1
|
Rodríguez-Montaño ÓL, Santoro L, Vaiani L, Lamberti L, Uva AE, Boccaccio A. Cell adhesion on substrates with variable curvature: Effects on genetic transcription processes. Comput Biol Med 2025; 189:109917. [PMID: 40023074 DOI: 10.1016/j.compbiomed.2025.109917] [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: 10/18/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Several studies suggest that changes in nuclear morphology due to forces and deformations as result of cell adhesion on biological substrates can induce molecular streaming through nuclear pore openings and alter chromatin structure. The condensed state of chromatin hinders transcription and replication, while its decompaction, induced by adhesion, plays a key role in differentiation. However, assessing nuclear stress/strain in vivo remains challenging, and the impact of substrate curvature on nuclear mechanics and chromatin structures is still unclear. In this study, we developed an axisymmetric finite element model of a mesenchymal stem cell adhering to substrates with different curvatures to analyze nuclear stress distribution and identify locations where adhesion-induced gene expression may occur. Results reveal a nuclear stress field with principal stresses in radial and circumferential directions, leading to chromatin decondensation and nuclear pore opening. The predicted forces acting on chromatin fibers, estimated and compared with experimental data, remain slightly below 5 pN-the threshold at which internucleosomal attraction is disrupted, triggering chromatin condensation-decondensation transition-. During early spreading, nuclear forces achieved through adhesion on convex substrates approach this threshold more closely than in concave or flat cases. These findings provide insights for tissue engineering and regenerative medicine, where early control of stem cell fate through substrate design is crucial. Understanding how mesenchymal stem cells respond to substrate curvature could lead to improved biomaterial surface topographies for guiding cell behavior. Tailoring curvature and mechanical properties may enhance early lineage commitment, optimizing regenerative strategies for tissue repair and organ regeneration.
Collapse
Affiliation(s)
| | - Lorenzo Santoro
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Lorenzo Vaiani
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Luciano Lamberti
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Antonio E Uva
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Antonio Boccaccio
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy.
| |
Collapse
|
2
|
Teng Y, Zhang X, Song L, Yang J, Li D, Shi Z, Guo X, Wang S, Fan H, Jiang L, Hou S, Ramakrishna S, Lv Q, Shi J. Construction of anti-calcification small-diameter vascular grafts using decellularized extracellular matrix/poly (L-lactide-co-ε-caprolactone) and baicalin-cathepsin S inhibitor. Acta Biomater 2025; 197:184-201. [PMID: 40120837 DOI: 10.1016/j.actbio.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The long-term transplantation of small-diameter vascular grafts (SDVGs) is associated with a risk of calcification, which is a key factor limiting the clinical translation of SDVG. Hence, there is an urgency attached to the development of new SDVGs with anti-calcification properties. Here, we used decellularized extracellular matrix (dECM) and poly (L-lactide-co-ε-caprolactone) (PLCL) as base materials and combined these with baicalin, cathepsin S (Cat S) inhibitor to prepare PBC-SDVGs by electrospinning. Baicalin contains carboxyl and hydroxyl groups that can interact with chemical groups in dECM powder, potentially blocking calcium nucleation sites. Cat S inhibitor prevents elastin degradation and further reduces the risk of calcification. PBC-SDVGs were biocompatible and when implanted in rat abdominal aorta, accelerated endothelialization, enhanced vascular tissue regeneration, inhibited elastin degradation, and promoted macrophage polarization M2 phenotype to regulate inflammation. After 3 months of implantation, the results of Doppler ultrasound, MicroCT, and histological staining revealed a significant reduction in calcification. In summary, the developed anti-calcification SDVGs offer a promising strategy for long-term implantation with significant clinical application potential. STATEMENT OF SIGNIFICANCE: The dECM and PLCL were used as base materials, connected with baicalin, and loaded with Cat S inhibitor to prepare PBC-SDVGs. The baicalin and dECM powder formed hydrogen bonds to crosslink together reducing the calcium deposition. In vitro, the vascular graft downregulated the expression level of osteogenic genes and promoted macrophage polarization toward an anti-inflammatory M2 phenotype, thereby reducing calcification. The PBC-SDVGs implanted in rat abdominal aorta can accelerate endothelialization, enhance vascular tissue regeneration, inhibit elastin degradation, reduce inflammation response and calcification.
Collapse
Affiliation(s)
- Yanjiao Teng
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaohai Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Jianing Yang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Duo Li
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Ziqi Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaoqin Guo
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Li Jiang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Tianjin 300021, PR China
| | - Shike Hou
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore.
| | - Qi Lv
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Jie Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| |
Collapse
|
3
|
Zhang Y, Fu Q, Sun W, Yue Q, He P, Niu D, Zhang M. Mechanical forces in the tumor microenvironment: roles, pathways, and therapeutic approaches. J Transl Med 2025; 23:313. [PMID: 40075523 PMCID: PMC11899831 DOI: 10.1186/s12967-025-06306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Tumors often exhibit greater stiffness compared to normal tissues, primarily due to increased deposition within the tumor stroma. Collagen, proteoglycans, laminin, and fibronectin are key components of the extracellular matrix (ECM), interacting to facilitate ECM assembly. Enhanced fiber density and cross-linking within the ECM result in elevated matrix stiffness and interstitial fluid pressure, subjecting tumors to significant physical stress during growth. This mechanical stress is transduced intracellularly via integrins, the Rho signaling pathway, and the Hippo signaling pathway, thereby promoting tumor invasion. Additionally, mechanical pressure fosters glycolysis in tumor cells, boosting energy production to support metastasis. Mechanical cues also regulate macrophage polarization, maintaining an inflammatory microenvironment conducive to tumor survival. In summary, mechanical signals within tumors play a crucial role in tumor growth and invasion. Understanding these signals and their involvement in tumor progression is essential for advancing our knowledge of tumor biology and enhancing therapeutic approaches.
Collapse
Affiliation(s)
- Yanli Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| | - Qi Fu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Wenyue Sun
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Qiujuan Yue
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Ping He
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Dong Niu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Min Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| |
Collapse
|
4
|
Smirnova O, Efremov Y, Klyucherev T, Peshkova M, Senkovenko A, Svistunov A, Timashev P. Direct and cell-mediated EV-ECM interplay. Acta Biomater 2024; 186:63-84. [PMID: 39043290 DOI: 10.1016/j.actbio.2024.07.029] [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: 02/02/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Extracellular vesicles (EV) are a heterogeneous group of lipid particles excreted by cells. They play an important role in regeneration, development, inflammation, and cancer progression, together with the extracellular matrix (ECM), which they constantly interact with. In this review, we discuss direct and indirect interactions of EVs and the ECM and their impact on different physiological processes. The ECM affects the secretion of EVs, and the properties of the ECM and EVs modulate EVs' diffusion and adhesion. On the other hand, EVs can affect the ECM both directly through enzymes and indirectly through the modulation of the ECM synthesis and remodeling by cells. This review emphasizes recently discovered types of EVs bound to the ECM and isolated by enzymatic digestion, including matrix-bound nanovesicles (MBV) and tissue-derived EV (TiEV). In addition to the experimental studies, computer models of the EV-ECM-cell interactions, from all-atom models to quantitative pharmacology models aiming to improve our understanding of the interaction mechanisms, are also considered. STATEMENT OF SIGNIFICANCE: Application of extracellular vesicles in tissue engineering is an actively developing area. Vesicles not only affect cells themselves but also interact with the matrix and change it. The matrix also influences both cells and vesicles. In this review, different possible types of interactions between vesicles, matrix, and cells are discussed. Furthermore, the united EV-ECM system and its regulation through the cellular activity are presented.
Collapse
Affiliation(s)
- Olga Smirnova
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Yuri Efremov
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Timofey Klyucherev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 119991 Moscow, Russia
| | - Alexey Senkovenko
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | | | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 119991 Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia.
| |
Collapse
|
5
|
Xia Y, Rao Z, Wu S, Huang J, Zhou H, Li H, Zheng H, Guo D, Quan D, Ou JS, Bai Y, Liu Y. Polyzwitterion-grafted decellularized bovine intercostal arteries as new substitutes of small-diameter arteries for vascular regeneration. Regen Biomater 2024; 11:rbae098. [PMID: 39224131 PMCID: PMC11368410 DOI: 10.1093/rb/rbae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Coronary artery bypass grafting is acknowledged as a major clinical approach for treatment of severe coronary artery atherosclerotic heart disease. This procedure typically requires autologous small-diameter vascular grafts. However, the limited availability of the donor vessels and associated trauma during tissue harvest underscore the necessity for artificial arterial alternatives. Herein, decellularized bovine intercostal arteries were successfully fabricated with lengths ranging from 15 to 30 cm, which also closely match the inner diameters of human coronary arteries. These decellularized arterial grafts exhibited great promise following poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) grafting from the inner surface. Such surface modification endowed the decellularized arteries with superior mechanical strength, enhanced anticoagulant properties and improved biocompatibility, compared to the decellularized bovine intercostal arteries alone, or even those decellularized grafts modified with both heparin and vascular endothelial growth factor. After replacement of the carotid arteries in rabbits, all surface-modified vascular grafts have shown good patency within 30 days post-implantation. Notably, strong signal was observed after α-SMA immunofluorescence staining on the PMPC-grafted vessels, indicating significant potential for regenerating the vascular smooth muscle layer and thereby restoring full structures of the artery. Consequently, the decellularized bovine intercostal arteries surface modified by PMPC can emerge as a potent candidate for small-diameter artificial blood vessels, and have shown great promise to serve as viable substitutes of arterial autografts.
Collapse
Affiliation(s)
- Yuan Xia
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Simin Wu
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiayao Huang
- Department of Medical Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Haiyun Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510160, China
| | - Hanzhao Li
- Department of Cardiac Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510160, China
| | - Hui Zheng
- Department of Cardiac Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510160, China
| | - Daxin Guo
- Department of Cardiac Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510160, China
| | - Daping Quan
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Engineering Technology Centre for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Yunqi Liu
- Department of Cardiac Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510160, China
| |
Collapse
|
6
|
Radman BA, Alhameed AMM, Shu G, Yin G, Wang M. Cellular elasticity in cancer: a review of altered biomechanical features. J Mater Chem B 2024; 12:5299-5324. [PMID: 38742281 DOI: 10.1039/d4tb00328d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
A large number of studies have shown that changes in biomechanical characteristics are an important indicator of tumor transformation in normal cells. Elastic deformation is one of the more studied biomechanical features of tumor cells, which plays an important role in tumourigenesis and development. Altered cell elasticity often brings many indications. This manuscript reviews the effects of altered cellular elasticity on cell characteristics, including adhesion viscosity, migration, proliferation, and differentiation elasticity and stiffness. Also, the physical factors that may affect cell elasticity, such as temperature, cell height, cell-viscosity, and aging, are summarized. Then, the effects of cell-matrix, cytoskeleton, in vitro culture medium, and cell-substrate with different three-dimensional structures on cell elasticity during cell tumorigenesis are outlined. Importantly, we summarize the current signaling pathways that may affect cellular elasticity, as well as tests for cellular elastic deformation. Finally, we summarize current hybrid materials: polymer-polymer, protein-protein, and protein-polymer hybrids, also, nano-delivery strategies that target cellular resilience and cases that are at least in clinical phase 1 trials. Overall, the behavior of cancer cell elasticity is modulated by biological, chemical, and physical changes, which in turn have the potential to alter cellular elasticity, and this may be an encouraging prediction for the future discovery of cancer therapies.
Collapse
Affiliation(s)
- Bakeel A Radman
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
- Department of Biology, College of Science and Education, Albaydha University, Yemen
| | | | - Guang Shu
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, 410013, China
- China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
| |
Collapse
|
7
|
Zhang H, Rahman T, Lu S, Adam AP, Wan LQ. Helical vasculogenesis driven by cell chirality. SCIENCE ADVANCES 2024; 10:eadj3582. [PMID: 38381835 PMCID: PMC10881055 DOI: 10.1126/sciadv.adj3582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
The cellular helical structure is well known for its crucial role in development and disease. Nevertheless, the underlying mechanism governing this phenomenon remains largely unexplored, particularly in recapitulating it in well-controlled engineering systems. Leveraging advanced microfluidics, we present compelling evidence of the spontaneous emergence of helical endothelial tubes exhibiting robust right-handedness governed by inherent cell chirality. To strengthen our findings, we identify a consistent bias toward the same chirality in mouse vascular tissues. Manipulating endothelial cell chirality using small-molecule drugs produces a dose-dependent reversal of the handedness in engineered vessels, accompanied by non-monotonic changes in vascular permeability. Moreover, our three-dimensional cell vertex model provides biomechanical insights into the chiral morphogenesis process, highlighting the role of cellular torque and tissue fluidity in its regulation. Our study unravels an intriguing mechanism underlying vascular chiral morphogenesis, shedding light on the broader implications and distinctive perspectives of tubulogenesis within biological systems.
Collapse
Affiliation(s)
- Haokang Zhang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Tasnif Rahman
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Shuhan Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
- Department of Ophthalmology, Albany Medical College, Albany, NY 12208, USA
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
8
|
Li YY, Ji SF, Fu XB, Jiang YF, Sun XY. Biomaterial-based mechanical regulation facilitates scarless wound healing with functional skin appendage regeneration. Mil Med Res 2024; 11:13. [PMID: 38369464 PMCID: PMC10874556 DOI: 10.1186/s40779-024-00519-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Scar formation resulting from burns or severe trauma can significantly compromise the structural integrity of skin and lead to permanent loss of skin appendages, ultimately impairing its normal physiological function. Accumulating evidence underscores the potential of targeted modulation of mechanical cues to enhance skin regeneration, promoting scarless repair by influencing the extracellular microenvironment and driving the phenotypic transitions. The field of skin repair and skin appendage regeneration has witnessed remarkable advancements in the utilization of biomaterials with distinct physical properties. However, a comprehensive understanding of the underlying mechanisms remains somewhat elusive, limiting the broader application of these innovations. In this review, we present two promising biomaterial-based mechanical approaches aimed at bolstering the regenerative capacity of compromised skin. The first approach involves leveraging biomaterials with specific biophysical properties to create an optimal scarless environment that supports cellular activities essential for regeneration. The second approach centers on harnessing mechanical forces exerted by biomaterials to enhance cellular plasticity, facilitating efficient cellular reprogramming and, consequently, promoting the regeneration of skin appendages. In summary, the manipulation of mechanical cues using biomaterial-based strategies holds significant promise as a supplementary approach for achieving scarless wound healing, coupled with the restoration of multiple skin appendage functions.
Collapse
Affiliation(s)
- Ying-Ying Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Shuai-Fei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| | - Yu-Feng Jiang
- Department of Tissue Regeneration and Wound Repair, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Xiao-Yan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| |
Collapse
|
9
|
Meijer E, Giles R, van Dijk CGM, Maringanti R, Wissing TB, Appels Y, Chrifi I, Crielaard H, Verhaar MC, Smits AI, Cheng C. Effect of Mechanical Stimuli on the Phenotypic Plasticity of Induced Pluripotent Stem-Cell-Derived Vascular Smooth Muscle Cells in a 3D Hydrogel. ACS APPLIED BIO MATERIALS 2023; 6:5716-5729. [PMID: 38032545 PMCID: PMC10731661 DOI: 10.1021/acsabm.3c00840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023]
Abstract
Introduction: Vascular smooth muscle cells (VSMCs) play a pivotal role in vascular homeostasis, with dysregulation leading to vascular complications. Human-induced pluripotent stem-cell (hiPSC)-derived VSMCs offer prospects for personalized disease modeling and regenerative strategies. Current research lacks comparative studies on the impact of three-dimensional (3D) substrate properties under cyclic strain on phenotypic adaptation in hiPSC-derived VSMCs. Here, we aim to investigate the impact of intrinsic substrate properties, such as the hydrogel's elastic modulus and cross-linking density in a 3D static and dynamic environment, on the phenotypical adaptation of human mural cells derived from hiPSC-derived organoids (ODMCs), compared to aortic VSMCs. Methods and results: ODMCs were cultured in two-dimensional (2D) conditions with synthetic or contractile differentiation medium or in 3D Gelatin Methacryloyl (GelMa) substrates with varying degrees of functionalization and percentages to modulate Young's modulus and cross-linking density. Cells in 3D substrates were exposed to cyclic, unidirectional strain. Phenotype characterization was conducted using specific markers through immunofluorescence and gene expression analysis. Under static 2D culture, ODMCs derived from hiPSCs exhibited a VSMC phenotype, expressing key mural markers, and demonstrated a level of phenotypic plasticity similar to primary human VSMCs. In static 3D culture, a substrate with a higher Young's modulus and cross-linking density promoted a contractile phenotype in ODMCs and VSMCs. Dynamic stimulation in the 3D substrate promoted a switch toward a contractile phenotype in both cell types. Conclusion: Our study demonstrates phenotypic plasticity of human ODMCs in response to 2D biological and 3D mechanical stimuli that equals that of primary human VSMCs. These findings may contribute to the advancement of tailored approaches for vascular disease modeling and regenerative strategies.
Collapse
Affiliation(s)
- Elana
M. Meijer
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Rachel Giles
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Christian G. M. van Dijk
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ranganath Maringanti
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Tamar B. Wissing
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Ymke Appels
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ihsan Chrifi
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Hanneke Crielaard
- Department
of Biomedical Engineering, Erasmus Medical
Center, Rotterdam 3000 CA, The Netherlands
| | - Marianne C. Verhaar
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Anthal I.P.M. Smits
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Caroline Cheng
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| |
Collapse
|
10
|
Mayner JM, Masutani EM, Demeester E, Kumar A, Macapugay G, Beri P, Lo Sardo V, Engler AJ. Heterogeneous expression of alternatively spliced lncRNA mediates vascular smooth cell plasticity. Proc Natl Acad Sci U S A 2023; 120:e2217122120. [PMID: 37276403 PMCID: PMC10268236 DOI: 10.1073/pnas.2217122120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/13/2023] [Indexed: 06/07/2023] Open
Abstract
9p21.3 locus polymorphisms have the strongest correlation with coronary artery disease, but as a noncoding locus, disease connection is enigmatic. The lncRNA ANRIL found in 9p21.3 may regulate vascular smooth muscle cell (VSMC) phenotype to contribute to disease risk. We observed significant heterogeneity in induced pluripotent stem cell-derived VSMCs from patients homozygous for risk versus isogenic knockout or nonrisk haplotypes. Subpopulations of risk haplotype cells exhibited variable morphology, proliferation, contraction, and adhesion. When sorted by adhesion, risk VSMCs parsed into synthetic and contractile subpopulations, i.e., weakly adherent and strongly adherent, respectively. Of note, >90% of differentially expressed genes coregulated by haplotype and adhesion and were associated with Rho GTPases, i.e., contractility. Weakly adherent subpopulations expressed more short isoforms of ANRIL, and when overexpressed in knockout cells, ANRIL suppressed adhesion, contractility, and αSMA expression. These data suggest that variable lncRNA penetrance may drive mixed functional outcomes that confound pathology.
Collapse
Affiliation(s)
- Jaimie M. Mayner
- Chien-Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA92093
| | - Evan M. Masutani
- Chien-Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA92093
| | - Elena Demeester
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Aditya Kumar
- Chien-Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA92093
| | - Gail Macapugay
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
| | - Pranjali Beri
- Chien-Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA92093
| | - Valentina Lo Sardo
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA92037
| |
Collapse
|
11
|
Walter C, Mathur J, Pathak A. Reciprocal intra- and extra-cellular polarity enables deep mechanosensing through layered matrices. Cell Rep 2023; 42:112362. [PMID: 37027304 PMCID: PMC11246724 DOI: 10.1016/j.celrep.2023.112362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/11/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Adherent cells migrate on layered tissue interfaces to drive morphogenesis, wound healing, and tumor invasion. Although stiffer surfaces are known to enhance cell migration, it remains unclear whether cells sense basal stiff environments buried under softer, fibrous matrix. Using layered collagen-polyacrylamide gel systems, we unveil a migration phenotype driven by cell-matrix polarity. Here, cancer (but not normal) cells with stiff base matrix generate stable protrusions, faster migration, and greater collagen deformation because of "depth mechanosensing" through the top collagen layer. Cancer cell protrusions with front-rear polarity produce polarized collagen stiffening and deformations. Disruption of either extracellular or intracellular polarity via collagen crosslinking, laser ablation, or Arp2/3 inhibition independently abrogates depth-mechanosensitive migration of cancer cells. Our experimental findings, validated by lattice-based energy minimization modeling, present a cell migration mechanism whereby polarized cellular protrusions and contractility are reciprocated by mechanical extracellular polarity, culminating in a cell-type-dependent ability to mechanosense through matrix layers.
Collapse
Affiliation(s)
- Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
12
|
Habbit NL, Anbiah B, Anderson L, Suresh J, Hassani I, Eggert M, Brannen A, Davis J, Tian Y, Prabhakarpandian B, Panizzi P, Arnold RD, Lipke EA. Tunable three-dimensional engineered prostate cancer tissues for in vitro recapitulation of heterogeneous in vivo prostate tumor stiffness. Acta Biomater 2022; 147:73-90. [PMID: 35551999 DOI: 10.1016/j.actbio.2022.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022]
Abstract
In this manuscript we report the establishment and characterization of a three-dimensional in vitro, coculture engineered prostate cancer tissue (EPCaT) disease model based upon and informed by our characterization of in vivo prostate cancer (PCa) xenograft tumor stiffness. In prostate cancer, tissue stiffness is known to impact changes in gene and protein expression, alter therapeutic response, and be positively correlated with an aggressive clinical presentation. To inform an appropriate stiffness range for our in vitro model, PC-3 prostate tumor xenografts were established. Tissue stiffness ranged from 95 to 6,750 Pa. Notably, xenograft cell seeding density significantly impacted tumor stiffness; a two-fold increase in the number of seeded cells not only widened the tissue stiffness range throughout the tumor but also resulted in significant spatial heterogeneity. To fabricate our in vitro EPCaT model, PC-3 castration-resistant prostate cancer cells were co-encapsulated with BJ-5ta fibroblasts within a poly(ethylene glycol)-fibrinogen matrix augmented with excess poly(ethylene glycol)-diacrylate to modulate the matrix mechanical properties. Encapsulated cells temporally remodeled their in vitro microenvironment and enrichment of gene sets associated with tumorigenic progression was observed in response to increased matrix stiffness. Through variation of matrix composition and culture duration, EPCaTs were tuned to mimic the wide range of biomechanical cues provided to PCa cells in vivo; collectively, a range of 50 to 10,000 Pa was achievable. Markedly, this also encompasses published clinical PCa stiffness data. Overall, this study serves to introduce our bioinspired, tunable EPCaT model and provide the foundation for future PCa progression and drug development studies. STATEMENT OF SIGNIFICANCE: The development of cancer models that mimic the native tumor microenvironment (TME) complexities is critical to not only develop effective drugs but also enhance our understanding of disease progression. Here we establish and characterize our 3D in vitro engineered prostate cancer tissue model with tunable matrix stiffness, that is inspired by this study's spatial characterization of in vivo prostate tumor xenograft stiffness. Notably, our model's mimicry of the TME is further augmented by the inclusion of matrix remodeling fibroblasts to introduce cancer-stromal cell-cell interactions. This study addresses a critical unmet need in the field by elucidating the prostate tumor xenograft stiffness range and establishing a foundation for recapitulating the biomechanics of site-of-origin and soft tissue metastatic prostate tumors in vitro.
Collapse
Affiliation(s)
- Nicole L Habbit
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Benjamin Anbiah
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Luke Anderson
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Joshita Suresh
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Iman Hassani
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Matthew Eggert
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Andrew Brannen
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Yuan Tian
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | | | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA.
| |
Collapse
|
13
|
Zhang XZ, Jiang YL, Hu JG, Zhao LM, Chen QZ, Liang Y, Zhang Y, Lei XX, Wang R, Lei Y, Zhang QY, Li-Ling J, Xie HQ. Procyanidins-crosslinked small intestine submucosa: A bladder patch promotes smooth muscle regeneration and bladder function restoration in a rabbit model. Bioact Mater 2021; 6:1827-1838. [PMID: 33336114 PMCID: PMC7721664 DOI: 10.1016/j.bioactmat.2020.11.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 02/05/2023] Open
Abstract
Currently the standard surgical treatment for bladder defects is augmentation cystoplasty with autologous tissues, which has many side effects. Biomaterials such as small intestine submucosa (SIS) can provide an alternative scaffold for the repair as bladder patches. Previous studies have shown that SIS could enhance the capacity and compliance of the bladder, but its application is hindered by issues like limited smooth muscle regeneration and stone formation since the fast degradation and poor mechanical properties of the SIS. Procyanidins (PC), a natural bio-crosslinking agent, has shown anti-calcification, anti-inflammatory and anti-oxidation properties. More importantly, PC and SIS can crosslink through hydrogen bonds, which may endow the material with enhanced mechanical property and stabilized functionalities. In this study, various concentrations of PC-crosslinked SIS (PC-SIS) were prepared to repair the full-thickness bladder defects, with an aim to reduce complications and enhance bladder functions. In vitro assays showed that the crosslinking has conferred the biomaterial with superior mechanical property and anti-calcification property, ability to promote smooth muscle cell adhesion and upregulate functional genes expression. Using a rabbit model with bladder defects, we demonstrated that the PC-SIS scaffold can rapidly promote in situ tissue regrowth and regeneration, in particular smooth muscle remodeling and improvement of urinary functions. The PC-SIS scaffold has therefore provided a promising material for the reconstruction of a functional bladder.
Collapse
Affiliation(s)
- Xiu-Zhen Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jun-Gen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Long-Mei Zhao
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qiu-Zhu Chen
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiong-Xin Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Rui Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yi Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jesse Li-Ling
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Hu M, Jia F, Huang WP, Li X, Hu DF, Wang J, Ren KF, Fu GS, Wang YB, Ji J. Substrate stiffness differentially impacts autophagy of endothelial cells and smooth muscle cells. Bioact Mater 2021; 6:1413-1422. [PMID: 33210033 PMCID: PMC7658328 DOI: 10.1016/j.bioactmat.2020.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 01/07/2023] Open
Abstract
Stiffening of blood vessels is one of the most important characteristics in the process of many cardiovascular pathologies such as atherosclerosis, angiosteosis, and vascular aging. Increased stiffness of the vascular extracellular matrix drives artery pathology and alters phenotypes of vascular cell. Understanding how substrate stiffness impacts vascular cell behaviors is of great importance to the biomaterial design in tissue engineering, regenerative medicine, and medical devices. Here we report that changing substrate stiffness has a significant impact on the autophagy of vascular endothelial cells (VECs) and smooth muscle cells (VSMCs). Interestingly, our findings demonstrate that, with the increase of substrate stiffness, the autophagy level of VECs and VSMCs showed differential changes: endothelial autophagy levels reduced, leading to the reductions in a range of gene expression associated with endothelial function; while, autophagy levels of VSMCs increased, showing a transition from contractile to the synthetic phenotype. We further demonstrate that, by inhibiting cell autophagy, the expressions of endothelial functional gene were further reduced and the expression of VSMC calponin increased, suggesting an important role of autophagy in response of the cells to the challenge of microenvironment stiffness changing. Although the underlying mechanism requires further study, this work highlights the relationship of substrate stiffness, autophagy, and vascular cell behaviors, and enlightening the design principles of surface stiffness of biomaterials in cardiovascular practical applications.
Collapse
Affiliation(s)
- Mi Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Fan Jia
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Wei-Pin Huang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xu Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Deng-Feng Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jing Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ke-Feng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Guo-Sheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yun-Bing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
15
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
16
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
17
|
Furdella KJ, Higuchi S, Behrangzade A, Kim K, Wagner WR, Vande Geest JP. In-vivo assessment of a tissue engineered vascular graft computationally optimized for target vessel compliance. Acta Biomater 2021; 123:298-311. [PMID: 33482362 DOI: 10.1016/j.actbio.2020.12.058] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 11/24/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) have the ability to be tuned to match a target vessel's compliance, diameter, wall thickness, and thereby prevent compliance mismatch. In this work, TEVG compliance was manipulated by computationally tuning its layered composition or by manipulating a crosslinking agent (genipin). In particular, these three acelluluar TEVGs were compared: a compliance matched graft (CMgel - high gelatin content); a hypocompliant PCL graft (HYPOpcl - high polycaprolactone content); and a hypocompliant genipin graft (HYPOgen - equivalent composition as CMgel but hypocompliant via increased genipin crosslinking). All constructs were implanted interpositionally into the abdominal aorta of 21 Sprague Dawley rats (n=7, males=11, females=10) for 28 days, imaged in-vivo using ultrasound, explanted, and assessed for remodeling using immunofluorescence and two photon excitation fluorescence imaging. Compliance matched grafts remained compliance-matched in-vivo compared to the hypocompliant grafts through 4 weeks (p<0.05). Construct degradation and cellular infiltration was increased in the CMgel and HYPOgen TEVGs. Contractile smooth muscle cell markers in the proximal anastomosis of the graft were increased in the CMgel group compared to the HYPOpcl (p=0.007) and HYPOgen grafts (p=0.04). Both hypocompliant grafts also had an increased pro-inflammatory response (increased ratio of CD163 to CD86 in the mid-axial location) compared to the CMgel group. Our results suggest that compliance matching using a computational optimization approach leads to the improved acute (28 day) remodeling of TEVGs. To the authors' knowledge, this is the first in-vivo rat study investigating TEVGs that have been computationally optimized for target vessel compliance.
Collapse
|
18
|
Synthesis of thermogel modified with biomaterials as carrier for hUSSCs differentiation into cardiac cells: Physicomechanical and biological assessment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111517. [DOI: 10.1016/j.msec.2020.111517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022]
|
19
|
Steering cell behavior through mechanobiology in 3D: A regenerative medicine perspective. Biomaterials 2020; 268:120572. [PMID: 33285439 DOI: 10.1016/j.biomaterials.2020.120572] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/04/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Abstract
Mechanobiology, translating mechanical signals into biological ones, greatly affects cellular behavior. Steering cellular behavior for cell-based regenerative medicine approaches requires a thorough understanding of the orchestrating molecular mechanisms, among which mechanotransducive ones are being more and more elucidated. Because of their wide use and highly mechanotransduction dependent differentiation, this review focuses on mesenchymal stromal cells (MSCs), while also briefly relating the discussed results to other cell types. While the mechanotransduction pathways are relatively well-studied in 2D, much remains unknown of the role and regulation of these pathways in 3D. Ultimately, cells need to be cultured in a 3D environment to create functional de novo tissue. In this review, we explore the literature on the roles of different material properties on cellular behavior and mechanobiology in 2D and 3D. For example, while stiffness plays a dominant role in 2D MSCs differentiation, it seems to be of subordinate importance in 3D MSCs differentiation, where matrix remodeling seems to be key. Also, the role and regulation of some of the main mechanotransduction players are discussed, focusing on MSCs. We have only just begun to fundamentally understand MSCs and other stem cells behavior in 3D and more fundamental research is required to advance biomaterials able to replicate the stem cell niche and control cell activity. This better understanding will contribute to smarter tissue engineering scaffold design and the advancement of regenerative medicine.
Collapse
|
20
|
Almeida Donanzam DDF, Donato TAG, Dos Reis KH, da Silva AP, Finato AC, Dos Santos AR, Cavalcante RS, Mendes RP, Venturini J. Exoantigens of Paracoccidioides spp. Promote Proliferation and Modulation of Human and Mouse Pulmonary Fibroblasts. Front Cell Infect Microbiol 2020; 10:590025. [PMID: 33194837 PMCID: PMC7662685 DOI: 10.3389/fcimb.2020.590025] [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: 07/31/2020] [Accepted: 10/07/2020] [Indexed: 11/13/2022] Open
Abstract
Paracoccidioidomycosis (PCM) is a systemic granulomatous fungal infection caused by thermally dimorphic fungi of the genus Paracoccidioides. Endemic in Latin America, PCM presents with high incidence in Brazil, Colombia, and Venezuela, especially among rural workers. The main clinical types are acute/subacute (AF) form and chronic form (CF). Even after effective antifungal treatment, patients with CF usually present sequelae, such as pulmonary fibrosis. In general, pulmonary fibrosis is associated with dysregulation wound healing and abnormal fibroblast activation. Although fibrogenesis is recognized as an early process in PCM, its mechanisms remain unknown. In the current study, we addressed the role of Paracoccidioides spp. exoantigens in pulmonary fibroblast proliferation and responsiveness. Human pulmonary fibroblasts (MRC-5) and pulmonary fibroblasts isolated from BALB/c mice were cultivated with 2.5, 5, 10, 100, and 250 µg/ml of exoantigens produced from P. brasiliensis (Pb18 and Pb326) and P. lutzii (Pb01, Pb8334, and Pb66) isolates. Purified gp43, the immunodominant protein of P. brasiliensis exoantigens, was also evaluated at concentrations of 5 and 10 µg/ml. After 24 h, proliferation and production of cytokines and growth factors by pulmonary fibroblasts were evaluated. Each exoantigen concentration promoted a different level of interference of the pulmonary fibroblasts. In general, exoantigens induced significant proliferation of both murine and human pulmonary fibroblasts (p < 0.05). All concentrations of exoantigens promoted decreased levels of IL-6 (p < 0.05) and VEGF (p < 0.05) in murine fibroblasts. Interestingly, decreased levels of bFGF (p < 0.05) and increased levels of TGF-β1 (p < 0.05) and pro-collagen I (p < 0.05) were observed in human fibroblasts. The gp43 protein induced increased TGF-β1 production by human cells (p = 0.02). In conclusion, our findings showed for the first time that components of P. brasiliensis and P. lutzii interfered in fibrogenesis by directly acting on the biology of pulmonary fibroblasts.
Collapse
Affiliation(s)
- Débora de Fátima Almeida Donanzam
- Faculdade de Medicina, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil.,Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | | | - Karoline Haghata Dos Reis
- Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - Adriely Primo da Silva
- Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - Angela Carolina Finato
- Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - Amanda Ribeiro Dos Santos
- Faculdade de Medicina, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil.,Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - Ricardo Souza Cavalcante
- Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - Rinaldo Poncio Mendes
- Faculdade de Medicina, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil.,Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| | - James Venturini
- Faculdade de Medicina, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil.,Faculdade de Medicina, Departamento de Doenças Tropicais e Diagnóstico por Imagem, UNESP, Botucatu, Brazil
| |
Collapse
|
21
|
Sanyour HJ, Rickel AP, Hong Z. The interplay of membrane cholesterol and substrate on vascular smooth muscle biomechanics. CURRENT TOPICS IN MEMBRANES 2020; 86:279-299. [PMID: 33837696 PMCID: PMC8041049 DOI: 10.1016/bs.ctm.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
Cardiovascular disease (CVD) remains the primary cause of death worldwide. Specifically, atherosclerosis is a CVD characterized as a slow progressing chronic inflammatory disease. During atherosclerosis, vascular walls accumulate cholesterol and cause fatty streak formation. The progressive changes in vascular wall stiffness exert alternating mechanical cues on vascular smooth muscle cells (VSMCs). The detachment of VSMCs in the media layer of the vessel and migration toward the intima is a critical step in atherosclerosis. VSMC phenotypic switching is a complicated process that modifies VSMC structure and biomechanical function. These changes affect the expression and function of cell adhesion molecules, thus impacting VSMC migration. Accumulating evidence has shown cholesterol is capable of regulating cellular migration, proliferation, and spreading. However, the interaction and coordinated effects of both cellular cholesterol and the extracellular matrix (ECM) stiffness/composition on VSMC biomechanics remains to be elucidated.
Collapse
Affiliation(s)
- Hanna J Sanyour
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Alex P Rickel
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Zhongkui Hong
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States.
| |
Collapse
|
22
|
Schwager SC, Reinhart-King CA. Mechanobiology of microvesicle release, uptake, and microvesicle-mediated activation. CURRENT TOPICS IN MEMBRANES 2020; 86:255-278. [PMID: 33837695 DOI: 10.1016/bs.ctm.2020.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Microvesicles are small, membrane-bound vesicles that are shed from the plasma membrane of cells into the extracellular space. Microvesicles contain a variety of cargo not typically thought to be released from cells, including receptor tyrosine kinases, cytosolic signaling proteins, and microRNAs, which are transferred from donor cells to recipient cells. The transfer of microvesicle cargo can result in the transformation of recipient cells thereby supporting disease progression, including modified fibroblast metabolism, epithelial cell contractility, vascular remodeling, and immune cell inflammatory signaling. Additionally, microvesicles are believed to play prominent roles in cell-cell communication and disease progression as they are detected at elevated concentrations in diseased tissues. As microvesicle uptake by recipient cells can modulate cell function to promote disease progression, understanding the mechanisms and mechanosensitivity of microvesicle release, internalization, and the resulting signaling is crucial to fully comprehend their functions in disease. Here, we review recent advances in the understanding of actomyosin-regulated microvesicle biogenesis, microvesicle uptake via pinocytosis, and the resulting cellular transformation. We discuss the effects of altered cell contractility, mode of cell migration, and extracellular matrix compliance on microvesicle signaling, with direct implications in disease progression and identifying future therapeutic targets.
Collapse
Affiliation(s)
- Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | | |
Collapse
|
23
|
Roveimiab Z, Lin F, Anderson JE. Traction and attraction: haptotaxis substrates collagen and fibronectin interact with chemotaxis by HGF to regulate myoblast migration in a microfluidic device. Am J Physiol Cell Physiol 2020; 319:C75-C92. [PMID: 32348173 DOI: 10.1152/ajpcell.00417.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell migration is central to development, wound healing, tissue regeneration, and immunity. Despite extensive knowledge of muscle regeneration, myoblast migration during regeneration is not well understood. C2C12 mouse myoblast migration and morphology were investigated using a triple-docking polydimethylsiloxane-based microfluidic device in which cells moved under gravity-driven laminar flow on uniform (=) collagen (CN=), fibronectin (FN=), or opposing gradients (CN-FN or FN-CN). In haptotaxis experiments, migration was faster on FN= than on CN=. At 10 h, cells were more elongated on FN-CN and migration was faster than on the CN-FN substrate. Net migration distance on FN-CN at 10 h was greater than on CN-FN, as cells rapidly entered the channel as a larger population (bulk-cell movement, wave 1). Hepatocyte growth factor (HGF) stimulated rapid chemotaxis on FN= but not CN=, increasing migration speed at 10 h early in the channel at low HGF in a steep HGF gradient. HGF accelerated migration on FN= and bulk-cell movement on both uniform substrates. An HGF gradient also slowed cells in wave 2 moving on FN-CN, not CN-FN. Both opposing-gradient substrates affected the shape, speed, and net distance of migrating cells. Gradient and uniform configurations of HGF and substrate differentially influenced migration behavior. Therefore, haptotaxis substrate configuration potently modifies myoblast chemotaxis by HGF. Innovative microfluidic experiments advance our understanding of intricate complexities of myoblast migration. Findings can be leveraged to engineer muscle-tissue volumes for transplantation after serious injury. New analytical approaches may generate broader insights into cell migration.
Collapse
Affiliation(s)
- Ziba Roveimiab
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Francis Lin
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Judy E Anderson
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
Mertgen AS, Trossmann VT, Guex AG, Maniura-Weber K, Scheibel T, Rottmar M. Multifunctional Biomaterials: Combining Material Modification Strategies for Engineering of Cell-Contacting Surfaces. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21342-21367. [PMID: 32286789 DOI: 10.1021/acsami.0c01893] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In the human body, cells in a tissue are exposed to signals derived from their specific extracellular matrix (ECM), such as architectural structure, mechanical properties, and chemical composition (proteins, growth factors). Research on biomaterials in tissue engineering and regenerative medicine aims to recreate such stimuli using engineered materials to induce a specific response of cells at the interface. Although traditional biomaterials design has been mostly limited to varying individual signals, increasing interest has arisen on combining several features in recent years to improve the mimicry of extracellular matrix properties. Tremendous progress in combinatorial surface modification exploiting, for example, topographical features or variations in mechanics combined with biochemical cues has enabled the identification of their key regulatory characteristics on various cell fate decisions. Gradients especially facilitated such research by enabling the investigation of combined continuous changes of different signals. Despite unravelling important synergies for cellular responses, challenges arise in terms of fabrication and characterization of multifunctional engineered materials. This review summarizes recent work on combinatorial surface modifications that aim to control biological responses. Modification and characterization methods for enhanced control over multifunctional material properties are highlighted and discussed. Thereby, this review deepens the understanding and knowledge of biomimetic combinatorial material modification, their challenges but especially their potential.
Collapse
Affiliation(s)
- Anne-Sophie Mertgen
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
- Laboratory for Biomimetic Membranes and Textiles, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Vanessa Tanja Trossmann
- Lehrstuhl für Biomaterialien, Universität Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, Bayreuth 95440, Germany
| | - Anne Géraldine Guex
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
- Laboratory for Biomimetic Membranes and Textiles, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Thomas Scheibel
- Lehrstuhl für Biomaterialien, Bayerisches Polymerinstitut (BPI), Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Universität Bayreuth, Bayreuth 95440, Germany
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| |
Collapse
|
25
|
Porter L, Minaisah RM, Ahmed S, Ali S, Norton R, Zhang Q, Ferraro E, Molenaar C, Holt M, Cox S, Fountain S, Shanahan C, Warren D. SUN1/2 Are Essential for RhoA/ROCK-Regulated Actomyosin Activity in Isolated Vascular Smooth Muscle Cells. Cells 2020; 9:cells9010132. [PMID: 31935926 PMCID: PMC7017107 DOI: 10.3390/cells9010132] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the blood vessel wall. Changes in VSMC actomyosin activity and morphology are prevalent in cardiovascular disease. The actin cytoskeleton actively defines cellular shape and the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex, comprised of nesprin and the Sad1p, UNC-84 (SUN)-domain family members SUN1/2, has emerged as a key regulator of actin cytoskeletal organisation. Although SUN1 and SUN2 function is partially redundant, they possess specific functions and LINC complex composition is tailored for cell-type-specific functions. We investigated the importance of SUN1 and SUN2 in regulating actomyosin activity and cell morphology in VSMCs. We demonstrate that siRNA-mediated depletion of either SUN1 or SUN2 altered VSMC spreading and impaired actomyosin activity and RhoA activity. Importantly, these findings were recapitulated using aortic VSMCs isolated from wild-type and SUN2 knockout (SUN2 KO) mice. Inhibition of actomyosin activity, using the rho-associated, coiled-coil-containing protein kinase1/2 (ROCK1/2) inhibitor Y27632 or blebbistatin, reduced SUN2 mobility in the nuclear envelope and decreased the association between SUN2 and lamin A, confirming that SUN2 dynamics and interactions are influenced by actomyosin activity. We propose that the LINC complex exists in a mechanical feedback circuit with RhoA to regulate VSMC actomyosin activity and morphology.
Collapse
Affiliation(s)
- Lauren Porter
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Rose-Marie Minaisah
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Sultan Ahmed
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Seema Ali
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Rosemary Norton
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Qiuping Zhang
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Elisa Ferraro
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Chris Molenaar
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Mark Holt
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, King’s College London, London SE1 1YR, UK
| | - Susan Cox
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, King’s College London, London SE1 1YR, UK
| | - Samuel Fountain
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Catherine Shanahan
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
| | - Derek Warren
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King’s College London, London SE5 9NU, UK
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Correspondence:
| |
Collapse
|
26
|
Burn-Induced Impairment of Ileal Muscle Contractility Is Associated with Increased Extracellular Matrix Components. J Gastrointest Surg 2020; 24:188-197. [PMID: 31637625 PMCID: PMC8634548 DOI: 10.1007/s11605-019-04400-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/05/2019] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Severe burns lead to marked impairment of gastrointestinal motility, such as delayed gastric emptying and small and large intestinal ileus. However, the cellular mechanism of these pathologic changes remains largely unknown. METHODS Male Sprague Dawley rats approximately 3 months old and weighing 300-350 g were randomized to either a 60% total body surface area full-thickness scald burn or sham procedure and were sacrificed 24 h after the procedure. Gastric emptying, gastric antrum contractility ileal smooth muscle contractility, and colonic contractility were measured. Muscularis externa was isolated from the ileal segment to prepare smooth muscle protein extracts for Western blot analysis. RESULTS Compared with sham controls, the baseline rhythmic contractile activities of the antral, ileal, and colonic smooth muscle strips were impaired in the burned rats. Simultaneously, our data showed that ileal muscularis ECM proteins fibronectin and laminin were significantly up-regulated in burned rats compared with sham rats. TGF-β signaling is an important stimulating factor for ECM protein expression. Our results revealed that TGF-β signaling was activated in the ileal muscle of burned rats evidenced by the activation of Smad2/3 expression and phosphorylation. In addition, the total and phosphorylated AKT, which is an important downstream factor of ECM signaling in smooth muscle cells, was also up-regulated in burned rats' ileal muscle. Notably, these changes were not seen in the colonic or gastric tissues. CONCLUSION Deposition of fibrosis-related proteins after severe burn is contributors to decreased small intestinal motility.
Collapse
|
27
|
Emerging Concepts and Tools in Cell Mechanomemory. Ann Biomed Eng 2019; 48:2103-2112. [DOI: 10.1007/s10439-019-02412-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/11/2019] [Indexed: 12/25/2022]
|
28
|
Co-immobilization of ACH 11 antithrombotic peptide and CAG cell-adhesive peptide onto vascular grafts for improved hemocompatibility and endothelialization. Acta Biomater 2019; 97:344-359. [PMID: 31377424 DOI: 10.1016/j.actbio.2019.07.057] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 11/20/2022]
Abstract
Surface modification by conjugating biomolecules has been widely proved to enhance biocompatibility of small-caliber artificial vascular grafts. In this study, we aimed at developing a multifunctional vascular graft that provides not only good hemocompatibility but also in situ rapid endothelialization. Herein, a vascular graft (inner diameter ∼2 mm) was fabricated by electrospinning with poly(lactic acid-co-caprolactone) and gelatin, and then biofunctionalized with antithrombotic peptide with sequence LTFPRIVFVLG (ACH11) and cell adhesion peptide with sequence CAG through adhesive poly(dopamine) coating. We developed this graft with the synergistic properties of low thrombogenicity and rapid endothelialization. The successful grafting of both CAG and ACH11 peptides was confirmed by Fourier transform infrared spectroscopy and X-ray photoelectron spectroscopy. The surface micromorphology of the modified surfaces was observed by field emission scanning electron microscopy. Our results demonstrated that the multifunctional surface suppressed the denaturation of absorbed fibrinogen, hindered coagulation factor Xa activation, and inhibited platelet adhesion and aggregation. Importantly, this modified surface could selectively enhance endothelial cells adhesion, proliferation and release of nitric oxide. Upon in vivo implantation of 6 weeks, the multifunctional vascular graft showed improved patency and superior vascular endothelialization. Overall, the results effectively demonstrated that the co-immobilization of ACH11 and CAG provided a promising method for the improvement of hemocompatibility and endothelialization of vascular grafts. STATEMENT OF SIGNIFICANCE: Electrospun small-caliber vascular grafts are increasingly used to treat cardiovascular diseases. Despite their success related to their good biodegradation and mechanical strength, they have some drawbacks, such as low hemocompatibility and endothelialization. The single-function ligands are insufficient to modify surface with both good hemocompatibility and rapid endothelialization simultaneously. Therefore, we functionalized electrospun vascular graft by novel antithrombotic peptide and cell-adhesive peptide to construct superior anticoagulation and ECs-selective adhesion surface in present study. The multifunctional vascular grafts benefit for high long-term patency and rapid endothelialization.
Collapse
|
29
|
Huebsch N. Translational mechanobiology: Designing synthetic hydrogel matrices for improved in vitro models and cell-based therapies. Acta Biomater 2019; 94:97-111. [PMID: 31129361 DOI: 10.1016/j.actbio.2019.05.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/27/2022]
Abstract
Synthetic hydrogels have ideal physiochemical properties to serve as reductionist mimics of the extracellular matrix (ECM) for studies on cellular mechanosensing. These studies range from basic observation of correlations between ECM mechanics and cell fate changes to molecular dissection of the underlying mechanisms. Despite intensive work on hydrogels to study mechanobiology, many fundamental questions regarding mechanosensing remain unanswered. In this review, I first discuss historical motivation for studying cellular mechanobiology, and challenges impeding this effort. I next overview recent efforts to engineer hydrogel properties to study cellular mechanosensing. Finally, I focus on in vitro modeling and cell-based therapies as applications of hydrogels that will exploit our ability to create micro-environments with physiologically relevant elasticity and viscoelasticity to control cell biology. These translational applications will not only use our current understanding of mechanobiology but will also bring new tools to study the fundamental problem of how cells sense their mechanical environment. STATEMENT OF SIGNIFICANCE: Hydrogels are an important tool for understanding how our cells can sense their mechanical environment, and to exploit that understanding in regenerative medicine. In the current review, I discuss historical work linking mechanics to cell behavior in vitro, and highlight the role hydrogels played in allowing us to understand how cells monitor mechanical cues. I then highlight potential translational applications of hydrogels with mechanical properties similar to those of the tissues where cells normally reside in our bodies, and discuss how these types of studies can provide clues to help us enhance our understanding of mechanosensing.
Collapse
Affiliation(s)
- Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, United States.
| |
Collapse
|
30
|
Gooch KJ, Firstenberg MS, Shrefler BS, Scandling BW. Biomechanics and Mechanobiology of Saphenous Vein Grafts. J Biomech Eng 2019; 140:2666246. [PMID: 29222565 DOI: 10.1115/1.4038705] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Indexed: 11/08/2022]
Abstract
Within several weeks of use as coronary artery bypass grafts (CABG), saphenous veins (SV) exhibit significant intimal hyperplasia (IH). IH predisposes vessels to thrombosis and atherosclerosis, the two major modes of vein graft failure. The fact that SV do not develop significant IH in their native venous environment coupled with the rapidity with which they develop IH following grafting into the arterial circulation suggests that factors associated with the isolation and preparation of SV and/or differences between the venous and arterial environments contribute to disease progression. There is strong evidence suggesting that mechanical trauma associated with traditional techniques of SV preparation can significantly damage the vessel and might potentially reduce graft patency though modern surgical techniques reduces these injuries. In contrast, it seems possible that modern surgical technique, specifically endoscopic vein harvest, might introduce other mechanical trauma that could subtly injure the vein and perhaps contribute to the reduced patency observed in veins harvested using endoscopic techniques. Aspects of the arterial mechanical environment influence remodeling of SV grafted into the arterial circulation. Increased pressure likely leads to thickening of the medial wall but its role in IH is less clear. Changes in fluid flow, including increased average wall shear stress, may reduce IH while disturbed flow likely increase IH. Nonmechanical stimuli, such as exposure to arterial levels of oxygen, may also have a significant but not widely recognized role in IH. Several potentially promising approaches to alter the mechanical environment to improve graft patency are including extravascular supports or altered graft geometries are covered.
Collapse
Affiliation(s)
- Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, 290 Bevis Hall 1080 Carmack Drive, Columbus, OH 43210.,Davis Heart Lung Research Institute, The Ohio State University, Columbus, OH 43210 e-mail:
| | - Michael S Firstenberg
- Surgery and Integrative Medicine, Northeast Ohio Medical Universities, Akron, OH 44309
| | - Brittany S Shrefler
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Benjamin W Scandling
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
31
|
Yosef A, Kossover O, Mironi‐Harpaz I, Mauretti A, Melino S, Mizrahi J, Seliktar D. Fibrinogen-Based Hydrogel Modulus and Ligand Density Effects on Cell Morphogenesis in Two-Dimensional and Three-Dimensional Cell Cultures. Adv Healthc Mater 2019; 8:e1801436. [PMID: 31081289 DOI: 10.1002/adhm.201801436] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/08/2019] [Indexed: 12/15/2022]
Abstract
There is a need to further explore the convergence of mechanobiology and dimensionality with systematic investigations of cellular response to matrix mechanics in 2D and 3D cultures. Here, a semisynthetic hydrogel capable of supporting both 2D and 3D cell culture is applied to investigate cell response to matrix modulus and ligand density. The culture materials are fabricated from adducts of polyethylene glycol (PEG) or PluronicF127 and fibrinogen fragments, formed into hydrogels by free-radical polymerization, and characterized by shear rheology. Control over the modulus of the materials is accomplished by changing the concentration of synthetic PEG-diacrylate crosslinker (0.5% w/v), and by altering the molecular length of the PEG (10 and 20 kDa). Control over ligand density is accomplished by changing fibrinogen concentrations from 3 to 12 mg mL-1 . In 2D culture, cell motility parameters, including cell speed and persistence time are significantly increased with increasing modulus. In both 2D and 3D culture, cells express vinculin and there is evidence of focal adhesion formation in the high stiffness materials. The modulus- and ligand-dependent morphogenesis response from the cells in 2D culture is contradictory to the same measured response in 3D culture. In 2D culture, anchorage-dependent cells become more elongated and significantly increase their size with increasing ligand density and matrix modulus. In 3D culture, the same anchorage-dependent cells become less spindled and significantly reduce their size in response to increasing ligand density and matrix modulus. These differences arise from dimensionality constraints, most notably the encapsulation of cells in a non-porous hydrogel matrix. These insights underscore the importance of mechanical properties in regulating cell morphogenesis in a 3D culture milieu. The versatility of the hydrogel culture environment further highlights the significance of a modular approach when developing materials that aim to optimize the cell culture environment.
Collapse
Affiliation(s)
- Andrei Yosef
- Faculty of Biomedical EngineeringTechnion—Israel Institute of Technology Haifa 32000 Israel
| | - Olga Kossover
- Faculty of Biomedical EngineeringTechnion—Israel Institute of Technology Haifa 32000 Israel
| | - Iris Mironi‐Harpaz
- Faculty of Biomedical EngineeringTechnion—Israel Institute of Technology Haifa 32000 Israel
| | - Arianna Mauretti
- Department of Chemical Sciences and TechnologiesUniversity of Rome “Tor Vergata” Via della Ricerca Scientifica 1 00133 Rome Italy
| | - Sonia Melino
- Department of Chemical Sciences and TechnologiesUniversity of Rome “Tor Vergata” Via della Ricerca Scientifica 1 00133 Rome Italy
- CIMER Center of Regenerative MedicineTor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| | - Joseph Mizrahi
- Faculty of Biomedical EngineeringTechnion—Israel Institute of Technology Haifa 32000 Israel
| | - Dror Seliktar
- Faculty of Biomedical EngineeringTechnion—Israel Institute of Technology Haifa 32000 Israel
| |
Collapse
|
32
|
Schwager SC, Bordeleau F, Zhang J, Antonyak MA, Cerione RA, Reinhart-King CA. Matrix stiffness regulates microvesicle-induced fibroblast activation. Am J Physiol Cell Physiol 2019; 317:C82-C92. [PMID: 31017799 PMCID: PMC6689748 DOI: 10.1152/ajpcell.00418.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 11/22/2022]
Abstract
Extracellular vesicles released by cancer cells have recently been implicated in the differentiation of stromal cells to their activated, cancer-supporting states. Microvesicles, a subset of extracellular vesicles released from the plasma membrane of cancer cells, contain biologically active cargo, including DNA, mRNA, and miRNA, which are transferred to recipient cells and induce a phenotypic change in behavior. While it is known that microvesicles can alter recipient cell phenotype, little is known about how the physical properties of the tumor microenvironment affect fibroblast response to microvesicles. Here, we utilized cancer cell-derived microvesicles and synthetic substrates designed to mimic the stiffness of the tumor and tumor stroma to investigate the effects of microvesicles on fibroblast phenotype as a function of the mechanical properties of the microenvironment. We show that microvesicles released by highly malignant breast cancer cells cause an increase in fibroblast spreading, α-smooth muscle actin expression, proliferation, cell-generated traction force, and collagen gel compaction. Notably, our data indicate that these phenotypic changes occur only on stiff matrices mimicking the stiffness of the tumor periphery and are dependent on the cell type from which the microvesicles are shed. Overall, these results show that the effects of cancer cell-derived microvesicles on fibroblast activation are regulated by the physical properties of the microenvironment, and these data suggest that microvesicles may have a more robust effect on fibroblasts located at the tumor periphery to influence cancer progression.
Collapse
Affiliation(s)
- Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Marc A Antonyak
- Department of Biomedical Science, Cornell University , Ithaca, New York
| | - Richard A Cerione
- Department of Biomedical Science, Cornell University , Ithaca, New York
- Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York
| | | |
Collapse
|
33
|
Sit B, Gutmann D, Iskratsch T. Costameres, dense plaques and podosomes: the cell matrix adhesions in cardiovascular mechanosensing. J Muscle Res Cell Motil 2019; 40:197-209. [PMID: 31214894 PMCID: PMC6726830 DOI: 10.1007/s10974-019-09529-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022]
Abstract
The stiffness of the cardiovascular environment changes during ageing and in disease and contributes to disease incidence and progression. For instance, increased arterial stiffness can lead to atherosclerosis, while stiffening of the heart due to fibrosis can increase the chances of heart failure. Cells can sense the stiffness of the extracellular matrix through integrin adhesions and other mechanosensitive structures and in response to this initiate mechanosignalling pathways that ultimately change the cellular behaviour. Over the past decades, interest in mechanobiology has steadily increased and with this also our understanding of the molecular basis of mechanosensing and transduction. However, much of our knowledge about the mechanisms is derived from studies investigating focal adhesions in non-muscle cells, which are distinct in several regards from the cell-matrix adhesions in cardiomyocytes (costameres) or vascular smooth muscle cells (dense plaques or podosomes). Therefore, we will look here first at the evidence for mechanical sensing in the cardiovascular system, before comparing the different cytoskeletal arrangements and adhesion sites in cardiomyocytes and vascular smooth muscle cells and what is known about mechanical sensing through the various structures.
Collapse
Affiliation(s)
- Brian Sit
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Daniel Gutmann
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK.
| |
Collapse
|
34
|
Yi B, Shen Y, Tang H, Wang X, Li B, Zhang Y. Stiffness of Aligned Fibers Regulates the Phenotypic Expression of Vascular Smooth Muscle Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:6867-6880. [PMID: 30676736 DOI: 10.1021/acsami.9b00293] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electrospun uniaxially aligned ultrafine fibers show great promise in constructing vascular grafts mimicking the anisotropic architecture of native blood vessels. However, understanding how the stiffness of aligned fibers would impose influences on the functionality of vascular cells has yet to be explored. The present study aimed to explore the stiffness effects of electrospun aligned fibrous substrates (AFSs) on phenotypic modulation in vascular smooth muscle cells (SMCs). A stable jet coaxial electrospinning (SJCES) method was employed to generate highly aligned ultrafine fibers of poly(l-lactide- co-caprolactone)/poly(l-lactic acid) (PLCL/PLLA) in shell-core configuration with a remarkably varying stiffness region from 0.09 to 13.18 N/mm. We found that increasing AFS stiffness had no significant influence on the cellular shape and orientation along the fiber direction with the cultured human umbilical artery SMCs (huaSMCs) but inhibited the cell adhesion rate, promoted cell proliferation and migration, and especially enhanced the F-actin fiber assembly in the huaSMCs. Notably, higher fiber stiffness resulted in significant downregulation of contractile markers like alpha-smooth muscle actin (α-SMA), smooth muscle myosin heavy chain, calponin, and desmin, whereas upregulated the gene expression of pathosis-associated osteopontin ( OPN) in the huaSMCs. These results allude to the phenotype of huaSMCs on stiffer AFSs being miserably modulated into a proliferative and pathological state. Consequently, it adversely affected the proliferation and migration behavior of human umbilical vein endothelial cells as well. Moreover, stiffer AFSs also revealed to incur significant upregulation of inflammatory gene expression, such as interleukin-6 ( IL-6), monocyte chemoattractant protein-1 ( MCP-1), and intercellular adhesion molecule-1 ( ICAM-1), in the huaSMCs. This study stresses that although electrospun aligned fibers are capable of modulating native-like oriented cell morphology and even desired phenotype realization or transition, they might not always direct cells into correct functionality. The integrated fiber stiffness underlying is thereby a critical parameter to consider in engineering structurally anisotropic tissue-engineered vascular grafts to ultimately achieve long-term patency.
Collapse
Affiliation(s)
| | | | | | | | - Bin Li
- Department of Orthopaedics , The First Affiliated Hospital of Soochow University , Suzhou 215006 , China
- Orthopaedic Institute, Medical College , Soochow University , Suzhou 215007 , China
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| | - Yanzhong Zhang
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| |
Collapse
|
35
|
Peter M, Singh A, Mohankumar K, Jeenger R, Joge PA, Gatne MM, Tayalia P. Gelatin-Based Matrices as a Tunable Platform To Study in Vitro and in Vivo 3D Cell Invasion. ACS APPLIED BIO MATERIALS 2019; 2:916-929. [PMID: 35016295 DOI: 10.1021/acsabm.8b00767] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydrogels have been used as synthetic mimics of 3D extracellular matrices (ECM) and their physical properties like stiffness, degradability, and porosity have been known to influence the behavior of encapsulated cells. However, to understand the role of individual properties, the influence of biophysical cues should be decoupled from biochemical ones. In this study, we have used hydrogels as a tunable model matrix to develop a 3D cell culture platform for studying cell invasion. Inert polyethylene (glycol) diacrylate (PEGDA) and cell adhesive gelatin methacryloyl (GELMA) were blended in varying compositions, followed by UV-mediated photo polymerization to obtain hydrogels with varying stiffness, degradation, and cell adhesive properties. We developed two hydrogel matrix systems, namely, PEGDA-GELMA (containing a larger proportion of PEGDA) and GELMA-PEGDA (containing predominantly GELMA), and characterized them for differences in pore size, swelling ratio, storage modulus, degradability, and biocompatibility of the matrix. Both hydrogel systems had similar pore dimensions and swelling behavior, but PEGDA-GELMA was found to be stiffer and nondegradable, while GELMA-PEGDA was softer and degradable. Accordingly, MDA-MB-231 breast cancer cells encapsulated in these matrices showed a spheroidal morphology in PEGDA-GELMA hydrogels and were more spindle-shaped in GELMA-PEGDA hydrogels, confirming that size and extent of spreading of cells were influenced by the type of these hydrogels. The softer GELMA-PEGDA matrices readily allowed invasion of MDA-MB-231 cells in 3D and showed differences in epithelial-mesenchymal transition (EMT) gene expression of these cells. We further demonstrated the invasion and sprouting of endothelial cells using a chick aortic arch assay, exhibiting the utility of softer matrices to study 3D cell invasion for multiple applications. We also implanted these matrices in mice and showed that soft gelatin-based hydrogels allow cell infiltration in vivo. Results from our study highlight the tunability of this matrix system and the role of matrix constitution in influencing cell invasion in a 3D microenvironment.
Collapse
Affiliation(s)
- Mathew Peter
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.,Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Archana Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Kumaravel Mohankumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.,Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843, United States
| | - Rajeev Jeenger
- Department of Veterinary Pharmacology and Toxicology, Bombay Veterinary College, Parel, Mumbai 400012, India
| | - Puja Arun Joge
- Department of Veterinary Pharmacology and Toxicology, Bombay Veterinary College, Parel, Mumbai 400012, India
| | - Madhumanjiri Mukulesh Gatne
- Department of Veterinary Pharmacology and Toxicology, Bombay Veterinary College, Parel, Mumbai 400012, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
36
|
Nabavinia M, Khoshfetrat AB, Naderi-Meshkin H. Nano-hydroxyapatite-alginate-gelatin microcapsule as a potential osteogenic building block for modular bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:67-77. [PMID: 30678955 DOI: 10.1016/j.msec.2018.12.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 11/08/2018] [Accepted: 12/10/2018] [Indexed: 01/01/2023]
Abstract
To develop osteogenic building blocks for modular bone tissue engineering applications, influence of gelatin as cell adhesive molecule and nano-hydroxyapatite (nHA) as osteoconductive component was examined on alginate-based hydrogel properties and microencapsulated osteoblast-like cell behavior by using factorial experimental design technique. nHA and alginate showed a statistically significant impact on swelling reduction, and improvement of stability and mechanical strength of hydrogels, respectively. Gelatin influence, however, was in a reverse manner. nHA played imperative roles in promoting microencapsulated osteoblastic cell proliferation and function due to its bioactivity and mechanical strength improvement of hydrogels to the modulus range of mineralized bone tissue in vivo. The results and their statistical analysis also revealed the importance of interaction effect of gelatin and nHA. Proliferation and osteogenic function of the cells fluctuated with increasing gelatin concentration of microcapsules in the presence of nHA, demonstrating that hydrogel properties should be balanced to provide an efficient 3D osteoconductive microcapsule. Alginate (1%)-gelatin (2.5%)-nHA (0.5%) microcapsule with compressive modulus of 0.19 MPa ± 0.02, swelling ratio of 52% ± 8 (24 h) and degradation rate of 12% ± 4 (96 h) revealed a maximum performance for the cell proliferation and function, indicating a potential microcapsule composition to prepare building blocks for modular bone tissue engineering.
Collapse
Affiliation(s)
- Mahboubeh Nabavinia
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran
| | - Ali Baradar Khoshfetrat
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran.
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Academic Center of Education, Culture, and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| |
Collapse
|
37
|
Chester D, Kathard R, Nortey J, Nellenbach K, Brown AC. Viscoelastic properties of microgel thin films control fibroblast modes of migration and pro-fibrotic responses. Biomaterials 2018; 185:371-382. [DOI: 10.1016/j.biomaterials.2018.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/20/2018] [Accepted: 09/07/2018] [Indexed: 12/22/2022]
|
38
|
Garoffolo G, Madonna R, de Caterina R, Pesce M. Cell based mechanosensing in vascular patho-biology: More than a simple go-with the flow. Vascul Pharmacol 2018; 111:7-14. [DOI: 10.1016/j.vph.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/10/2018] [Accepted: 06/16/2018] [Indexed: 12/12/2022]
|
39
|
Zhu Y, Li X, Janairo RRR, Kwong G, Tsou AD, Chu JS, Wang A, Yu J, Wang D, Li S. Matrix stiffness modulates the differentiation of neural crest stem cells in vivo. J Cell Physiol 2018; 234:7569-7578. [PMID: 30368818 DOI: 10.1002/jcp.27518] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Stem cells are often transplanted with scaffolds for tissue regeneration; however, how the mechanical property of a scaffold modulates stem cell fate in vivo is not well understood. Here we investigated how matrix stiffness modulates stem cell differentiation in a model of vascular graft transplantation. Multipotent neural crest stem cells (NCSCs) were differentiated from induced pluripotent stem cells, embedded in the hydrogel on the outer surface of nanofibrous polymer grafts, and implanted into rat carotid arteries by anastomosis. After 3 months, NCSCs differentiated into smooth muscle cells (SMCs) near the outer surface of the polymer grafts; in contrast, NCSCs differentiated into glial cells in the most part of the hydrogel. Atomic force microscopy demonstrated a stiffer matrix near the polymer surface but much lower stiffness away from the polymer graft. Consistently, in vitro studies confirmed that stiff surface induced SMC genes whereas soft surface induced glial genes. These results suggest that the scaffold's mechanical properties play an important role in directing stem cell differentiation in vivo, which has important implications in biomaterials design for stem cell delivery and tissue engineering.
Collapse
Affiliation(s)
- Yiqian Zhu
- Department of Bioengineering, University of California, Berkeley, California.,Department of Neurosurgery, Fudan University Huashan Hospital, Shanghai, China
| | - Xian Li
- Department of Bioengineering, University of California, Berkeley, California.,Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | | | - George Kwong
- Department of Bioengineering, University of California, Berkeley, California
| | - Anchi D Tsou
- Department of Bioengineering, University of California, Berkeley, California
| | - Julia S Chu
- Department of Bioengineering, University of California, Berkeley, California
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Sacramento, California
| | - Jian Yu
- Department of Bioengineering, University of California, Berkeley, California.,Department of Neurosurgery, Fudan University Huashan Hospital, Shanghai, China
| | - Dong Wang
- Department of Bioengineering, University of California, Los Angeles, California
| | - Song Li
- Department of Bioengineering, University of California, Berkeley, California.,Department of Bioengineering, University of California, Los Angeles, California.,Department of Medicine, University of California, Los Angeles, California
| |
Collapse
|
40
|
Schöneberg J, De Lorenzi F, Theek B, Blaeser A, Rommel D, Kuehne AJC, Kießling F, Fischer H. Engineering biofunctional in vitro vessel models using a multilayer bioprinting technique. Sci Rep 2018; 8:10430. [PMID: 29992981 PMCID: PMC6041340 DOI: 10.1038/s41598-018-28715-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of bioprinting have led to the development of perfusable complex structures. However, most of the existing printed vascular channels lack the composition or key structural and physiological features of natural blood vessels or they make use of more easily printable but less biocompatible hydrogels. Here, we use a drop-on-demand bioprinting technique to generate in vitro blood vessel models, consisting of a continuous endothelium imitating the tunica intima, an elastic smooth muscle cell layer mimicking the tunica media, and a surrounding fibrous and collagenous matrix of fibroblasts mimicking the tunica adventitia. These vessel models with a wall thickness of up to 425 µm and a diameter of about 1 mm were dynamically cultivated in fluidic bioreactors for up to three weeks under physiological flow conditions. High cell viability (>83%) after printing and the expression of VE-Cadherin, smooth muscle actin, and collagen IV were observed throughout the cultivation period. It can be concluded that the proposed novel technique is suitable to achieve perfusable vessel models with a biofunctional multilayer wall composition. Such structures hold potential for the creation of more physiologically relevant in vitro disease models suitable especially as platforms for the pre-screening of drugs.
Collapse
Affiliation(s)
- Jan Schöneberg
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Blaeser
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Dirk Rommel
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Alexander J C Kuehne
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Fabian Kießling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
41
|
Ding Y, Xu X, Sharma S, Floren M, Stenmark K, Bryant SJ, Neu CP, Tan W. Biomimetic soft fibrous hydrogels for contractile and pharmacologically responsive smooth muscle. Acta Biomater 2018; 74:121-130. [PMID: 29753912 DOI: 10.1016/j.actbio.2018.05.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 01/22/2023]
Abstract
The ability to assess changes in smooth muscle contractility and pharmacological responsiveness in normal or pathological-relevant vascular tissue environments is critical to enable vascular drug discovery. However, major challenges remain in both capturing the complexity of in vivo vascular remodeling and evaluating cell contractility in complex, tissue-like environments. Herein, we developed a biomimetic fibrous hydrogel with tunable structure, stiffness, and composition to resemble the native vascular tissue environment. This hydrogel platform was further combined with the combinatory protein array technology as well as advanced approaches to measure cell mechanics and contractility, thus permitting evaluation of smooth muscle functions in a variety of tissue-like microenvironments. Our results demonstrated that biomimetic fibrous structure played a dominant role in smooth muscle function, while the presentation of adhesion proteins co-regulated it to various degrees. Specifically, fibre networks enabled cell infiltration and upregulated expression of actomyosin proteins in contrast to flat hydrogels. Remarkably, fibrous structure and physiologically relevant stiffness of hydrogels cooperatively enhanced smooth muscle contractility and pharmacological responses to vasoactive drugs at both the single cell and intact tissue levels. Together, this study is the first to demonstrate alterations of human vascular smooth muscle contractility and pharmacological responsiveness in biomimetic soft, fibrous environments with a cellular array platform. The integrated platform produced here could enable investigations for pathobiology and pharmacological interventions by developing a broad range of patho-physiologically relevant in vitro tissue models. STATEMENT OF SIGNIFICANCE Engineering functional smooth muscle in vitro holds the great potential for diseased tissue replacement and drug testing. A central challenge is recapitulating the smooth muscle contractility and pharmacological responses given its significant phenotypic plasticity in response to changes in environment. We present a biomimetic fibrous hydrogel with tunable structure, stiffness, and composition that enables the creation of functional smooth muscle tissues in the native-like vascular tissue microenvironment. Such fibrous hydrogel is further combined with the combinatory protein array technology to construct a cellular array for evaluation of smooth muscle phenotype, contraction, and cell mechanics. The integrated platform produced here could be promising for developing a broad range of normal or diseased in vitro tissue models.
Collapse
Affiliation(s)
- Yonghui Ding
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Xin Xu
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Sadhana Sharma
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Michael Floren
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; Cardiovascular Pulmonary Research Laboratories, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; BioFrontiers Institute, Material Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Wei Tan
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
42
|
Cooke ME, Jones SW, Ter Horst B, Moiemen N, Snow M, Chouhan G, Hill LJ, Esmaeli M, Moakes RJA, Holton J, Nandra R, Williams RL, Smith AM, Grover LM. Structuring of Hydrogels across Multiple Length Scales for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705013. [PMID: 29430770 DOI: 10.1002/adma.201705013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/20/2017] [Indexed: 06/08/2023]
Abstract
The development of new materials for clinical use is limited by an onerous regulatory framework, which means that taking a completely new material into the clinic can make translation economically unfeasible. One way to get around this issue is to structure materials that are already approved by the regulator, such that they exhibit very distinct physical properties and can be used in a broader range of clinical applications. Here, the focus is on the structuring of soft materials at multiple length scales by modifying processing conditions. By applying shear to newly forming materials, it is possible to trigger molecular reorganization of polymer chains, such that they aggregate to form particles and ribbon-like structures. These structures then weakly interact at zero shear forming a solid-like material. The resulting self-healing network is of particular use for a range of different biomedical applications. How these materials are used to allow the delivery of therapeutic entities (cells and proteins) and as a support for additive layer manufacturing of larger-scale tissue constructs is discussed. This technology enables the development of a range of novel materials and structures for tissue augmentation and regeneration.
Collapse
Affiliation(s)
- Megan E Cooke
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Inflammation and Ageing, MRC Musculoskeletal Ageing Centre, QE Hospital, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC Musculoskeletal Ageing Centre, QE Hospital, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Britt Ter Horst
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Scar Free Foundation Centre for Burns Research, QE Hospital, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Naiem Moiemen
- Scar Free Foundation Centre for Burns Research, QE Hospital, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Martyn Snow
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Gurpreet Chouhan
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Lisa J Hill
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Maryam Esmaeli
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Richard J A Moakes
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - James Holton
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Rajpal Nandra
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Richard L Williams
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Alan M Smith
- Department of Pharmacy, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Liam M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
43
|
Gao J, Jiang L, Liang Q, Shi J, Hou D, Tang D, Chen S, Kong D, Wang S. The grafts modified by heparinization and catalytic nitric oxide generation used for vascular implantation in rats. Regen Biomater 2018; 5:105-114. [PMID: 29644092 PMCID: PMC5888227 DOI: 10.1093/rb/rby003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
Small-diameter (<6 mm) vascular grafts are increasingly needed in peripheral vascular surgery but have few successes because of acute thrombosis, incomplete endothelialization and intimal hyperplasia after implantation. This study used electrospun poly(ε-caprolactone) as the matrix material. Heparin and selenium-containing catalyst-organoselenium modified polyethyleneimine were introduced through layer-by-layer assembly in order to build a vascular graft with in situ nitric oxide (NO) generation. The aim of this study was to explore the application of the graft with improved histocompatibility and biological function for vascular implantation in rats. After implantation in rats, compared to poly(ε-caprolactone), the modified grafts could promote the adhesion and proliferation of endothelial cells, and inhibit the adhesion of smooth muscle cells. The modified grafts remarkably promoted endothelialization, inhibited intimal hyperplasia and increased the ratio of alternatively activated macrophages (M2) to classical activated macrophages (M1). This work constructed a vascular graft with heparinization and catalytic NO generation for improving the vascularization, and accelerating the tissue regeneration by regulating the inflammatory response. The present study indicates that it is a promising method for regulating response and tissue regeneration of small diameter vascular grafts by a novel approach of combining heparinization and catalytic NO generation.
Collapse
Affiliation(s)
- Jingchen Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Li Jiang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qinge Liang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ding Hou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Di Tang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Siyuan Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shufang Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
44
|
Sprouting angiogenesis induces significant mechanical heterogeneities and ECM stiffening across length scales in fibrin hydrogels. Biomaterials 2018; 162:99-108. [PMID: 29438884 DOI: 10.1016/j.biomaterials.2018.02.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/21/2018] [Accepted: 02/03/2018] [Indexed: 11/23/2022]
Abstract
Matrix stiffness is a well-established instructive cue in two-dimensional cell cultures. Its roles in morphogenesis in 3-dimensional (3D) cultures, and the converse effects of cells on the mechanics of their surrounding microenvironment, have been more elusive given the absence of suitable methods to quantify stiffness on a length-scale relevant for individual cell-extracellular matrix (ECM) interactions. In this study, we applied traditional bulk rheology and laser tweezers-based active microrheology to probe mechanics across length scales during the complex multicellular process of capillary morphogenesis in 3D, and further characterized the relative contributions of neovessels and supportive stromal cells to dynamic changes in stiffness over time. Our data show local ECM stiffness was highly heterogeneous around sprouting capillaries, and the variation progressively increased with time. Both endothelial cells and stromal support cells progressively stiffened the ECM, with the changes in bulk properties dominated by the latter. Interestingly, regions with high micro-stiffness did not necessarily correlate with remodeled regions of high ECM density as shown by confocal reflectance microscopy. Collectively, these findings, especially the large spatiotemporal variations in local stiffness around cells during morphogenesis in soft 3D fibrin gels, underscore that characterizing ECM mechanics across length scales. provides an opportunity to attain a deeper mechanobiological understanding of the microenvironment's roles in cell fate and tissue patterning.
Collapse
|
45
|
Xie SA, Zhang T, Wang J, Zhao F, Zhang YP, Yao WJ, Hur SS, Yeh YT, Pang W, Zheng LS, Fan YB, Kong W, Wang X, Chiu JJ, Zhou J. Matrix stiffness determines the phenotype of vascular smooth muscle cell in vitro and in vivo: Role of DNA methyltransferase 1. Biomaterials 2018; 155:203-216. [DOI: 10.1016/j.biomaterials.2017.11.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/23/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
|
46
|
Brooks EA, Jansen LE, Gencoglu MF, Yurkevicz AM, Peyton SR. Complementary, Semiautomated Methods for Creating Multidimensional PEG-Based Biomaterials. ACS Biomater Sci Eng 2018; 4:707-718. [PMID: 33418758 DOI: 10.1021/acsbiomaterials.7b00737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tunable biomaterials that mimic selected features of the extracellular matrix (ECM) such as its stiffness, protein composition, and dimensionality are increasingly popular for studying how cells sense and respond to ECM cues. In the field, there exists a significant trade-off for how complex and how well these biomaterials represent the in vivo microenvironment versus how easy they are to make and how adaptable they are to automated fabrication techniques. To address this need to integrate more complex biomaterials design with high-throughput screening approaches, we present several methods to fabricate synthetic biomaterials in 96-well plates and demonstrate that they can be adapted to semiautomated liquid handling robotics. These platforms include (1) glass bottom plates with covalently attached ECM proteins and (2) hydrogels with tunable stiffness and protein composition with either cells seeded on the surface or (3) laden within the three-dimensional hydrogel matrix. This study includes proof-of-concept results demonstrating control over breast cancer cell line phenotypes via these ECM cues in a semiautomated fashion. We foresee the use of these methods as a mechanism to bridge the gap between high-throughput cell-matrix screening and engineered ECM-mimicking biomaterials.
Collapse
Affiliation(s)
- Elizabeth A Brooks
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Maria F Gencoglu
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Annali M Yurkevicz
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| |
Collapse
|
47
|
Tseng H, Daquinag AC, Souza GR, Kolonin MG. Three-Dimensional Magnetic Levitation Culture System Simulating White Adipose Tissue. Methods Mol Biol 2018; 1773:147-154. [PMID: 29687387 DOI: 10.1007/978-1-4939-7799-4_12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
White adipose tissue (WAT) has attracted interest for tissue engineering and cell-based therapies as an abundant source of adipose stem/stromal cells (ASC). However, technical challenges in WAT cell culture have limited its applications in regenerative medicine. Traditional two-dimensional (2D) cell culture models, which are essentially monolayers of cells on glass or plastic substrates, inadequately represent tissue architecture, biochemical concentration gradients, substrate stiffness, and most importantly for WAT research, cell phenotypic heterogeneity. Physiological cell culture platforms for WAT modeling must recapitulate the native diversity of cell types and their coordination within the organ. For this purpose, we developed a three-dimensional (3D) model using magnetic levitation. Here, we describe our protocol that we successfully employed to build adipose tissue organoids (adipospheres) that preserve the heterogeneity of the constituent cell types in vitro. We demonstrate the capacity of assembling adipospheres from multiple cell types, including ASCs, endohtelial cells, and leukocytes that recreate tissue organization. These adipospheres mimicked WAT organogenesis in that they enabled the formation of vessel-like endothelial structures with lumens and differentiation of unilocular adipocytes. Altogether, magnetic levitation is a cell culture platform that recreates tissue structure, function, and heterogeneity in vitro, and serves as a foundation for high-throughput WAT tissue culture and analysis.
Collapse
Affiliation(s)
| | - Alexes C Daquinag
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Glauco R Souza
- Nano3D Biosciences, Houston, TX, USA.,Oncology Division, Department of Internal Medicine, UT Health Science Center at Houston, Houston, TX, USA
| | - Mikhail G Kolonin
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
48
|
Türker E, Demirçak N, Arslan-Yildiz A. Scaffold-free three-dimensional cell culturing using magnetic levitation. Biomater Sci 2018; 6:1745-1753. [DOI: 10.1039/c8bm00122g] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Magnetic levitation platform ensures a scaffold-free 3D cell culture formation by utilizing Gadolinium(iii) chelates, which provide paramagnetic environment for levitation; therefore, the cells are assembled into complex 3D structures.
Collapse
Affiliation(s)
- Esra Türker
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| | - Nida Demirçak
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| | - Ahu Arslan-Yildiz
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| |
Collapse
|
49
|
Cheng B, Lin M, Huang G, Li Y, Ji B, Genin GM, Deshpande VS, Lu TJ, Xu F. Cellular mechanosensing of the biophysical microenvironment: A review of mathematical models of biophysical regulation of cell responses. Phys Life Rev 2017; 22-23:88-119. [PMID: 28688729 PMCID: PMC5712490 DOI: 10.1016/j.plrev.2017.06.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 12/11/2022]
Abstract
Cells in vivo reside within complex microenvironments composed of both biochemical and biophysical cues. The dynamic feedback between cells and their microenvironments hinges upon biophysical cues that regulate critical cellular behaviors. Understanding this regulation from sensing to reaction to feedback is therefore critical, and a large effort is afoot to identify and mathematically model the fundamental mechanobiological mechanisms underlying this regulation. This review provides a critical perspective on recent progress in mathematical models for the responses of cells to the biophysical cues in their microenvironments, including dynamic strain, osmotic shock, fluid shear stress, mechanical force, matrix rigidity, porosity, and matrix shape. The review highlights key successes and failings of existing models, and discusses future opportunities and challenges in the field.
Collapse
Affiliation(s)
- Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baohua Ji
- Biomechanics and Biomaterials Laboratory, Department of Applied Mechanics, Beijing Institute of Technology, Beijing, China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Mechanical Engineering & Materials Science, and NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis 63130, MO, USA
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, United Kingdom
| | - Tian Jian Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
50
|
Seeto WJ, Tian Y, Winter RL, Caldwell FJ, Wooldridge AA, Lipke EA. Encapsulation of Equine Endothelial Colony Forming Cells in Highly Uniform, Injectable Hydrogel Microspheres for Local Cell Delivery. Tissue Eng Part C Methods 2017; 23:815-825. [DOI: 10.1089/ten.tec.2017.0233] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Wen J. Seeto
- Department of Chemical Engineering, Auburn University, Auburn, Alabama
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, Alabama
| | - Randolph L. Winter
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Fred J. Caldwell
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Anne A. Wooldridge
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | | |
Collapse
|