the main elastic fiber genes, tropoelastin (ELN) and fibrillin-1 (FBN1). Significant increases in elastin protein and ELN and FBN1 mRNA were produced from samples subjected to a 0.5 Hz, 10% strain regimen, as well as samples stretched at higher amplitude (15%, 0.5 Hz) and higher frequency (1 Hz, 10%); however, no significant effects because of third-trimester mimetic hormone treatment were determined. These results establish that a minimum level of strain is required to stimulate the synthesis of elastic fiber components in our culture model and show this response can be similarly enhanced by increasing either the amplitude or frequency parameter of applied strain. Further, our results demonstrate strain alone is sufficient to stimulate elastic fiber production and suggest hormones may not be a significant factor in regulating elastin synthesis. This 3D culture model will provide a useful tool to further investigate mechanisms underlying pregnancy-induced de novo elastic fiber synthesis and assembly by uterine smooth muscle cells.
Collapse
Affiliation(s)
- Carolyn A Nietupski
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andreja Moset Zupan
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stacey C Schutte
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
8
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
9
|
Coppini A, Falconieri A, Mualem O, Nasrin SR, Roudon M, Saper G, Hess H, Kakugo A, Raffa V, Shefi O. Can repetitive mechanical motion cause structural damage to axons? Front Mol Neurosci 2024; 17:1371738. [PMID: 38912175 PMCID: PMC11191579 DOI: 10.3389/fnmol.2024.1371738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Biological structures have evolved to very efficiently generate, transmit, and withstand mechanical forces. These biological examples have inspired mechanical engineers for centuries and led to the development of critical insights and concepts. However, progress in mechanical engineering also raises new questions about biological structures. The past decades have seen the increasing study of failure of engineered structures due to repetitive loading, and its origin in processes such as materials fatigue. Repetitive loading is also experienced by some neurons, for example in the peripheral nervous system. This perspective, after briefly introducing the engineering concept of mechanical fatigue, aims to discuss the potential effects based on our knowledge of cellular responses to mechanical stresses. A particular focus of our discussion are the effects of mechanical stress on axons and their cytoskeletal structures. Furthermore, we highlight the difficulty of imaging these structures and the promise of new microscopy techniques. The identification of repair mechanisms and paradigms underlying long-term stability is an exciting and emerging topic in biology as well as a potential source of inspiration for engineers.
Collapse
Affiliation(s)
| | | | - Oz Mualem
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| | - Syeda Rubaiya Nasrin
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | - Marine Roudon
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Gadiel Saper
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Akira Kakugo
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | | | - Orit Shefi
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
10
|
Chen Y, Topo EJ, Nan B, Chen J. Mathematical modeling of mechanosensitive reversal control in Myxococcus xanthus. Front Microbiol 2024; 14:1294631. [PMID: 38260904 PMCID: PMC10803039 DOI: 10.3389/fmicb.2023.1294631] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Adjusting motility patterns according to environmental cues is important for bacterial survival. Myxococcus xanthus, a bacterium moving on surfaces by gliding and twitching mechanisms, modulates the reversal frequency of its front-back polarity in response to mechanical cues like substrate stiffness and cell-cell contact. In this study, we propose that M. xanthus's gliding machinery senses environmental mechanical cues during force generation and modulates cell reversal accordingly. To examine our hypothesis, we expand an existing mathematical model for periodic polarity reversal in M. xanthus, incorporating the experimental data on the intracellular dynamics of the gliding machinery and the interaction between the gliding machinery and a key polarity regulator. The model successfully reproduces the dependence of cell reversal frequency on substrate stiffness observed in M. xanthus gliding. We further propose reversal control networks between the gliding and twitching motility machineries to explain the opposite reversal responses observed in wild type M. xanthus cells that possess both motility mechanisms. These results provide testable predictions for future experimental investigations. In conclusion, our model suggests that the gliding machinery in M. xanthus can function as a mechanosensor, which transduces mechanical cues into a cell reversal signal.
Collapse
Affiliation(s)
- Yirui Chen
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Genetics, Bioinformatics and Computational Biology Graduate Program, Virginia Tech, Blacksburg, VA, United States
| | - Elias J. Topo
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Beiyan Nan
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Jing Chen
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
- Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
11
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [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: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
12
|
Giverso C, Loy N, Lucci G, Preziosi L. Cell orientation under stretch: A review of experimental findings and mathematical modelling. J Theor Biol 2023; 572:111564. [PMID: 37391125 DOI: 10.1016/j.jtbi.2023.111564] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/15/2023] [Indexed: 07/02/2023]
Abstract
The key role of electro-chemical signals in cellular processes had been known for many years, but more recently the interplay with mechanics has been put in evidence and attracted substantial research interests. Indeed, the sensitivity of cells to mechanical stimuli coming from the microenvironment turns out to be relevant in many biological and physiological circumstances. In particular, experimental evidence demonstrated that cells on elastic planar substrates undergoing periodic stretches, mimicking native cyclic strains in the tissue where they reside, actively reorient their cytoskeletal stress fibres. At the end of the realignment process, the cell axis forms a certain angle with the main stretching direction. Due to the importance of a deeper understanding of mechanotransduction, such a phenomenon was studied both from the experimental and the mathematical modelling point of view. The aim of this review is to collect and discuss both the experimental results on cell reorientation and the fundamental features of the mathematical models that have been proposed in the literature.
Collapse
Affiliation(s)
- Chiara Giverso
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10126, Italy.
| | - Nadia Loy
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10126, Italy.
| | - Giulio Lucci
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10126, Italy.
| | - Luigi Preziosi
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10126, Italy.
| |
Collapse
|
13
|
Lien JC, Wang YL. Cyclic stretching combined with cell-cell adhesion is sufficient for inducing cell intercalation. Biophys J 2023; 122:3146-3158. [PMID: 37408306 PMCID: PMC10432222 DOI: 10.1016/j.bpj.2023.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 02/09/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023] Open
Abstract
Although the important role of cell intercalation within a collective has long been recognized particularly for morphogenesis, the underlying mechanism remains poorly understood. Here we investigate the possibility that cellular responses to cyclic stretching play a major role in this process. By applying synchronized imaging and cyclic stretching to epithelial cells cultured on micropatterned polyacrylamide (PAA) substrates, we discovered that uniaxial cyclic stretching induces cell intercalation along with cell shape change and cell-cell interfacial remodeling. The process involved intermediate steps as previously reported for cell intercalation during embryonic morphogenesis, including the appearance of cell vertices, anisotropic vertex resolution, and directional expansion of cell-cell interface. Using mathematical modeling, we further found that cell shape change in conjunction with dynamic cell-cell adhesions was sufficient to account for the observations. Further investigation with small-molecule inhibitors indicated that disruption of myosin II activities suppressed cyclic stretching-induced intercalation while inhibiting the appearance of oriented vertices. Inhibition of Wnt signaling did not suppress stretch-induced cell shape change but disrupted cell intercalation and vertex resolution. Our results suggest that cyclic stretching, by inducing cell shape change and reorientation in the presence of dynamic cell-cell adhesions, can induce at least some aspects of cell intercalation and that this process is dependent in distinct ways on myosin II activities and Wnt signaling.
Collapse
Affiliation(s)
- Jui-Chien Lien
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Yu-Li Wang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
14
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 140] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
15
|
Colombi A, Preziosi L, Scianna M. Modelling Cell Orientation Under Stretch: The Effect of Substrate Elasticity. Bull Math Biol 2023; 85:79. [PMID: 37460873 PMCID: PMC10352433 DOI: 10.1007/s11538-023-01180-1] [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: 12/29/2022] [Accepted: 06/19/2023] [Indexed: 07/20/2023]
Abstract
When cells are seeded on a cyclically deformed substrate like silicon, they tend to reorient their major axis in two ways: either perpendicular to the main stretching direction, or forming an oblique angle with it. However, when the substrate is very soft such as a collagen gel, the oblique orientation is no longer observed, and the cells align either along the stretching direction, or perpendicularly to it. To explain this switch, we propose a simplified model of the cell, consisting of two elastic elements representing the stress fiber/focal adhesion complexes in the main and transverse directions. These elements are connected by a torsional spring that mimics the effect of crosslinking molecules among the stress fibers, which resist shear forces. Our model, consistent with experimental observations, predicts that there is a switch in the asymptotic behaviour of the orientation of the cell determined by the stiffness of the substratum, related to a change from a supercritical bifurcation scenario, whereby the oblique configuration is stable for a sufficiently large stiffness, to a subcritical bifurcation scenario at a lower stiffness. Furthermore, we investigate the effect of cell elongation and find that the region of the parameter space leading to an oblique orientation decreases as the cell becomes more elongated. This implies that elongated cells, such as fibroblasts and smooth muscle cells, are more likely to maintain an oblique orientation with respect to the main stretching direction. Conversely, rounder cells, such as those of epithelial or endothelial origin, are more likely to switch to a perpendicular or parallel orientation on soft substrates.
Collapse
Affiliation(s)
- Annachiara Colombi
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129, Turin, Italy
| | - Luigi Preziosi
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129, Turin, Italy.
| | - Marco Scianna
- Department of Mathematical Sciences "G.L. Lagrange", Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129, Turin, Italy
| |
Collapse
|
16
|
Soto Veliz D, Lin K, Sahlgren C. Organ-on-a-chip technologies for biomedical research and drug development: A focus on the vasculature. SMART MEDICINE 2023; 2:e20220030. [PMID: 37089706 PMCID: PMC7614466 DOI: 10.1002/smmd.20220030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/20/2023] [Indexed: 04/25/2023]
Abstract
Current biomedical models fail to replicate the complexity of human biology. Consequently, almost 90% of drug candidates fail during clinical trials after decades of research and billions of investments in drug development. Despite their physiological similarities, animal models often misrepresent human responses, and instead, trigger ethical and societal debates regarding their use. The overall aim across regulatory entities worldwide is to replace, reduce, and refine the use of animal experimentation, a concept known as the Three Rs principle. In response, researchers develop experimental alternatives to improve the biological relevance of in vitro models through interdisciplinary approaches. This article highlights the emerging organ-on-a-chip technologies, also known as microphysiological systems, with a focus on models of the vasculature. The cardiovascular system transports all necessary substances, including drugs, throughout the body while in charge of thermal regulation and communication between other organ systems. In addition, we discuss the benefits, limitations, and challenges in the widespread use of new biomedical models. Coupled with patient-derived induced pluripotent stem cells, organ-on-a-chip technologies are the future of drug discovery, development, and personalized medicine.
Collapse
Affiliation(s)
- Diosangeles Soto Veliz
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Kai‐Lan Lin
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Cecilia Sahlgren
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
17
|
Navaee F, Renaud P, Kleger A, Braschler T. Highly Efficient Cardiac Differentiation and Maintenance by Thrombin-Coagulated Fibrin Hydrogels Enriched with Decellularized Porcine Heart Extracellular Matrix. Int J Mol Sci 2023; 24:2842. [PMID: 36769166 PMCID: PMC9917900 DOI: 10.3390/ijms24032842] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Biochemical and biophysical properties instruct cardiac tissue morphogenesis. Here, we are reporting on a blend of cardiac decellularized extracellular matrix (dECM) from porcine ventricular tissue and fibrinogen that is suitable for investigations employing an in vitro 3D cardiac cell culture model. Rapid and specific coagulation with thrombin facilitates the gentle inclusion of cells while avoiding sedimentation during formation of the dECM-fibrin composite. Our investigations revealed enhanced cardiogenic differentiation in the H9c2 myoblast cells when using the system in a co-culture with Nor-10 fibroblasts. Further enhancement of differentiation efficiency was achieved by 3D embedding of rat neonatal cardiomyocytes in the 3D system. Calcium imaging and analysis of beating motion both indicate that the dECM-fibrin composite significantly enhances recovery, frequency, synchrony, and the maintenance of spontaneous beating, as compared to various controls including Matrigel, pure fibrin and collagen I as well as a fibrin-collagen I blend.
Collapse
Affiliation(s)
- Fatemeh Navaee
- Microsystems Laboratory-LMIS4, EPFL, 1015 Lausanne, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, CMU, 1211 Geneva, Switzerland
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, 89081 Ulm, Germany
| | - Philippe Renaud
- Microsystems Laboratory-LMIS4, EPFL, 1015 Lausanne, Switzerland
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, 89081 Ulm, Germany
- Interdisciplinary Pancreatology, Department of Internal Medicine 1, Ulm University Hospital, 89081 Ulm, Germany
- Organoid Core Facility, Medical Faculty, Ulm University Hospital, 89081 Ulm, Germany
| | - Thomas Braschler
- Department of Pathology and Immunology, Faculty of Medicine, CMU, 1211 Geneva, Switzerland
| |
Collapse
|
18
|
Fang X, Ni K, Guo J, Li Y, Zhou Y, Sheng H, Bu B, Luo M, Ouyang M, Deng L. FRET Visualization of Cyclic Stretch-Activated ERK via Calcium Channels Mechanosensation While Not Integrin β1 in Airway Smooth Muscle Cells. Front Cell Dev Biol 2022; 10:847852. [PMID: 35663392 PMCID: PMC9162487 DOI: 10.3389/fcell.2022.847852] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/05/2022] [Indexed: 12/19/2022] Open
Abstract
Mechanical stretch is one type of common physiological activities such as during heart beating, lung breathing, blood flow through the vessels, and physical exercise. The mechanical stimulations regulate cellular functions and maintain body homeostasis. It still remains to further characterize the mechanical-biomechanical coupling mechanism. Here we applied fluorescence resonance energy transfer (FRET) technology to visualize ERK activity in airway smooth muscle (ASM) cells under cyclic stretch stimulation in airway smooth muscle (ASM) cells, and studied the mechanosensing pathway. FRET measurements showed apparent ERK activation by mechanical stretch, which was abolished by ERK inhibitor PD98059 pretreatment. Inhibition of extracellular Ca2+ influx reduced ERK activation, and selective inhibition of inositol 1,4,5-trisphosphate receptor (IP3R) Ca2+ channel or SERCA Ca2+ pump on endoplasmic reticulum (ER) blocked the activation. Chemical inhibition of the L-type or store-operated Ca2+ channels on plasma membrane, or inhibition of integrin β1 with siRNA had little effect on ERK activation. Disruption of actin cytoskeleton but not microtubule one inhibited the stretch-induced ERK activation. Furthermore, the ER IP3R-dependent ERK activation was not dependent on phospholipase C-IP3 signal, indicating possibly more mechanical mechanism for IP3R activation. It is concluded from our study that the mechanical stretch activated intracellular ERK signal in ASM cells through membrane Ca2+ channels mechanosensation but not integrin β1, which was mediated by actin cytoskeleton.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Linhong Deng
- *Correspondence: Mingxing Ouyang, ; Linhong Deng,
| |
Collapse
|
19
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
20
|
Aguilera Suarez S, Sekar NC, Nguyen N, Lai A, Thurgood P, Zhou Y, Needham S, Pirogova E, Khoshmanesh K, Baratchi S. Studying the Mechanobiology of Aortic Endothelial Cells Under Cyclic Stretch Using a Modular 3D Printed System. Front Bioeng Biotechnol 2021; 9:791116. [PMID: 34957080 PMCID: PMC8698250 DOI: 10.3389/fbioe.2021.791116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Here, we describe a motorized cam-driven system for the cyclic stretch of aortic endothelial cells. Our modular design allows for generating customized spatiotemporal stretch profiles by varying the profile and size of 3D printed cam and follower elements. The system is controllable, compact, inexpensive, and amenable for parallelization and long-term experiments. Experiments using human aortic endothelial cells show significant changes in the cytoskeletal structure and morphology of cells following exposure to 5 and 10% cyclic stretch over 9 and 16 h. The system provides upportunities for exploring the complex molecular and cellular processes governing the response of mechanosensitive cells under cyclic stretch.
Collapse
Affiliation(s)
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Austin Lai
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Ying Zhou
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | | | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | | | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
22
|
Cook BL, Chao CJ, Alford PW. Architecture-Dependent Mechano-Adaptation in Single Vascular Smooth Muscle Cells. J Biomech Eng 2021; 143:101002. [PMID: 33972987 DOI: 10.1115/1.4051117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 01/03/2023]
Abstract
Arteries grow and remodel following mechanical perturbation. Vascular smooth muscle cells (VSMCs) within the artery undergo hyperplasia, hypertrophy, or change their contractility following sustained changes in loading. Experimental evidence in vivo and in vitro suggests that VSMCs grow and remodel to maintain a constant transmural stress, or "target" stress. This behavior is often described using a stress-dependent finite growth framework. Typically, computational models of arterial growth and remodeling account for VSMC behavior in a constrained mixture formulation that incorporates behavior of each component of the artery. However, these models do not account for differential VSMC architecture observed in situ, which may significantly influence growth and remodeling behavior. Here, we used cellular microbiaxial stretching (CμBS) to characterize how VSMCs with different cytoskeletal architectures respond to a sustained step change in strain. We find that VSMC F-actin architecture becomes more aligned following stretch and retains this alignment after 24 h. Further, we find that VSMC stress magnitude depends on cellular architecture. Qualitatively, however, stress behavior following stretch is consistent across cell architectures-stress increases following stretch and returns to prestretch magnitudes after 24 h. Finally, we formulated an architecture-dependent targeted growth law that accounts for experimentally measured cytoskeletal alignment and attributes stress evolution to individual fiber growth and find that this model robustly captures long-term stress evolution in single VSMCs. These results suggest that VSMC mechano-adaptation depends on cellular architecture, which has implications for growth and remodeling in regions of arteries with differential architecture, such as at bifurcations.
Collapse
Affiliation(s)
- Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Christina J Chao
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| |
Collapse
|
23
|
Cyclic stretching-induced epithelial cell reorientation is driven by microtubule-modulated transverse extension during the relaxation phase. Sci Rep 2021; 11:14803. [PMID: 34285275 PMCID: PMC8292395 DOI: 10.1038/s41598-021-93987-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/29/2021] [Indexed: 11/29/2022] Open
Abstract
Many types of adherent cells are known to reorient upon uniaxial cyclic stretching perpendicularly to the direction of stretching to facilitate such important events as wound healing, angiogenesis, and morphogenesis. While this phenomenon has been documented for decades, the underlying mechanism remains poorly understood. Using an on-stage stretching device that allowed programmable stretching with synchronized imaging, we found that the reorientation of NRK epithelial cells took place primarily during the relaxation phase when cells underwent rapid global retraction followed by extension transverse to the direction of stretching. Inhibition of myosin II caused cells to orient along the direction of stretching, whereas disassembly of microtubules enhanced transverse reorientation. Our results indicate distinct roles of stretching and relaxation in cell reorientation and implicate a role of myosin II-dependent contraction via a microtubule-modulated mechanism. The importance of relaxation phase also explains the difference between the responses to cyclic and static stretching.
Collapse
|
24
|
Andreu I, Falcones B, Hurst S, Chahare N, Quiroga X, Le Roux AL, Kechagia Z, Beedle AEM, Elosegui-Artola A, Trepat X, Farré R, Betz T, Almendros I, Roca-Cusachs P. The force loading rate drives cell mechanosensing through both reinforcement and cytoskeletal softening. Nat Commun 2021; 12:4229. [PMID: 34244477 PMCID: PMC8270983 DOI: 10.1038/s41467-021-24383-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/15/2021] [Indexed: 01/08/2023] Open
Abstract
Cell response to force regulates essential processes in health and disease. However, the fundamental mechanical variables that cells sense and respond to remain unclear. Here we show that the rate of force application (loading rate) drives mechanosensing, as predicted by a molecular clutch model. By applying dynamic force regimes to cells through substrate stretching, optical tweezers, and atomic force microscopy, we find that increasing loading rates trigger talin-dependent mechanosensing, leading to adhesion growth and reinforcement, and YAP nuclear localization. However, above a given threshold the actin cytoskeleton softens, decreasing loading rates and preventing reinforcement. By stretching rat lungs in vivo, we show that a similar phenomenon may occur. Our results show that cell sensing of external forces and of passive mechanical parameters (like tissue stiffness) can be understood through the same mechanisms, driven by the properties under force of the mechanosensing molecules involved.
Collapse
Affiliation(s)
- Ion Andreu
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | | | - Sebastian Hurst
- Institute of Cell Biology, Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Nimesh Chahare
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat Politècnica de Catalunya (UPC), Campus Nord, Barcelona, Spain
| | - Xarxa Quiroga
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Anabel-Lise Le Roux
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Zanetta Kechagia
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Amy E M Beedle
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Department of Physics, King's College London, Strand, London, UK
| | - Alberto Elosegui-Artola
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Boston, MA, USA
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Ramon Farré
- Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Timo Betz
- Institute of Cell Biology, Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Isaac Almendros
- Universitat de Barcelona, Barcelona, Spain.
- CIBER de Enfermedades Respiratorias, Madrid, Spain.
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain.
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain.
- Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
25
|
Decoding mechanical cues by molecular mechanotransduction. Curr Opin Cell Biol 2021; 72:72-80. [PMID: 34218181 DOI: 10.1016/j.ceb.2021.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 12/29/2022]
Abstract
Cells are exposed to a variety of mechanical cues, including forces from their local environment and physical properties of the tissue. These mechanical cues regulate a vast number of cellular processes, relying on a repertoire of mechanosensors that transduce forces into biochemical pathways through mechanotransduction. Forces can act on different parts of the cell, carry information regarding magnitude and direction, and have distinct temporal profiles. Thus, the specific cellular response to mechanical forces is dependent on the ability of cells to sense and transduce these physical parameters. In this review, we will highlight recent findings that provide insights into the mechanisms by which different mechanosensors decode mechanical cues and how their coordinated response determines the cellular outcomes.
Collapse
|
26
|
Lucci G, Giverso C, Preziosi L. Cell orientation under stretch: Stability of a linear viscoelastic model. Math Biosci 2021; 337:108630. [PMID: 34015301 DOI: 10.1016/j.mbs.2021.108630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
The sensitivity of cells to alterations in the microenvironment and in particular to external mechanical stimuli is significant in many biological and physiological circumstances. In this regard, experimental assays demonstrated that, when a monolayer of cells cultured on an elastic substrate is subject to an external cyclic stretch with a sufficiently high frequency, a reorganization of actin stress fibres and focal adhesions happens in order to reach a stable equilibrium orientation, characterized by a precise angle between the cell major axis and the largest strain direction. To examine the frequency effect on the orientation dynamics, we propose a linear viscoelastic model that describes the coupled evolution of the cellular stress and the orientation angle. We find that cell orientation oscillates tending to an angle that is predicted by the minimization of a very general orthotropic elastic energy, as confirmed by a bifurcation analysis. Moreover, simulations show that the speed of convergence towards the predicted equilibrium orientation presents a changeover related to the viscous-elastic transition for viscoelastic materials. In particular, when the imposed oscillation period is lower than the characteristic turnover rate of the cytoskeleton and of adhesion molecules such as integrins, reorientation is significantly faster.
Collapse
Affiliation(s)
- Giulio Lucci
- Department of Mathematical Sciences "G.L. Lagrange" Dipartimento di Eccellenza 2018-2022, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; Department of Mathematics "G. Peano", Università degli Studi di Torino, Via Carlo Alberto 10, 10123 Turin, Italy.
| | - Chiara Giverso
- Department of Mathematical Sciences "G.L. Lagrange" Dipartimento di Eccellenza 2018-2022, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy.
| | - Luigi Preziosi
- Department of Mathematical Sciences "G.L. Lagrange" Dipartimento di Eccellenza 2018-2022, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy.
| |
Collapse
|
27
|
Uniaxially fixed mechanical boundary condition elicits cellular alignment in collagen matrix with induction of osteogenesis. Sci Rep 2021; 11:9009. [PMID: 33907271 PMCID: PMC8079399 DOI: 10.1038/s41598-021-88505-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Osteocytes differentiated from osteoblasts play significant roles as mechanosensors in modulating the bone remodeling process. While the well-aligned osteocyte network along the trabeculae with slender cell processes perpendicular to the trabeculae surface is known to facilitate the sensing of mechanical stimuli by cells and the intracellular communication in the bone matrix, the mechanisms underlying osteocyte network formation remains unclear. Here, we developed a novel in vitro collagen matrix system exerting a uniaxially-fixed mechanical boundary condition on which mouse osteoblast-like MC3T3-E1 cells were subcultured, evoking cellular alignment along the uniaxial boundary condition. Using a myosin II inhibitor, blebbistatin, we showed that the intracellular tension via contraction of actin fibers contributed to the cellular alignment under the influence of isometric matrix condition along the uniaxially-fixed mechanical boundary condition. Furthermore, the cells actively migrated inside the collagen matrix and promoted the expression of osteoblast and osteocyte genes with their orientations aligned along the uniaxially-fixed boundary condition. Collectively, our results suggest that the intracellular tension of osteoblasts under a uniaxially-fixed mechanical boundary condition is one of the factors that determines the osteocyte alignment inside the bone matrix.
Collapse
|
28
|
Mao T, He Y, Gu Y, Yang Y, Yu Y, Wang X, Ding J. Critical Frequency and Critical Stretching Rate for Reorientation of Cells on a Cyclically Stretched Polymer in a Microfluidic Chip. ACS APPLIED MATERIALS & INTERFACES 2021; 13:13934-13948. [PMID: 33739805 DOI: 10.1021/acsami.0c21186] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The ability of cells to sense and respond to mechanical signals from their surrounding microenvironments is one of the key issues in tissue engineering and regeneration, yet a fundamental study of cells with both cell observation and mechanical stimulus is challenging and should be based upon an appropriate microdevice. Herein we designed and fabricated a two-layer microfluidic chip to enable simultaneous observation of live cells and cyclic stretching of an elastic polymer, polydimethylsiloxane (PDMS), with a modified surface for enhanced cell adhesion. Human mesenchymal stem cells (hMSCs) were examined with a series of frequencies from 0.00003 to 2 Hz and varied amplitudes of 2%, 5%, or 10%. The cells with an initial random orientation were confirmed to be reoriented perpendicular to the stretching direction at frequencies greater than a threshold value, which we term critical frequency (fc); additionally, the critical frequency fc was amplitude-dependent. We further introduced the concept of critical stretching rate (Rc) and found that this quantity can unify both frequency and amplitude dependences. The reciprocal value of Rc in this study reads 8.3 min, which is consistent with the turnover time of actin filaments reported in the literature, suggesting that the supramolecular relaxation in the cytoskeleton within a cell might be responsible for the underlying cell mechanotransduction. The theoretical calculation of cell reorientation based on a two-dimensional tensegrity model under uniaxial cyclic stretching is well consistent with our experiments. The above findings provide new insight into the crucial role of critical frequency and critical stretching rate in regulating cells on biomaterials under biomechanical stimuli.
Collapse
Affiliation(s)
- Tianjiao Mao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yingning He
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yexin Gu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yuqian Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yue Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Xinlei Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
29
|
A Fully Integrated Arduino-Based System for the Application of Stretching Stimuli to Living Cells and Their Time-Lapse Observation: A Do-It-Yourself Biology Approach. Ann Biomed Eng 2021; 49:2243-2259. [PMID: 33728867 DOI: 10.1007/s10439-021-02758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
Mechanobiology has nowadays acquired the status of a topic of fundamental importance in a degree in Biological Sciences. It is inherently a multidisciplinary topic where biology, physics and engineering competences are required. A course in mechanobiology should include lab experiences where students can appreciate how mechanical stimuli from outside affect living cell behaviour. Here we describe all the steps to build a cell stretcher inside an on-stage cell incubator. This device allows exposing living cells to a periodic mechanical stimulus similar to what happens in physiological conditions such as, for example, in the vascular system or in the lungs. The reaction of the cells to the periodic mechanical stretching represents a prototype of a mechanobiological signal integrated by living cells. We also provide the theoretical and experimental aspects related to the calibration of the stretcher apparatus at a level accessible to researchers not used to dealing with topics like continuum mechanics and analysis of deformations. We tested our device by stretching cells of two different lines, U87-MG and Balb-3T3 cells, and we analysed and discussed the effect of the periodic stimulus on both cell reorientation and migration. We also discuss the basic aspects related to the quantitative analysis of the reorientation process and of cell migration. We think that the device we propose can be easily reproduced at low-cost within a project-oriented course in the fields of biology, biotechnology and medical engineering.
Collapse
|
30
|
Ghavamian A, Mousavi SJ, Avril S. Computational Study of Growth and Remodeling in Ascending Thoracic Aortic Aneurysms Considering Variations of Smooth Muscle Cell Basal Tone. Front Bioeng Biotechnol 2020; 8:587376. [PMID: 33224937 PMCID: PMC7670047 DOI: 10.3389/fbioe.2020.587376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/28/2020] [Indexed: 11/16/2022] Open
Abstract
In this paper, we investigate the progression of Ascending Thoracic Aortic Aneurysms (ATAA) using a computational model of Growth and Remodeling (G&R) taking into account the composite (elastin, four collagen fiber families and Smooth Muscle Cells—SMCs) and multi-layered (media and adventitia) nature of the aorta. The G&R model, which is based on the homogenized Constrained Mixture theory, is implemented as a UMAT in the Abaqus finite-element package. Each component of the mixture is assigned a strain energy density function: nearly-incompressible neo-Hookean for elastin and Fung-type for collagen and SMCs. Active SMCs tension is additionally considered, through a length-tension relationship having a classic inverted parabola shape, in order to investigate its effects on the progression of ATAA in a patient-specific model. A sensitivity analysis is performed to evaluate the potential impact of variations in the parameters of the length-tension relationships. These variations reflect in variations of SMCs normal tone during ATAA progression, with active stress contributions ranging between 30% (best case scenario) and 0% (worst case scenario) of the total wall circumferential stress. Low SMCs active stress in the worst case scenarios, in fact, affect the rates of collagen deposition by which the elastin loss is gradually compensated by collagen deposition in the simulated ATAA progression, resulting eventually in larger aneurysm diameters. The types of length-tension relationships leading to a drop of SMCs active stress in our simulations reveal a critical condition which could also result in SMCs apoptosis.
Collapse
Affiliation(s)
- Ataollah Ghavamian
- Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Étienne, France
| | - S Jamaleddin Mousavi
- Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Étienne, France
| | - Stéphane Avril
- Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Étienne, France
| |
Collapse
|
31
|
Mierke CT. Mechanical Cues Affect Migration and Invasion of Cells From Three Different Directions. Front Cell Dev Biol 2020; 8:583226. [PMID: 33043017 PMCID: PMC7527720 DOI: 10.3389/fcell.2020.583226] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Cell migration and invasion is a key driving factor for providing essential cellular functions under physiological conditions or the malignant progression of tumors following downward the metastatic cascade. Although there has been plentiful of molecules identified to support the migration and invasion of cells, the mechanical aspects have not yet been explored in a combined and systematic manner. In addition, the cellular environment has been classically and frequently assumed to be homogeneous for reasons of simplicity. However, motility assays have led to various models for migration covering only some aspects and supporting factors that in some cases also include mechanical factors. Instead of specific models, in this review, a more or less holistic model for cell motility in 3D is envisioned covering all these different aspects with a special emphasis on the mechanical cues from a biophysical perspective. After introducing the mechanical aspects of cell migration and invasion and presenting the heterogeneity of extracellular matrices, the three distinct directions of cell motility focusing on the mechanical aspects are presented. These three different directions are as follows: firstly, the commonly used invasion tests using structural and structure-based mechanical environmental signals; secondly, the mechano-invasion assay, in which cells are studied by mechanical forces to migrate and invade; and thirdly, cell mechanics, including cytoskeletal and nuclear mechanics, to influence cell migration and invasion. Since the interaction between the cell and the microenvironment is bi-directional in these assays, these should be accounted in migration and invasion approaches focusing on the mechanical aspects. Beyond this, there is also the interaction between the cytoskeleton of the cell and its other compartments, such as the cell nucleus. In specific, a three-element approach is presented for addressing the effect of mechanics on cell migration and invasion by including the effect of the mechano-phenotype of the cytoskeleton, nucleus and the cell's microenvironment into the analysis. In precise terms, the combination of these three research approaches including experimental techniques seems to be promising for revealing bi-directional impacts of mechanical alterations of the cellular microenvironment on cells and internal mechanical fluctuations or changes of cells on the surroundings. Finally, different approaches are discussed and thereby a model for the broad impact of mechanics on cell migration and invasion is evolved.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
32
|
Tyson J, Bundy K, Roach C, Douglas H, Ventura V, Segars MF, Schwartz O, Simpson CL. Mechanisms of the Osteogenic Switch of Smooth Muscle Cells in Vascular Calcification: WNT Signaling, BMPs, Mechanotransduction, and EndMT. Bioengineering (Basel) 2020; 7:bioengineering7030088. [PMID: 32781528 PMCID: PMC7552614 DOI: 10.3390/bioengineering7030088] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 12/16/2022] Open
Abstract
Characterized by the hardening of arteries, vascular calcification is the deposition of hydroxyapatite crystals in the arterial tissue. Calcification is now understood to be a cell-regulated process involving the phenotypic transition of vascular smooth muscle cells into osteoblast-like cells. There are various pathways of initiation and mechanisms behind vascular calcification, but this literature review highlights the wingless-related integration site (WNT) pathway, along with bone morphogenic proteins (BMPs) and mechanical strain. The process mirrors that of bone formation and remodeling, as an increase in mechanical stress causes osteogenesis. Observing the similarities between the two may aid in the development of a deeper understanding of calcification. Both are thought to be regulated by the WNT signaling cascade and bone morphogenetic protein signaling and can also be activated in response to stress. In a pro-calcific environment, integrins and cadherins of vascular smooth muscle cells respond to a mechanical stimulus, activating cellular signaling pathways, ultimately resulting in gene regulation that promotes calcification of the vascular extracellular matrix (ECM). The endothelium is also thought to contribute to vascular calcification via endothelial to mesenchymal transition, creating greater cell plasticity. Each of these factors contributes to calcification, leading to increased cardiovascular mortality in patients, especially those suffering from other conditions, such as diabetes and kidney failure. Developing a better understanding of the mechanisms behind calcification may lead to the development of a potential treatment in the future.
Collapse
|
33
|
Yamashita T, Nishina T, Matsushita I, Sudo R. Air-pressure-driven Separable Microdevice to Control the Anisotropic Curvature of Cell Culture Surface. ANAL SCI 2020; 36:1015-1019. [PMID: 32201406 DOI: 10.2116/analsci.20a001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We report on a novel microdevice to tune the curvature of a cell-adhering surface by controlling the air-pressure and micro-slit. Human aortic smooth muscle cells were cultured on demi-cylindrical concaves formed on a microdevice. Their shape-adapting behavior could be tracked when the groove direction was changed to the orthogonal direction. This microdevice demonstrated live observation of cells responding to dynamic changes of the anisotropic curvature of the adhering surface and could serve as a new platform to pursue mechanobiology on curved surfaces.
Collapse
Affiliation(s)
| | - Takuya Nishina
- Department of System Design Engineering, Keio University
| | | | - Ryo Sudo
- Department of System Design Engineering, Keio University
| |
Collapse
|
34
|
Molina JJ, Yamamoto R. Modeling the mechanosensitivity of fast-crawling cells on cyclically stretched substrates. SOFT MATTER 2019; 15:683-698. [PMID: 30623962 DOI: 10.1039/c8sm01903g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The mechanosensitivity of cells, which determines how they are able to respond to mechanical signals, is crucial for the functioning of biological systems. Experimentally, this is investigated by studying the reorientation of cells on cyclically stretched substrates. The reorientation depends on the type of cell and on the stretching protocol, but the mechanisms responsible for the response are still not completely understood. Here, we introduce a computational model for fast crawling cells on cyclically stretched substrates that accounts for the sub-cellular elements responsible for cell shape and motility. This includes the dynamics of the cell membrane, the actin cytoskeleton, and the focal adhesions with the stretching substrate. These processes evolve over characteristic time scales that can vary by orders of magnitude and naturally give rise to the frequency dependent reorientation observed experimentally. Depending on which processes are being probed by the stretching and on the type of coupling with the substrate, our simulations predict either no reorientation, a bi-stability in the parallel and perpendicular directions, or a complete reorientation in either the parallel or perpendicular direction. In particular, we show that an asymmetry in the adhesion dynamics during the loading and unloading phases of the stretching, whether it comes from the response of the cell itself or from the precise stretching protocol, can be used to selectively align the cells. Our results provide further evidence for the importance of focal adhesion dynamics in determining the mechanosensitive response of cells, as well as a way to interpret recent experiments.
Collapse
Affiliation(s)
- John J Molina
- Department of Chemical Engineering, Kyoto University, Kyoto, Japan.
| | - Ryoichi Yamamoto
- Department of Chemical Engineering, Kyoto University, Kyoto, Japan. and Institute of Industrial Science, The University of Tokyo, Tokyo, 153-8505, Japan
| |
Collapse
|
35
|
Lévesque L, Loy C, Lainé A, Drouin B, Chevallier P, Mantovani D. Incrementing the Frequency of Dynamic Strain on SMC-Cellularised Collagen-Based Scaffolds Affects Extracellular Matrix Remodeling and Mechanical Properties. ACS Biomater Sci Eng 2018; 4:3759-3767. [PMID: 33429603 DOI: 10.1021/acsbiomaterials.7b00395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Notwithstanding the efforts injected in vascular tissue engineering in the past 30 years, the clinical translation of engineered artery constructs is far from being successful. One common approach to improve artery regeneration is the use of cyclic mechanical stimuli to guide cellular remodeling. However, there is a lack of information on the effect of cyclic strain on cells within a 3D environment. To this end, this work explored the effect of gradual increase in stimulation frequency on the response of human umbilical artery smooth muscle cells (HUASMCs) embedded in a 3D collagen matrix. The results demonstrate that, with an applied strain of 5%, the gradual increase of frequency from 0.1 to 1 Hz improved collagen remodeling by HUASMCs compared to samples constantly stimulated at 1 Hz. The expression of collagen, elastin and matrix metalloproteinase-2 (MMP-2) genes was similar at 7 days for gradual and 1 Hz samples which showed lower amounts than static counterparts. Interestingly the mechanical properties of the constructs, specifically the amplitude of the time constants and the elastic equilibrium modulus, were enhanced by gradual increase of frequency. Taken together, these results show an increase in collagen remodeling by the HUASMCs overtime under incremental cyclic mechanical strain. This work suggests that only the in-depth investigation of the effects of stimulation parameters on the behavior of vSMC under cyclic strain in a 3D environment could lead to the design of optimized control strategies for enhanced vascular tissue generation and maturation in bioreactors.
Collapse
Affiliation(s)
- L Lévesque
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | - C Loy
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | - A Lainé
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | - B Drouin
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | - P Chevallier
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | - D Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met- Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Université Laval, Quebec, Quebec, Canada G1V 0A6
| |
Collapse
|
36
|
Venugopal B, Mogha P, Dhawan J, Majumder A. Cell density overrides the effect of substrate stiffness on human mesenchymal stem cells' morphology and proliferation. Biomater Sci 2018. [PMID: 29528341 PMCID: PMC5933002 DOI: 10.1039/c7bm00853h] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The effect of substrate stiffness on the cellular morphology, proliferation, and differentiation of human mesenchymal stem cells (hMSCs) has been extensively researched and well established. However, the majority of these studies are done with a low seeding density where cell to cell interactions do not play a significant role. While these conditions permit an analysis of cell-substratum interactions at the single cell level, such a model system fails to capture a critical aspect of the cellular micro-environment in vivo, i.e. the cell-cell interaction via matrix deformation (i.e., strain). To address this question, we seeded hMSCs on soft poly-acrylamide (PAA) gels, at a seeding density that permits cells to be mechanically interacting via the underlying substrate. We found that as the intercellular distance decreases with the increasing seeding density, cellular sensitivity towards the substrate rigidity becomes significantly diminished. With the increasing seeding density, the cell spread area increased on a soft substrate (500 Pa) but reduced on an even slightly stiffer substrate (2 kPa) as well as on glass making them indistinguishable at a high seeding density. Not only in terms of cell spread area but also at a high seeding density, cells formed mature focal adhesions and prominent stress fibres on a soft substrate similar to that of the cells being cultured on a stiff substrate. The decreased intercellular distance also influenced the proliferation rate of the cells: higher seeding density on the soft substrate showed cell cycle progression similar to that of the cells on glass substrates. In summary, this paper demonstrates how the effect of substrate rigidity on the cell morphology and fate is a function of inter-cellular distance when seeded on a soft substrate. Our AFM data suggest that such changes happen due to local strain stiffening of the soft PAA gel, an effect that has been rarely reported in the literature so far.
Collapse
Affiliation(s)
- Balu Venugopal
- Institute of Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India.
| | | | | | | |
Collapse
|
37
|
Holzapfel GA, Ogden RW. Biomechanical relevance of the microstructure in artery walls with a focus on passive and active components. Am J Physiol Heart Circ Physiol 2018; 315:H540-H549. [DOI: 10.1152/ajpheart.00117.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The microstructure of arteries, consisting, in particular, of collagen, elastin, and vascular smooth muscle cells, plays a very significant role in their biomechanical response during a cardiac cycle. In this article, we highlight the microstructure and the contributions of each of its components to the overall mechanical behavior. We also describe the changes of the microstructure that occur as a result of abdominal aortic aneurysms and disease, such as atherosclerosis. We also focus on how the passive and active constituents are incorporated into a mathematical model without going into detail of the mathematical formulation. We conclude by mentioning open problems toward a better characterization of the biomechanical aspects of arteries that will be beneficial for a better understanding of cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Norwegian University of Science and Technology, Faculty of Engineering Science and Technology, Trondheim, Norway
| | - Ray W. Ogden
- School of Mathematics and Statistics, University of Glasgow, Scotland, United Kingdom
| |
Collapse
|
38
|
Wu T, Zhang J, Wang Y, Sun B, Yin M, Bowlin GL, Mo X. Design and Fabrication of a Biomimetic Vascular Scaffold Promoting in Situ Endothelialization and Tunica Media Regeneration. ACS APPLIED BIO MATERIALS 2018; 1:833-844. [DOI: 10.1021/acsabm.8b00269] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Tong Wu
- State Key Lab for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Jialing Zhang
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Yuanfei Wang
- State Key Laboratory of Bioreactor Engineering, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Binbin Sun
- State Key Lab for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Meng Yin
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Gary L. Bowlin
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee 38017, United States
| | - Xiumei Mo
- State Key Lab for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
39
|
Cyclic Mechanical Stretch Up-regulates Hepatoma-Derived Growth Factor Expression in Cultured Rat Aortic Smooth Muscle Cells. Biosci Rep 2018; 38:BSR20171398. [PMID: 29467272 PMCID: PMC5857908 DOI: 10.1042/bsr20171398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/14/2018] [Accepted: 02/21/2018] [Indexed: 11/19/2022] Open
Abstract
Hepatoma-derived growth factor (HDGF) is a potent mitogen for vascular smooth muscle cells (SMCs) during embryogenesis and injury repair of vessel walls. Whether mechanical stimuli modulate HDGF expression remains unknown. The present study aimed at investigating whether cyclic mechanical stretch plays a regulatory role in HDGF expression and regenerative cytokine production in aortic SMCs. A SMC cell line was grown on a silicone-based elastomer chamber with extracellular matrix coatings (either type I collagen or fibronectin) and received cyclic and uniaxial mechanical stretches with 10% deformation at frequency 1 Hz. Morphological observation showed that fibronectin coating provided better cell adhesion and spreading and that consecutive 6 h of cyclic mechanical stretch remarkably induced reorientation and realignment of SMCs. Western blotting detection demonstrated that continuous mechanical stimuli elicited up-regulation of HDGF and proliferative cell nuclear antigen, a cell proliferative marker. Signal kinetic profiling study indicated that cyclic mechanical stretch induced signaling activity in RhoA/ROCK and PI3K/Akt cascades. Kinase inhibition study further showed that blockade of PI3K activity suppressed the stretch-induced tumor necrosis factor-α (TNF-α), whereas RhoA/ROCK inhibition significantly blunted the interleukin-6 (IL-6) production and HDGF overexpression. Moreover, siRNA-mediated HDGF gene silencing significantly suppressed constitutive expression of IL-6, but not TNF-α, in SMCs. These findings support the role of HDGF in maintaining vascular expression of IL-6, which has been regarded a crucial regenerative factor for acute vascular injury. In conclusion, cyclic mechanical stretch may maintain constitutive expression of HDGF in vascular walls and be regarded an important biophysical regulator in vascular regeneration.
Collapse
|
40
|
Profiles of long noncoding RNAs in hypertensive rats: long noncoding RNA XR007793 regulates cyclic strain-induced proliferation and migration of vascular smooth muscle cells. J Hypertens 2017; 35:1195-1203. [PMID: 28319593 DOI: 10.1097/hjh.0000000000001304] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are being discovered in multiple diseases at a rapid pace. However, the contribution of lncRNAs to hypertension remains largely unknown. In hypertension, the vascular walls are exposed to abnormal mechanical cyclic strain, which leads to vascular remodelling. Here, we investigated the mechanobiological role of lncRNAs in hypertension. METHODS AND RESULTS Differences in the lncRNAs and mRNAs between spontaneously hypertensive rats and Wistar-Kyoto rats were screened using a gene microarray. The results showed that 68 lncRNAs and 255 mRNAs were upregulated in the aorta of spontaneously hypertensive rats, whereas 167 lncRNAs and 272 mRNAs were downregulated. Expressions of the screened lncRNAs, including XR007793, were validated by real-time PCR. A coexpression network was composed, and gene function was analysed using Ingenuity Pathway Analysis. In vitro, vascular smooth muscle cells (VSMCs) were subjected to cyclic strain at a magnitude of 5 (physiological normotensive cyclic strain) or 15% (pathological hypertensive cyclic strain) by Flexcell-4000T. A total of 15% cyclic strain increased XR007793 expression. XR007793 knockdown attenuated VSMC proliferation and migration and inhibited coexpressed genes such as signal transducers and activators of transcription 2 (stat2), LIM domain only 2 (lmo2) and interferon regulatory factor 7 (irf7). CONCLUSION The profile of lncRNAs was varied in response to hypertension, and pathological elevated cyclic strain may play crucial roles during this process. Our data revealed a novel mechanoresponsive lncRNA-XR007793, which modulates VSMC proliferation and migration, and participates in vascular remodelling during hypertension.
Collapse
|
41
|
Bade ND, Kamien RD, Assoian RK, Stebe KJ. Curvature and Rho activation differentially control the alignment of cells and stress fibers. SCIENCE ADVANCES 2017; 3:e1700150. [PMID: 28913421 PMCID: PMC5587136 DOI: 10.1126/sciadv.1700150] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 08/10/2017] [Indexed: 05/27/2023]
Abstract
In vivo, cells respond to a host of physical cues ranging from substrate stiffness to the organization of micro- and nanoscale fibrous networks. We show that macroscale substrates with radii of curvature from tens to hundreds of micrometers influence cell alignment. In a model system of fibroblasts, isolated cells aligned strongly in the axial direction on cylinders with radii similar to the cell length and more weakly on cylinders of much larger radius. Isolated vascular smooth muscle cells did not align as effectively as fibroblasts. However, both cell types aligned robustly in weak curvature fields when in confluent monolayers. We identified two distinct populations of stress fibers in both cell types: long, apical stress fibers that aligned axially and short, basal stress fibers that aligned circumferentially. Circumferential alignment of the basal stress fibers is in apparent disagreement with a long-standing hypothesis that energetic penalties for bending enforce axial alignment on cylinders. To explore this phenomenon, we manipulated stress fibers by activating Rho, a small guanosine triphosphatase that regulates stress fiber assembly. In response, apical stress fibers disassembled, whereas basal stress fibers thickened and aligned more strongly in the circumferential direction. By activating Rho in confluent monolayers of vascular smooth muscle cells, we recapitulated the circumferential alignment pattern of F-actin within these cells that is observed in cylindrical vessels in vivo. In agreement with recent theory, these results suggest that stress fiber bending penalties are overcome when stress fiber contractility is enhanced and motivate deeper study of the mechanics of these distinct stress fiber populations.
Collapse
Affiliation(s)
- Nathan D. Bade
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Randall D. Kamien
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Richard K. Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen J. Stebe
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Walters B, Uynuk-Ool T, Rothdiener M, Palm J, Hart ML, Stegemann JP, Rolauffs B. Engineering the geometrical shape of mesenchymal stromal cells through defined cyclic stretch regimens. Sci Rep 2017; 7:6640. [PMID: 28747783 PMCID: PMC5529555 DOI: 10.1038/s41598-017-06794-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cells have been predicted to improve disease outcomes and patient lives. Steering stem cell fate - through controlling cell shape - may substantially accelerate progress towards this goal. As mesenchymal stromal cells (MSCs) are continuously exposed in vivo to a dynamically changing biomechanical environment, we hypothesized that exogenous forces can be applied for engineering a variety of significantly different MSC shapes. We applied specific cyclic stretch regimens to human MSCs and quantitatively measured the resulting cell shape, alignment, and expression of smooth muscle (SMC) differentiation markers, as those have been associated with elongated morphology. As proof of principle, a range of different shapes, alignments, and correlating SMC marker levels were generated by varying strain, length, and repetition of stretch. However, the major determinant of biomechanically engineering cellular shape was the repetition of a chosen stretch regimen, indicating that the engineered shape and associated differentiation were complex non-linear processes relying on sustained biomechanical stimulation. Thus, forces are key regulators of stem cell shape and the targeted engineering of specific MSC shapes through biomechanical forces represents a novel mechanobiology concept that could exploit naturally occurring in vivo forces for improving stem cell fate in clinical regenerative therapies.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, United States
| | - Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Waldhoernlestr. 22, 72072, Tuebingen, Germany
| | - Miriam Rothdiener
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Waldhoernlestr. 22, 72072, Tuebingen, Germany
| | - Julian Palm
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Waldhoernlestr. 22, 72072, Tuebingen, Germany
| | - Melanie L Hart
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, United States
| | - Bernd Rolauffs
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany. .,Massachusetts Institute of Technology, Center for Biomedical Engineering, Cambridge, MA, 02319, USA.
| |
Collapse
|
43
|
Steucke KE, Win Z, Stemler TR, Walsh EE, Hall JL, Alford PW. Empirically Determined Vascular Smooth Muscle Cell Mechano-Adaptation Law. J Biomech Eng 2017; 139:2619314. [PMID: 28418526 PMCID: PMC5467037 DOI: 10.1115/1.4036454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/20/2017] [Indexed: 01/28/2023]
Abstract
Cardiovascular disease can alter the mechanical environment of the vascular system, leading to mechano-adaptive growth and remodeling. Predictive models of arterial mechano-adaptation could improve patient treatments and outcomes in cardiovascular disease. Vessel-scale mechano-adaptation includes remodeling of both the cells and extracellular matrix. Here, we aimed to experimentally measure and characterize a phenomenological mechano-adaptation law for vascular smooth muscle cells (VSMCs) within an artery. To do this, we developed a highly controlled and reproducible system for applying a chronic step-change in strain to individual VSMCs with in vivo like architecture and tracked the temporal cellular stress evolution. We found that a simple linear growth law was able to capture the dynamic stress evolution of VSMCs in response to this mechanical perturbation. These results provide an initial framework for development of clinically relevant models of vascular remodeling that include VSMC adaptation.
Collapse
Affiliation(s)
- Kerianne E Steucke
- Department of Biomedical Engineering, University of Minnesota Twin Cities, 312 Church Street SE NHH 7-105, Minneapolis, MN 55455 e-mail:
| | - Zaw Win
- Department of Biomedical Engineering, University of Minnesota Twin Cities, 312 Church Street SE NHH 7-105, Minneapolis, MN 55455 e-mail:
| | - Taylor R Stemler
- Department of Biomedical Engineering, University of Minnesota Twin Cities, 312 Church Street SE NHH 7-105, Minneapolis, MN 55455 e-mail:
| | - Emily E Walsh
- Department of Biomedical Engineering, University of Minnesota Twin Cities, 312 Church Street SE NHH 7-105, Minneapolis, MN 55455 e-mail:
| | - Jennifer L Hall
- Division of Cardiology, Department of Medicine, University of Minnesota Twin Cities, 2231 6th Street SE CCRB, Minneapolis, MN 55455 e-mail:
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota Twin Cities, 312 Church Street SE NHH 7-105, Minneapolis, MN 55455 e-mail:
| |
Collapse
|
44
|
Mantella LE, Singh KK, Sandhu P, Kantores C, Ramadan A, Khyzha N, Quan A, Al-Omran M, Fish JE, Jankov RP, Verma S. Fingerprint of long non-coding RNA regulated by cyclic mechanical stretch in human aortic smooth muscle cells: implications for hypertension. Mol Cell Biochem 2017; 435:163-173. [PMID: 28526936 DOI: 10.1007/s11010-017-3065-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 05/05/2017] [Indexed: 11/26/2022]
Abstract
Emerging evidence suggests that long non-coding RNAs (lncRNAs) represent a cellular hub coordinating various cellular processes that are critical in health and disease. Mechanical stress triggers changes in vascular smooth muscle cells (VSMCs) that in turn contribute to pathophysiological changes within the vasculature. We sought to evaluate the role that lncRNAs play in mechanical stretch-induced alterations of human aortic smooth muscle cells (HASMCs). RNA (lncRNA and mRNA) samples isolated from HASMCs that had been subjected to 10 or 20% elongation (1 Hz) for 24 h were profiled with the Arraystar Human LncRNA Microarray V3.0. LncRNA expression was quantified in parallel via qRT-PCR. Of the 30,586 human lncRNAs screened, 580 were differentially expressed (DE, P < 0.05) in stretched HASMCs. Amongst the 26,109 protein-coding transcripts evaluated, 25 of those DE were associated with 25 of the aforementioned DE lncRNAs (P < 0.05). Subsequent Kyoto Encyclopedia of Genes and Genomes analysis revealed that the DE mRNAs were largely associated with the tumor necrosis factor signaling pathway and inflammation. Gene Ontology analysis indicated that the DE mRNAs were associated with cell differentiation, stress response, and response to external stimuli. We describe the first transcriptome profile of stretch-induced changes in HASMCs and provide novel insights into the regulatory switches that may be fundamental in governing aberrant VSMC remodeling.
Collapse
Affiliation(s)
- Laura-Eve Mantella
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Krishna K Singh
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Paul Sandhu
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
| | - Crystal Kantores
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Azza Ramadan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Nadiya Khyzha
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, ON, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
| | - Mohammed Al-Omran
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Department of Surgery, King Saud University, Riyadh, Kingdom of Saudi Arabia
- The King Saud University-Li Ka Shing Collaborative Research Program, Riyadh, Kingdom of Saudi Arabia
| | - Jason E Fish
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, ON, Canada
| | - Robert P Jankov
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, ON, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Surgery, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
45
|
Ribas J, Zhang YS, Pitrez PR, Leijten J, Miscuglio M, Rouwkema J, Dokmeci MR, Nissan X, Ferreira L, Khademhosseini A. Biomechanical Strain Exacerbates Inflammation on a Progeria-on-a-Chip Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:10.1002/smll.201603737. [PMID: 28211642 PMCID: PMC5545787 DOI: 10.1002/smll.201603737] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/02/2017] [Indexed: 05/22/2023]
Abstract
Organ-on-a-chip platforms seek to recapitulate the complex microenvironment of human organs using miniaturized microfluidic devices. Besides modeling healthy organs, these devices have been used to model diseases, yielding new insights into pathophysiology. Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease showing accelerated vascular aging, leading to the death of patients due to cardiovascular diseases. HGPS targets primarily vascular cells, which reside in mechanically active tissues. Here, a progeria-on-a-chip model is developed and the effects of biomechanical strain are examined in the context of vascular aging and disease. Physiological strain induces a contractile phenotype in primary smooth muscle cells (SMCs), while a pathological strain induces a hypertensive phenotype similar to that of angiotensin II treatment. Interestingly, SMCs derived from human induced pluripotent stem cells of HGPS donors (HGPS iPS-SMCs), but not from healthy donors, show an exacerbated inflammatory response to strain. In particular, increased levels of inflammation markers as well as DNA damage are observed. Pharmacological intervention reverses the strain-induced damage by shifting gene expression profile away from inflammation. The progeria-on-a-chip is a relevant platform to study biomechanics in vascular biology, particularly in the setting of vascular disease and aging, while simultaneously facilitating the discovery of new drugs and/or therapeutic targets.
Collapse
Affiliation(s)
- João Ribas
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Doctoral Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Patrícia R. Pitrez
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mario Miscuglio
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mehmet Remzi Dokmeci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Xavier Nissan
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex 91030, France
| | | | | |
Collapse
|
46
|
Uynuk-Ool T, Rothdiener M, Walters B, Hegemann M, Palm J, Nguyen P, Seeger T, Stöckle U, Stegemann JP, Aicher WK, Kurz B, Hart ML, Klein G, Rolauffs B. The geometrical shape of mesenchymal stromal cells measured by quantitative shape descriptors is determined by the stiffness of the biomaterial and by cyclic tensile forces. J Tissue Eng Regen Med 2017; 11:3508-3522. [PMID: 28371409 DOI: 10.1002/term.2263] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 04/15/2016] [Accepted: 07/03/2016] [Indexed: 12/21/2022]
Abstract
Controlling mesenchymal stromal cell (MSC) shape is a novel method for investigating and directing MSC behaviour in vitro. it was hypothesized that specifigc MSC shapes can be generated by using stiffness-defined biomaterial surfaces and by applying cyclic tensile forces. Biomaterials used were thin and thick silicone sheets, fibronectin coating, and compacted collagen type I sheets. The MSC morphology was quantified by shape descriptors describing dimensions and membrane protrusions. Nanoscale stiffness was measured by atomic force microscopy and the expression of smooth muscle cell (SMC) marker genes (ACTA2, TAGLN, CNN1) by quantitative reverse-transcription polymerase chain reaction. Cyclic stretch was applied with 2.5% or 5% amplitudes. Attachment to biomaterials with a higher stiffness yielded more elongated MSCs with fewer membrane protrusions compared with biomaterials with a lower stiffness. For cyclic stretch, compacted collagen sheets were selected, which were associated with the most elongated MSC shape across all investigated biomaterials. As expected, cyclic stretch elongated MSCs during stretch. One hour after cessation of stretch, however, MSC shape was rounder again, suggesting loss of stretch-induced shape. Different shape descriptor values obtained by different stretch regimes correlated significantly with the expression levels of SMC marker genes. Values of approximately 0.4 for roundness and 3.4 for aspect ratio were critical for the highest expression levels of ACTA2 and CNN1. Thus, specific shape descriptor values, which can be generated using biomaterial-associated stiffness and tensile forces, can serve as a template for the induction of specific gene expression levels in MSC. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Miriam Rothdiener
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Brandan Walters
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Miriam Hegemann
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Julian Palm
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Phong Nguyen
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Tanja Seeger
- University Medical Clinic, Department II, Centre for Medical Research, University of Tübingen, Tübingen, Germany
| | - Ulrich Stöckle
- Clinic for Trauma and Restorative Surgery, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Wilhelm K Aicher
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University, Kiel, Germany
| | - Melanie L Hart
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tübingen, University of Tübingen, Tübingen, Germany
| | - Gerd Klein
- University Medical Clinic, Department II, Centre for Medical Research, University of Tübingen, Tübingen, Germany
| | - Bernd Rolauffs
- Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| |
Collapse
|
47
|
Babahosseini H, Strobl JS, Agah M. Microfluidic Iterative Mechanical Characteristics (iMECH) Analyzer for Single-Cell Metastatic Identification. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2017; 9:847-855. [PMID: 29034007 PMCID: PMC5637398 DOI: 10.1039/c6ay03342c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
This study describes the development of a microfluidic biosensor called the iterative mechanical characteristics (iMECH) analyzer which enables label-free biomechanical profiling of individual cells for distinction between metastatic and non-metastatic human mammary cell lines. Previous results have demonstrated that pulsed mechanical nanoindentation can modulate the biomechanics of cells resulting in distinctly different biomechanical responses in metastatic and non-metastatic cell lines. The iMECH analyzer aims to move this concept into a microfluidic, clinically more relevant platform. The iMECH analyzer directs a cyclic deformation regimen by pulling cells through a test channel comprised of narrow deformation channels and interspersed with wider relaxation regions which together simulate a dynamic microenvironment. The results of the iMECH analysis of human breast cell lines revealed that cyclic deformations produce a resistance in non-metastatic 184A1 and MCF10A cells as determined by a drop in their average velocity in the iterative deformation channels after each relaxation. In contrast, metastatic MDA-MB-231 and MDA-MB-468 cells exhibit a loss of resistance as measured by a velocity raise after each relaxation. These distinctive modulatory mechanical responses of normal-like non-metastatic and metastatic cancer breast cells to the pulsed indentations paradigm provide a unique bio-signature. The iMECH analyzer represents a diagnostic microchip advance for discriminating metastatic cancer at the single-cell level.
Collapse
Affiliation(s)
- Hesam Babahosseini
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, US
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, US
| | - Jeannine S. Strobl
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, US
| | - Masoud Agah
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, US
| |
Collapse
|
48
|
Rothdiener M, Hegemann M, Uynuk-Ool T, Walters B, Papugy P, Nguyen P, Claus V, Seeger T, Stoeckle U, Boehme KA, Aicher WK, Stegemann JP, Hart ML, Kurz B, Klein G, Rolauffs B. Stretching human mesenchymal stromal cells on stiffness-customized collagen type I generates a smooth muscle marker profile without growth factor addition. Sci Rep 2016; 6:35840. [PMID: 27775041 PMCID: PMC5075785 DOI: 10.1038/srep35840] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 10/05/2016] [Indexed: 12/18/2022] Open
Abstract
Using matrix elasticity and cyclic stretch have been investigated for inducing mesenchymal stromal cell (MSC) differentiation towards the smooth muscle cell (SMC) lineage but not in combination. We hypothesized that combining lineage-specific stiffness with cyclic stretch would result in a significantly increased expression of SMC markers, compared to non-stretched controls. First, we generated dense collagen type I sheets by mechanically compressing collagen hydrogels. Atomic force microscopy revealed a nanoscale stiffness range known to support myogenic differentiation. Further characterization revealed viscoelasticity and stable biomechanical properties under cyclic stretch with >99% viable adherent human MSC. MSCs on collagen sheets demonstrated a significantly increased mRNA but not protein expression of SMC markers, compared to on culture flasks. However, cyclic stretch of MSCs on collagen sheets significantly increased both mRNA and protein expression of α-smooth muscle actin, transgelin, and calponin versus plastic and non-stretched sheets. Thus, lineage-specific stiffness and cyclic stretch can be applied together for inducing MSC differentiation towards SMCs without the addition of recombinant growth factors or other soluble factors. This represents a novel stimulation method for modulating the phenotype of MSCs towards SMCs that could easily be incorporated into currently available methodologies to obtain a more targeted control of MSC phenotype.
Collapse
Affiliation(s)
- Miriam Rothdiener
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | | | - Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | - Brandan Walters
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Piruntha Papugy
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | - Phong Nguyen
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | - Valentin Claus
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | - Tanja Seeger
- Center for Medical Research, Medical University Clinic II, University of Tuebingen, Germany
| | - Ulrich Stoeckle
- Clinic for Trauma and Restorative Surgery, BG Trauma Clinic Tuebingen, University of Tuebingen, Germany
| | - Karen A. Boehme
- Department of Orthopaedic Surgery, University of Tuebingen, Germany
| | | | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Melanie L. Hart
- Department of Orthopedics and Trauma Surgery, Albert-Ludwigs-University, Freiburg, Germany
| | - Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University, Kiel, Germany
| | - Gerd Klein
- Center for Medical Research, Medical University Clinic II, University of Tuebingen, Germany
| | - Bernd Rolauffs
- Department of Orthopedics and Trauma Surgery, Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
49
|
Kamble H, Barton MJ, Jun M, Park S, Nguyen NT. Cell stretching devices as research tools: engineering and biological considerations. LAB ON A CHIP 2016; 16:3193-203. [PMID: 27440436 DOI: 10.1039/c6lc00607h] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cells within the human body are subjected to continuous, cyclic mechanical strain caused by various organ functions, movement, and growth. Cells are well known to have the ability to sense and respond to mechanical stimuli. This process is referred to as mechanotransduction. A better understanding of mechanotransduction is of great interest to clinicians and scientists alike to improve clinical diagnosis and understanding of medical pathology. However, the complexity involved in in vivo biological systems creates a need for better in vitro technologies, which can closely mimic the cells' microenvironment using induced mechanical strain. This technology gap motivates the development of cell stretching devices for better understanding of the cell response to mechanical stimuli. This review focuses on the engineering and biological considerations for the development of such cell stretching devices. The paper discusses different types of stretching concepts, major design consideration and biological aspects of cell stretching and provides a perspective for future development in this research area.
Collapse
Affiliation(s)
- Harshad Kamble
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan Campus, 170 Kessels Road, QLD 4111, Australia.
| | - Matthew J Barton
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Myeongjun Jun
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, Korea
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan Campus, 170 Kessels Road, QLD 4111, Australia.
| |
Collapse
|
50
|
Wilson K, Terlouw A, Roberts K, Wolchok JC. The characterization of decellularized human skeletal muscle as a blueprint for mimetic scaffolds. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:125. [PMID: 27324779 PMCID: PMC6260795 DOI: 10.1007/s10856-016-5735-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/28/2016] [Indexed: 05/08/2023]
Abstract
The use of decellularized skeletal muscle (DSM) as a cell substrate and scaffold for the repair of volumetric muscle loss injuries has shown therapeutic promise. The performance of DSM materials motivated our interest in exploring the chemical and physical properties of this promising material. We suggest that these properties could serve as a blueprint for the development of next generation engineered materials with DSM mimetic properties. In this study, whole human lower limb rectus femoris (n = 10) and upper limb supraspinatus muscle samples (n = 10) were collected from both male and female tissue donors. Skeletal muscle samples were decellularized and nine property values, capturing key compositional, architectural, and mechanical properties, were measured and statistically analyzed. Mean values for each property were determined across muscle types and sexes. Additionally, the influence of muscle type (upper vs lower limb) and donor sex (male vs female) on each of the DSM material properties was examined. The data suggests that DSM materials prepared from lower limb rectus femoris samples have an increased modulus and contain a higher collagen content then upper limb supraspinatus muscles. Specifically, lower limb rectus femoris DSM material modulus and collagen content was approximately twice that of lower limb supraspinatus DSM samples. While muscle type did show some influence on material properties, we did not find significant trends related to sex. The material properties reported herein may be used as a blueprint for the data-driven design of next generation engineered scaffolds with muscle mimetic properties, as well as inputs for computational and physical models of skeletal muscle.
Collapse
Affiliation(s)
- Klaire Wilson
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Abby Terlouw
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Kevin Roberts
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, USA
| | - Jeffrey C Wolchok
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA.
| |
Collapse
|