1
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
2
|
Wu Q, Li L, Zhang Y, Ming X, Feng N. Measurement methods, influencing factors and applications of intercellular receptor-ligand binding kinetics in diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:43-54. [PMID: 39491758 DOI: 10.1016/j.pbiomolbio.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Receptor-ligand binding on contacting cells dictates the extent of transmembrane signaling through membrane receptors during cell communication, influencing both the physiological and pathological activities of cells. This process is integral to fundamental biological mechanisms including signal transduction, cancer metastasis, immune responses, and inflammatory cascades, all of which are profoundly influenced by the cell microenvironment. This article provides an overview of the kinetic theory of receptor-ligand binding and examines methods for measuring this interaction, along with their respective advantages and disadvantages. Furthermore, it comprehensively explores the factors that impact receptor-ligand binding, encompassing protein-membrane interactions, the bioelectric microenvironment, auxiliary factors, hydrogen bond strength, pH levels, cis and trans interactions between ligands and receptors. The application of receptor-ligand binding kinetics in various diseases such as immunity, cancer, and inflammation are also discussed. Additionally, the investigation into how functional substances alter receptor-ligand binding dynamics within specific cellular microenvironments presents a promising new approach to treating related diseases.
Collapse
Affiliation(s)
- Qian Wu
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Liangchao Li
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Yuyan Zhang
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Xiaozhi Ming
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Nianjie Feng
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| |
Collapse
|
3
|
Din M, Paul S, Ullah S, Yang H, Xu RG, Abidin NAZ, Sun A, Chen YC, Gao R, Chowdhury B, Zhou F, Rogers S, Miller M, Biswas A, Hu L, Fan Z, Zahner C, Fan J, Chen Z, Berman M, Xue L, Ju LA, Chen Y. Multi-parametric thrombus profiling microfluidics detects intensified biomechanical thrombogenesis associated with hypertension and aging. Nat Commun 2024; 15:9067. [PMID: 39433750 PMCID: PMC11494109 DOI: 10.1038/s41467-024-53069-9] [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: 01/03/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
Arterial thrombosis is a leading cause of death and disability worldwide with no effective bioassay for clinical prediction. As a symbolic feature of arterial thrombosis, severe stenosis in the blood vessel creates a high-shear, high-gradient flow environment that facilitates platelet aggregation towards vessel occlusion. Here, we present a thrombus profiling assay that monitors the multi-dimensional attributes of thrombi forming in such biomechanical conditions. Using this assay, we demonstrate that different receptor-ligand interactions contribute distinctively to the composition and activation status of the thrombus. Our investigation into hypertensive and older individuals reveals intensified biomechanical thrombogenesis and multi-dimensional thrombus profile abnormalities, endorsing the diagnostic potential of the assay. Furthermore, we identify the hyperactivity of GPIbα-integrin αIIbβ3 mechanosensing axis as a molecular mechanism that contributes to hypertension-associated arterial thrombosis. By studying drug-disease interactions and inter-individual variability, our work reveals a need for personalized anti-thrombotic drug selection that accommodates each patient's pathological profile.
Collapse
Affiliation(s)
- Misbahud Din
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Souvik Paul
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sana Ullah
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Haoyi Yang
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Rong-Guang Xu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Allan Sun
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yiyao Catherine Chen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Rui Gao
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Bari Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Stephenie Rogers
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mariel Miller
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Atreyee Biswas
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, 06030, USA
| | - Christopher Zahner
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jing Fan
- Department of Mechanical Engineering, The City University of New York - City College, New York, NY, 10031, USA
| | - Zi Chen
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Megan Berman
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
4
|
Sala S, Caillier A, Oakes PW. Principles and regulation of mechanosensing. J Cell Sci 2024; 137:jcs261338. [PMID: 39297391 PMCID: PMC11423818 DOI: 10.1242/jcs.261338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
Research over the past two decades has highlighted that mechanical signaling is a crucial component in regulating biological processes. Although many processes and proteins are termed 'mechanosensitive', the underlying mechanisms involved in mechanosensing can vary greatly. Recent studies have also identified mechanosensing behaviors that can be regulated independently of applied force. This important finding has major implications for our understanding of downstream mechanotransduction, the process by which mechanical signals are converted into biochemical signals, as it offers another layer of biochemical regulatory control for these crucial signaling pathways. In this Review, we discuss the different molecular and cellular mechanisms of mechanosensing, how these processes are regulated and their effects on downstream mechanotransduction. Together, these discussions provide an important perspective on how cells and tissues control the ways in which they sense and interpret mechanical signals.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Alexia Caillier
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| |
Collapse
|
5
|
Dasetty S, Bidone TC, Ferguson AL. Data-driven prediction of α IIbβ 3 integrin activation paths using manifold learning and deep generative modeling. Biophys J 2024; 123:2716-2729. [PMID: 38098231 PMCID: PMC11393677 DOI: 10.1016/j.bpj.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024] Open
Abstract
The integrin heterodimer is a transmembrane protein critical for driving cellular process and is a therapeutic target in the treatment of multiple diseases linked to its malfunction. Activation of integrin involves conformational transitions between bent and extended states. Some of the conformations that are intermediate between bent and extended states of the heterodimer have been experimentally characterized, but the full activation pathways remain unresolved both experimentally due to their transient nature and computationally due to the challenges in simulating rare barrier crossing events in these large molecular systems. An understanding of the activation pathways can provide new fundamental understanding of the biophysical processes associated with the dynamic interconversions between bent and extended states and can unveil new putative therapeutic targets. In this work, we apply nonlinear manifold learning to coarse-grained molecular dynamics simulations of bent, extended, and two intermediate states of αIIbβ3 integrin to learn a low-dimensional embedding of the configurational phase space. We then train deep generative models to learn an inverse mapping between the low-dimensional embedding and high-dimensional molecular space and use these models to interpolate the molecular configurations constituting the activation pathways between the experimentally characterized states. This work furnishes plausible predictions of integrin activation pathways and reports a generic and transferable multiscale technique to predict transition pathways for biomolecular systems.
Collapse
Affiliation(s)
- Siva Dasetty
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Tamara C Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois.
| |
Collapse
|
6
|
Dong L, Li L, Chen H, Cao Y, Lei H. Mechanochemistry: Fundamental Principles and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403949. [PMID: 39206931 DOI: 10.1002/advs.202403949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Mechanochemistry is an emerging research field at the interface of physics, mechanics, materials science, and chemistry. Complementary to traditional activation methods in chemistry, such as heat, electricity, and light, mechanochemistry focuses on the activation of chemical reactions by directly or indirectly applying mechanical forces. It has evolved as a powerful tool for controlling chemical reactions in solid state systems, sensing and responding to stresses in polymer materials, regulating interfacial adhesions, and stimulating biological processes. By combining theoretical approaches, simulations and experimental techniques, researchers have gained intricate insights into the mechanisms underlying mechanochemistry. In this review, the physical chemistry principles underpinning mechanochemistry are elucidated and a comprehensive overview of recent significant achievements in the discovery of mechanically responsive chemical processes is provided, with a particular emphasis on their applications in materials science. Additionally, The perspectives and insights into potential future directions for this exciting research field are offered.
Collapse
Affiliation(s)
- Liang Dong
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Luofei Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Huiyan Chen
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
- Institute of Advanced Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| |
Collapse
|
7
|
Din M, Paul S, Ullah S, Yang H, Xu RG, Abidin NAZ, Sun A, Chen YC, Gao R, Chowdhury B, Zhou F, Rogers S, Miller M, Biswas A, Hu L, Fan Z, Zahner C, Fan J, Chen Z, Berman M, Xue L, Ju LA, Chen Y. Multi-parametric thrombus profiling microfluidics detects intensified biomechanical thrombogenesis associated with hypertension and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598290. [PMID: 38915705 PMCID: PMC11195082 DOI: 10.1101/2024.06.11.598290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Arterial thrombosis, which represents a critical complication of cardiovascular diseases, is a leading cause of death and disability worldwide with no effective bioassay for clinical prediction. As a symbolic feature of arterial thrombosis, severe stenosis in the blood vessel creates a high-shear, high-gradient flow environment that effectively facilitates platelet aggregation towards vessel occlusion even with platelet amplification loops inhibited. However, no approach is currently available to comprehensively characterize the size, composition and platelet activation status of thrombi forming under this biorheological condition. Here, we present a thrombus profiling assay that monitors the multi-dimensional attributes of thrombi forming in conditions mimicking the physiological scenario of arterial thrombosis. Using this platform, we demonstrate that different receptor-ligand interactions contribute distinctively to the composition and activation status of the thrombus. Our investigation into hypertensive and older individuals reveals intensified biomechanical thrombogenesis and multi-dimensional thrombus profile abnormalities, demonstrating a direct contribution of mechanobiology to arterial thrombosis and endorsing the diagnostic potential of the assay. Furthermore, we identify the hyperactivity of GPIbα-integrin αIIbβ3 mechanosensing axis as a molecular mechanism that contributes to hypertension-associated arterial thrombosis. By studying the interactions between anti-thrombotic inhibitors and hypertension, and the inter-individual variability in personal thrombus profiles, our work reveals a critical need for personalized anti-thrombotic drug selection that accommodates each patient's pathological profile.
Collapse
Affiliation(s)
- Misbahud Din
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Souvik Paul
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Sana Ullah
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Haoyi Yang
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Rong-Guang Xu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Division of Thoracic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | - Allan Sun
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| | - Yiyao Catherine Chen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Rui Gao
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Bari Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Stephenie Rogers
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Mariel Miller
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Atreyee Biswas
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut 06030, USA
| | - Christopher Zahner
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Jing Fan
- Department of Mechanical Engineering, The City University of New York - City College, New York, New York 10031, USA
| | - Zi Chen
- Division of Thoracic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Megan Berman
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| |
Collapse
|
8
|
Goh T, Gao L, Singh J, Totaro R, Carey R, Yang K, Cartwright B, Dennis M, Ju LA, Waterhouse A. Platelet Adhesion and Activation in an ECMO Thrombosis-on-a-Chip Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401524. [PMID: 38757670 PMCID: PMC11321669 DOI: 10.1002/advs.202401524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/03/2024] [Indexed: 05/18/2024]
Abstract
Use of extracorporeal membrane oxygenation (ECMO) for cardiorespiratory failure remains complicated by blood clot formation (thrombosis), triggered by biomaterial surfaces and flow conditions. Thrombosis may result in ECMO circuit changes, cause red blood cell hemolysis, and thromboembolic events. Medical device thrombosis is potentiated by the interplay between biomaterial properties, hemodynamic flow conditions and patient pathology, however, the contribution and importance of these factors are poorly understood because many in vitro models lack the capability to customize material and flow conditions to investigate thrombosis under clinically relevant medical device conditions. Therefore, an ECMO thrombosis-on-a-chip model is developed that enables highly customizable biomaterial and flow combinations to evaluate ECMO thrombosis in real-time with low blood volume. It is observed that low flow rates, decelerating conditions, and flow stasis significantly increased platelet adhesion, correlating with clinical thrombus formation. For the first time, it is found that tubing material, polyvinyl chloride, caused increased platelet P-selectin activation compared to connector material, polycarbonate. This ECMO thrombosis-on-a-chip model can be used to guide ECMO operation, inform medical device design, investigate embolism, occlusion and platelet activation mechanisms, and develop anti-thrombotic biomaterials to ultimately reduce medical device thrombosis, anti-thrombotic drug use and therefore bleeding complications, leading to safer blood-contacting medical devices.
Collapse
Affiliation(s)
- Tiffany Goh
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Heart Research InstituteNewtownNSW2042Australia
- Charles Perkins CentreThe University of SydneySydneyNSW2006Australia
- The University of Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
| | - Lingzi Gao
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Heart Research InstituteNewtownNSW2042Australia
- Charles Perkins CentreThe University of SydneySydneyNSW2006Australia
- The University of Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
| | - Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Heart Research InstituteNewtownNSW2042Australia
- Charles Perkins CentreThe University of SydneySydneyNSW2006Australia
- The University of Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
| | - Richard Totaro
- Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Intensive Care DepartmentRoyal Prince Alfred HospitalMissenden Road, CamperdownSydneyNSW2050Australia
| | - Ruaidhri Carey
- Intensive Care DepartmentRoyal Prince Alfred HospitalMissenden Road, CamperdownSydneyNSW2050Australia
| | - Kevin Yang
- Intensive Care DepartmentRoyal Prince Alfred HospitalMissenden Road, CamperdownSydneyNSW2050Australia
| | - Bruce Cartwright
- Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Anaesthetics DepartmentRoyal Prince Alfred HospitalCamperdownSydneyNSW2050Australia
| | - Mark Dennis
- Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Cardiology DepartmentRoyal Prince Alfred HospitalMissenden Road, CamperdownSydneyNSW2050Australia
| | - Lining Arnold Ju
- Heart Research InstituteNewtownNSW2042Australia
- Charles Perkins CentreThe University of SydneySydneyNSW2006Australia
- The University of Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
- School of Biomedical EngineeringFaculty of EngineeringThe University of SydneyDarlingtonNSW2008Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSW2006Australia
- Charles Perkins CentreThe University of SydneySydneyNSW2006Australia
- The University of Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
| |
Collapse
|
9
|
Joshi O, Skóra T, Yarema A, Rabbitt RD, Bidone TC. Contributions of the individual domains of α IIbβ 3 integrin to its extension: Insights from multiscale modeling. Cytoskeleton (Hoboken) 2024; 81:393-408. [PMID: 38682753 PMCID: PMC11333186 DOI: 10.1002/cm.21865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/01/2024]
Abstract
The platelet integrin αIIbβ3 undergoes long-range conformational transitions between bent and extended conformations to regulate platelet aggregation during hemostasis and thrombosis. However, how exactly αIIbβ3 transitions between conformations remains largely elusive. Here, we studied how transitions across bent and extended-closed conformations of αIIbβ3 integrin are regulated by effective interactions between its functional domains. We first carried out μs-long equilibrium molecular dynamics (MD) simulations of full-length αIIbβ3 integrins in bent and intermediate conformations, the latter characterized by an extended headpiece and closed legs. Then, we built heterogeneous elastic network models, perturbed inter-domain interactions, and evaluated their relative contributions to the energy barriers between conformations. Results showed that integrin extension emerges from: (i) changes in interfaces between functional domains; (ii) allosteric coupling of the head and upper leg domains with flexible lower leg domains. Collectively, these results provide new insights into integrin conformational activation based on short- and long-range interactions between its functional domains and highlight the importance of the lower legs in the regulation of integrin allostery.
Collapse
Affiliation(s)
- Onkar Joshi
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, USA
| | - Tomasz Skóra
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, USA
| | - Anna Yarema
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Tamara C Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Ding S, Chen Y, Huang C, Song L, Liang Z, Wei B. Perception and response of skeleton to mechanical stress. Phys Life Rev 2024; 49:77-94. [PMID: 38564907 DOI: 10.1016/j.plrev.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Mechanical stress stands as a fundamental factor in the intricate processes governing the growth, development, morphological shaping, and maintenance of skeletal mass. The profound influence of stress in shaping the skeletal framework prompts the assertion that stress essentially births the skeleton. Despite this acknowledgment, the mechanisms by which the skeleton perceives and responds to mechanical stress remain enigmatic. In this comprehensive review, our scrutiny focuses on the structural composition and characteristics of sclerotin, leading us to posit that it serves as the primary structure within the skeleton responsible for bearing and perceiving mechanical stress. Furthermore, we propose that osteocytes within the sclerotin emerge as the principal mechanical-sensitive cells, finely attuned to perceive mechanical stress. And a detailed analysis was conducted on the possible transmission pathways of mechanical stress from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Sicheng Ding
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yiren Chen
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Chengshuo Huang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lijun Song
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhen Liang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Bo Wei
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
11
|
Liu J, Tan YY, Zheng W, Wang Y, Ju LA, Su QP. Nanoscale insights into hematology: super-resolved imaging on blood cell structure, function, and pathology. J Nanobiotechnology 2024; 22:363. [PMID: 38910248 PMCID: PMC11194919 DOI: 10.1186/s12951-024-02605-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
Fluorescence nanoscopy, also known as super-resolution microscopy, has transcended the conventional resolution barriers and enabled visualization of biological samples at nanometric resolutions. A series of super-resolution techniques have been developed and applied to investigate the molecular distribution, organization, and interactions in blood cells, as well as the underlying mechanisms of blood-cell-associated diseases. In this review, we provide an overview of various fluorescence nanoscopy technologies, outlining their current development stage and the challenges they are facing in terms of functionality and practicality. We specifically explore how these innovations have propelled forward the analysis of thrombocytes (platelets), erythrocytes (red blood cells) and leukocytes (white blood cells), shedding light on the nanoscale arrangement of subcellular components and molecular interactions. We spotlight novel biomarkers uncovered by fluorescence nanoscopy for disease diagnosis, such as thrombocytopathies, malignancies, and infectious diseases. Furthermore, we discuss the technological hurdles and chart out prospective avenues for future research directions. This review aims to underscore the significant contributions of fluorescence nanoscopy to the field of blood cell analysis and disease diagnosis, poised to revolutionize our approach to exploring, understanding, and managing disease at the molecular level.
Collapse
Affiliation(s)
- Jinghan Liu
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yuping Yolanda Tan
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
| | - Wen Zheng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
| | - Qian Peter Su
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
- Heart Research Institute, Newtown, NSW, 2042, Australia.
| |
Collapse
|
12
|
Huang X, Meng L, Cao G, Prominski A, Hu Y, Yang C, Chen M, Shi J, Gallagher C, Cao T, Yue J, Huang J, Tian B. Multimodal probing of T-cell recognition with hexapod heterostructures. Nat Methods 2024; 21:857-867. [PMID: 38374262 DOI: 10.1038/s41592-023-02165-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/26/2023] [Indexed: 02/21/2024]
Abstract
Studies using antigen-presenting systems at the single-cell and ensemble levels can provide complementary insights into T-cell signaling and activation. Although crucial for advancing basic immunology and immunotherapy, there is a notable absence of synthetic material toolkits that examine T cells at both levels, and especially those capable of single-molecule-level manipulation. Here we devise a biomimetic antigen-presenting system (bAPS) for single-cell stimulation and ensemble modulation of T-cell recognition. Our bAPS uses hexapod heterostructures composed of a submicrometer cubic hematite core (α-Fe2O3) and nanostructured silica branches with diverse surface modifications. At single-molecule resolution, we show T-cell activation by a single agonist peptide-loaded major histocompatibility complex; distinct T-cell receptor (TCR) responses to structurally similar peptides that differ by only one amino acid; and the superior antigen recognition sensitivity of TCRs compared with that of chimeric antigen receptors (CARs). We also demonstrate how the magnetic field-induced rotation of hexapods amplifies the immune responses in suspended T and CAR-T cells. In addition, we establish our bAPS as a precise and scalable method for identifying stimulatory antigen-specific TCRs at the single-cell level. Thus, our multimodal bAPS represents a unique biointerface tool for investigating T-cell recognition, signaling and function.
Collapse
Affiliation(s)
- Xiaodan Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Lingyuan Meng
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | - Yifei Hu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Chuanwang Yang
- The James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Min Chen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Jiuyun Shi
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | | | - Thao Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Jiping Yue
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| | - Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL, USA.
- The James Franck Institute, University of Chicago, Chicago, IL, USA.
- The Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Tuna R, Yi W, Crespo Cruz E, Romero JP, Ren Y, Guan J, Li Y, Deng Y, Bluestein D, Liu ZL, Sheriff J. Platelet Biorheology and Mechanobiology in Thrombosis and Hemostasis: Perspectives from Multiscale Computation. Int J Mol Sci 2024; 25:4800. [PMID: 38732019 PMCID: PMC11083691 DOI: 10.3390/ijms25094800] [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: 02/11/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Thrombosis is the pathological clot formation under abnormal hemodynamic conditions, which can result in vascular obstruction, causing ischemic strokes and myocardial infarction. Thrombus growth under moderate to low shear (<1000 s-1) relies on platelet activation and coagulation. Thrombosis at elevated high shear rates (>10,000 s-1) is predominantly driven by unactivated platelet binding and aggregating mediated by von Willebrand factor (VWF), while platelet activation and coagulation are secondary in supporting and reinforcing the thrombus. Given the molecular and cellular level information it can access, multiscale computational modeling informed by biology can provide new pathophysiological mechanisms that are otherwise not accessible experimentally, holding promise for novel first-principle-based therapeutics. In this review, we summarize the key aspects of platelet biorheology and mechanobiology, focusing on the molecular and cellular scale events and how they build up to thrombosis through platelet adhesion and aggregation in the presence or absence of platelet activation. In particular, we highlight recent advancements in multiscale modeling of platelet biorheology and mechanobiology and how they can lead to the better prediction and quantification of thrombus formation, exemplifying the exciting paradigm of digital medicine.
Collapse
Affiliation(s)
- Rukiye Tuna
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Wenjuan Yi
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Esmeralda Crespo Cruz
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - JP Romero
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Yi Ren
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Jingjiao Guan
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Yan Li
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Yuefan Deng
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Zixiang Leonardo Liu
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA;
| |
Collapse
|
14
|
Wang J, Jin W, Huang S, Wang W, Wang S, Yu Z, Gao L, Gao Y, Han H, Wang L. Microbubble Biointerfacing by Regulation of the Platelet Membrane Surfactant Activity at the Gas-Liquid Interface for Acute Thrombosis Targeting. Angew Chem Int Ed Engl 2024; 63:e202314583. [PMID: 38196289 DOI: 10.1002/anie.202314583] [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: 09/28/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 01/11/2024]
Abstract
Biointerfacing nanomaterials with cell membranes has been successful in the functionalization of nanoparticles or nanovesicles, but microbubble functionalization remains challenging due to the unique conformation of the lipid monolayer structure at the gas-liquid interface that provides insufficient surfactant activity. Here, we describe a strategy to rationally regulate the surfactant activity of platelet membrane vesicles by adjusting the ratio of proteins to lipids through fusion with synthetic phospholipids (i.e., liposomes). A "platesome" with the optimized protein-to-lipid ratio can be assembled at the gas-liquid interface in the same manner as pulmonary surfactants to stabilize a microsized gas bubble. Platesome microbubbles (PMBs) inherited 61.4 % of the platelet membrane vesicle proteins and maintained the active conformation of integrin αIIbβ3 without the talin 1 for fibrin binding. We demonstrated that the PMBs had good stability, long circulation, and superior functionality both in vitro and in vivo. Moreover, by molecular ultrasound imaging, the PMBs provide up to 11.8 dB of ultrasound signal-to-noise ratio enhancement for discriminating between acute and chronic thrombi. This surface tension regulating strategy may provide a paradigm for biointerfacing microbubbles with cell membranes, offering a potential new approach for the construction of molecular ultrasound contrast agents for the diagnosis of different diseases.
Collapse
Affiliation(s)
- Jiahui Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Weikui Jin
- Department of Ultrasound Diagnostics, The Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, P. R. China
| | - Shengyu Huang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Wenqi Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Siyu Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Zhen Yu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Li Gao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Yu Gao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Hao Han
- Department of Ultrasound Diagnostics, The Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, P. R. China
| | - Lianhui Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| |
Collapse
|
15
|
Bershadsky ES, Ermokhin DA, Kurattsev VA, Panteleev MA, Nechipurenko DY. Force balance ratio is a robust predictor of arterial thrombus stability. Biophys J 2024; 123:464-477. [PMID: 38204165 PMCID: PMC10912926 DOI: 10.1016/j.bpj.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/07/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
Thrombus formation on a damaged vessel wall can lead to the formation of a stable occlusive/subocclusive clot or unstable embolizing thrombus. Both outcomes can cause significant health damage. The mechanisms that regulate maximum thrombus size, its stability, and embolization in both micro- and macrocirculation are poorly understood. To investigate the impact of flow and intrathrombus forces on the stability of homogeneous and heterogeneous platelet thrombi in a wide range of thrombus geometries, critical interplatelet forces, vessel diameters, and hydrodynamic conditions, we took advantage of the recently developed in silico models. To perform analysis of thrombus stability/embolization in arterioles, we used our previously developed particle-based 2D model with a single-platelet resolution. Its results and predictions were further extended to a 3D case and the large spatial scales of arteries using novel particle-based and continuum 3D models. We found a robust quantitative parameter, termed force balance ratio, which quantifies the balance between destabilizing hydrodynamic and stabilizing interplatelet forces. This parameter predicts whether a homogeneous thrombus (or the shell of a heterogeneous thrombus) with a particular value of critical interplatelet forces will embolize under given hydrodynamic conditions. Our simulations also predict that, for a given magnitude of critical interplatelet forces, the longer thrombi are more stable than the shorter ones. Furthermore, the aggregates formed on top of the severe stenosis are more stable than thrombi formed at moderate stenosis. Taken together, our results give new insights into the interplay between critical interplatelet forces, local hydrodynamics, and overall thrombus stability against the flow.
Collapse
Affiliation(s)
- Efim S Bershadsky
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia
| | - Daniel A Ermokhin
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | | | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry Y Nechipurenko
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
16
|
Chen Y, Li Z, Kong F, Ju LA, Zhu C. Force-Regulated Spontaneous Conformational Changes of Integrins α 5β 1 and α Vβ 3. ACS NANO 2024; 18:299-313. [PMID: 38105535 PMCID: PMC10786158 DOI: 10.1021/acsnano.3c06253] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Integrins are cell surface nanosized receptors crucial for cell motility and mechanosensing of the extracellular environment, which are often targeted for the development of biomaterials and nanomedicines. As a key feature of integrins, their activity, structure and behavior are highly mechanosensitive, which are regulated by mechanical forces down to pico-Newton scale. Using single-molecule biomechanical approaches, we compared the force-modulated ectodomain bending/unbending conformational changes of two integrin species, α5β1 and αVβ3. It was found that the conformation of integrin α5β1 is determined by a threshold head-to-tail tension. By comparison, integrin αVβ3 exhibits bistability even without force and can spontaneously transition between the bent and extended conformations with an apparent transition time under a wide range of forces. Molecular dynamics simulations observed almost concurrent disruption of ∼2 hydrogen bonds during integrin α5β1 unbending, but consecutive disruption of ∼7 hydrogen bonds during integrin αVβ3 unbending. Accordingly, we constructed a canonical energy landscape for integrin α5β1 with a single energy well that traps the integrin in the bent state until sufficient force tilts the energy landscape to allow the conformational transition. In contrast, the energy landscape of integrin αVβ3 conformational changes was constructed with hexa-stable intermediate states and intermediate energy barriers that segregate the conformational change process into multiple small steps. Our study elucidates the different biomechanical inner workings of integrins α5β1 and αVβ3 at the submolecular level, helps understand their mechanosignaling processes and how their respective functions are facilitated by their distinctive mechanosensitivities, and provides useful design principles for the engineering of protein-based biomechanical nanomachines.
Collapse
Affiliation(s)
- Yunfeng Chen
- Woodruff School of Mechanical Engineering and Petit Institute
for Bioengineering
and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Department
of Biochemistry and Molecular Biology and Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Zhenhai Li
- Shanghai
Key Laboratory of Mechanics in Energy Engineering, Shanghai Institute
of Applied Mathematics and Mechanics, School of Mechanics and Engineering
Science, Shanghai University, Shanghai 200072, China
| | - Fang Kong
- Woodruff School of Mechanical Engineering and Petit Institute
for Bioengineering
and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
- School of
Biological Science, Nanyang Technological
University, Singapore 637551, Singapore
| | - Lining Arnold Ju
- Woodruff School of Mechanical Engineering and Petit Institute
for Bioengineering
and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
- School
of Biomedical Engineering, The University
of Sydney, Darlington, New South Wales 2008, Australia
- Charles
Perkins Centre, The University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Cheng Zhu
- Woodruff School of Mechanical Engineering and Petit Institute
for Bioengineering
and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
17
|
Duan Y, Szlam F, Hu Y, Chen W, Li R, Ke Y, Sniecinski R, Salaita K. Detection of cellular traction forces via the force-triggered Cas12a-mediated catalytic cleavage of a fluorogenic reporter strand. Nat Biomed Eng 2023; 7:1404-1418. [PMID: 37957275 PMCID: PMC11289779 DOI: 10.1038/s41551-023-01114-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 09/26/2023] [Indexed: 11/15/2023]
Abstract
Molecular forces generated by cell receptors are infrequent and transient, and hence difficult to detect. Here we report an assay that leverages the CRISPR-associated protein 12a (Cas12a) to amplify the detection of cellular traction forces generated by as few as 50 adherent cells. The assay involves the immobilization of a DNA duplex modified with a ligand specific for a cell receptor. Traction forces of tens of piconewtons trigger the dehybridization of the duplex, exposing a cryptic Cas12-activating strand that sets off the indiscriminate Cas12-mediated cleavage of a fluorogenic reporter strand. We used the assay to perform hundreds of force measurements using human platelets from a single blood draw to extract individualized dose-response curves and half-maximal inhibitory concentrations for a panel of antiplatelet drugs. For seven patients who had undergone cardiopulmonary bypass, platelet dysfunction strongly correlated with the need for platelet transfusion to limit bleeding. The Cas12a-mediated detection of cellular traction forces may be used to assess cell state, and to screen for genes, cell-adhesion ligands, drugs or metabolites that modulate cell mechanics.
Collapse
Affiliation(s)
- Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Fania Szlam
- Department of Anesthesiology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Wenchun Chen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Departments of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Departments of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Roman Sniecinski
- Department of Anesthesiology, School of Medicine, Emory University, Atlanta, GA, USA.
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA.
| |
Collapse
|
18
|
陶 人, 谢 旭, 吴 建, 方 颖. [Molecular dynamics simulation of force-regulated interaction between glycoprotein Ib α and filamin]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2023; 40:876-885. [PMID: 37879916 PMCID: PMC10600417 DOI: 10.7507/1001-5515.202302043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/06/2023] [Indexed: 10/27/2023]
Abstract
In resting platelets, the 17 th domain of filamin a (FLNa17) constitutively binds to the platelet membrane glycoprotein Ibα (GPIbα) at its cytoplasmic tail (GPIbα-CT) and inhibits the downstream signal activation, while the binding of ligand and blood shear force can activate platelets. To imitate the pull force transmitted from the extracellular ligand of GPIbα and the lateral tension from platelet cytoskeleton deformation, two pulling modes were applied on the GPIbα-CT/FLNa17 complex, and the molecular dynamics simulation method was used to explore the mechanical regulation on the affinity and mechanical stability of the complex. In this study, at first, nine pairs of key hydrogen bonds on the interface between GPIbα-CT and FLNa17 were identified, which was the basis for maintaining the complex structural stability. Secondly, it was found that these hydrogen bonding networks would be broken down and lead to the dissociation of FLNa17 from GPIbα-CT only under the axial pull force; but, under the lateral tension, the secondary structures at both terminals of FLNa17 would unfold to protect the interface of the GPIbα-CT/FLNa17 complex from mechanical damage. In the range of 0~40 pN, the increase of pull force promoted outward-rotation of the nitrogen atom of the 563 rd phenylalanine (PHE 563-N) at GPIbα-CT and the dissociation of the complex. This study for the first time revealed that the extracellular ligand-transmitted axial force could more effectively relieve the inhibition of FLNa17 on the downstream signal of GPIbα than pure mechanical tension at the atomic level, and would be useful for further understanding the platelet intracellular force-regulated signal pathway.
Collapse
Affiliation(s)
- 人才 陶
- 华南理工大学 生物科学与工程学院(广州 510006)School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - 旭斌 谢
- 华南理工大学 生物科学与工程学院(广州 510006)School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - 建华 吴
- 华南理工大学 生物科学与工程学院(广州 510006)School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - 颖 方
- 华南理工大学 生物科学与工程学院(广州 510006)School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
19
|
Belyaev AV, Fedotova IV. Molecular mechanisms of catch bonds and their implications for platelet hemostasis. Biophys Rev 2023; 15:1233-1256. [PMID: 37974999 PMCID: PMC10643804 DOI: 10.1007/s12551-023-01144-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 11/19/2023] Open
Abstract
Adhesive molecular bonds between blood cells are essential for thrombosis and hemostasis as they provide means for platelet adhesion, aggregation, and signaling in flowing blood. According to the nowadays conventional definition, a "catch" bond is a type of non-covalent bio-molecular bridge, whose dissociation lifetime counter-intuitively increases with applied tensile force. Following recent experimental findings, such receptor-ligand protein bonds are vital to the blood cells involved in the prevention of bleeding (hemostatic response) and infection (immunity). In this review, we examine the up-to-date experimental discoveries and theoretical insights about catch bonds between the blood cells, their biomechanical principles at the molecular level, and their role in platelet thrombosis and hemostasis.
Collapse
Affiliation(s)
- Aleksey V. Belyaev
- Faculty of Physics, M.V.Lomonosov Moscow State University, 1, Leninskiye Gory, build.2, Moscow, 119991 Russia
| | - Irina V. Fedotova
- Faculty of Physics, M.V.Lomonosov Moscow State University, 1, Leninskiye Gory, build.2, Moscow, 119991 Russia
| |
Collapse
|
20
|
Oshinowo O, Azer SS, Lin J, Lam WA. Why platelet mechanotransduction matters for hemostasis and thrombosis. J Thromb Haemost 2023; 21:2339-2353. [PMID: 37331517 PMCID: PMC10529432 DOI: 10.1016/j.jtha.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023]
Abstract
Mechanotransduction is the ability of cells to "feel" or sense their mechanical microenvironment and integrate and convert these physical stimuli into adaptive biochemical cellular responses. This phenomenon is vital for the physiology of numerous nucleated cell types to affect their various cellular processes. As the main drivers of hemostasis and clot retraction, platelets also possess this ability to sense the dynamic mechanical microenvironments of circulation and convert those signals into biological responses integral to clot formation. Like other cell types, platelets leverage their "hands" or receptors/integrins to mechanotransduce important signals in responding to vascular injury to achieve hemostasis. The clinical relevance of cellular mechanics and mechanotransduction is imperative as pathologic alterations or aberrant mechanotransduction in platelets has been shown to lead to bleeding and thrombosis. As such, the aim of this review is to provide an overview of the most recent research related to platelet mechanotransduction, from platelet generation to platelet activation, within the hemodynamic environment and clot contraction at the site of vascular injury, thereby covering the entire "life cycle" of platelets. Additionally, we describe the key mechanoreceptors in platelets and discuss the new biophysical techniques that have enabled the field to understand how platelets sense and respond to their mechanical microenvironment via those receptors. Finally, the clinical significance and importance of continued exploration of platelet mechanotransduction have been discussed as the key to better understanding of both thrombotic and bleeding disorders lies in a more complete mechanistic understanding of platelet function by way of mechanotransduction.
Collapse
Affiliation(s)
- Oluwamayokun Oshinowo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA
| | - Sally S Azer
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA
| | - Jessica Lin
- The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Wilbur A Lam
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA.
| |
Collapse
|
21
|
Chen L, Yu L, Liu Y, Xu H, Li W, Wang F, Zhu J, Yi K, Ma L, Xiao H, Zhou F, Chen M, Cheng Y, Wang F, Zhu C, Xiao X, Yang Y. Valve-Adjustable Optofluidic Bio-Imaging Platform for Progressive Stenosis Investigation. ACS Sens 2023; 8:3104-3115. [PMID: 37477650 DOI: 10.1021/acssensors.3c00754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The clinical evidence has proven that valvular stenosis is closely related to many vascular diseases, which attracts great academic attention to the corresponding pathological mechanisms. The investigation is expected to benefit from the further development of an in vitro model that is tunable for bio-mimicking progressive valvular stenosis and enables accurate optical recognition in complex blood flow. Here, we develop a valve-adjustable optofluidic bio-imaging recognition platform to fulfill it. Specifically, the bionic valve was designed with in situ soft membrane, and the internal air-pressure chamber could be regulated from the inside out to bio-mimic progressive valvular stenosis. The developed imaging algorithm enhances the recognition of optical details in blood flow imaging and allows for quantitative analysis. In a prospective clinical study, we examined the effect of progressive valvular stenosis on hemodynamics within the typical physiological range of veins by this way, where the inhomogeneity and local enhancement effect in the altered blood flow field were precisely described and the optical differences were quantified. The effectiveness and consistency of the results were further validated through statistical analysis. In addition, we tested it on fluorescence and noticed its good performance in fluorescent tracing of the clotting process. In virtue of theses merits, this system should be able to contribute to mechanism investigation, pharmaceutical development, and therapeutics of valvular stenosis-related diseases.
Collapse
Affiliation(s)
- Longfei Chen
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| | - Le Yu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Yantong Liu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Hongshan Xu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Wei Li
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Fang Wang
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Jiaomeng Zhu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
| | - Kezhen Yi
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Linlu Ma
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hui Xiao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Ming Chen
- Department of Blood Transfusion, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yanxiang Cheng
- School of Medicine, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Yi Yang
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital of Wuhan University, Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & technology, Wuhan University, Wuhan 430072, China
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| |
Collapse
|
22
|
Moldovan L, Song CH, Chen YC, Wang HJ, Ju LA. Biomembrane force probe (BFP): Design, advancements, and recent applications to live-cell mechanobiology. EXPLORATION (BEIJING, CHINA) 2023; 3:20230004. [PMID: 37933233 PMCID: PMC10624387 DOI: 10.1002/exp.20230004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/18/2023] [Indexed: 11/08/2023]
Abstract
Mechanical forces play a vital role in biological processes at molecular and cellular levels, significantly impacting various diseases such as cancer, cardiovascular disease, and COVID-19. Recent advancements in dynamic force spectroscopy (DFS) techniques have enabled the application and measurement of forces and displacements with high resolutions, providing crucial insights into the mechanical pathways underlying these diseases. Among DFS techniques, the biomembrane force probe (BFP) stands out for its ability to measure bond kinetics and cellular mechanosensing with pico-newton and nano-meter resolutions. Here, a comprehensive overview of the classical BFP-DFS setup is presented and key advancements are emphasized, including the development of dual biomembrane force probe (dBFP) and fluorescence biomembrane force probe (fBFP). BFP-DFS allows us to investigate dynamic bond behaviors on living cells and significantly enhances the understanding of specific ligand-receptor axes mediated cell mechanosensing. The contributions of BFP-DFS to the fields of cancer biology, thrombosis, and inflammation are delved into, exploring its potential to elucidate novel therapeutic discoveries. Furthermore, future BFP upgrades aimed at improving output and feasibility are anticipated, emphasizing its growing importance in the field of cell mechanobiology. Although BFP-DFS remains a niche research modality, its impact on the expanding field of cell mechanobiology is immense.
Collapse
Affiliation(s)
- Laura Moldovan
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
| | - Caroline Haoran Song
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| | - Yiyao Catherine Chen
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
| | - Haoqing Jerry Wang
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| | - Lining Arnold Ju
- School of Biomedical EngineeringThe University of SydneyDarlingtonNew South WalesAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNew South WalesAustralia
- Heart Research InstituteNewtownNew South WalesAustralia
- Sydney Nano Institute (Sydney Nano)The University of SydneyCamperdownNew South WalesAustralia
| |
Collapse
|
23
|
Choi HK, Travaglino S, Münchhalfen M, Görg R, Zhong Z, Lyu J, Reyes-Aguilar DM, Wienands J, Singh A, Zhu C. Mechanotransduction governs CD40 function and underlies X-linked Hyper IgM syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550231. [PMID: 37546834 PMCID: PMC10401940 DOI: 10.1101/2023.07.23.550231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
B cell maturation in germinal centers (GCs) depends on cognate interactions between the T and B cells. Upon interaction with CD40 ligand (CD40L) on T cells, CD40 delivers co-stimulatory signals alongside B cell antigen receptor (BCR) signaling to regulate affinity maturation and antibody class-switch during GC reaction. Mutations in CD40L disrupt interactions with CD40, which lead to abnormal antibody responses in immune deficiencies known as X-linked Hyper IgM syndrome (X-HIgM). Assuming that physical interactions between highly mobile T and B cells generate mechanical forces on CD40-CD40L bonds, we set out to study the B cell mechanobiology mediated by CD40-CD40L interaction. Using a suite of biophysical assays we find that CD40 forms catch bond with CD40L where the bond lasts longer at larger forces, B cells exert tension on CD40-CD40L bonds, and force enhances CD40 signaling and antibody class-switch. Significantly, X-HIgM CD40L mutations impair catch bond formation, suppress endogenous tension, and reduce force-enhanced CD40 signaling, leading to deficiencies in antibody class switch. Our findings highlight the critical role of mechanotransduction in CD40 function and provide insights into the molecular mechanisms underlying X-HIgM syndrome.
Collapse
|
24
|
Choi HK, Cong P, Ge C, Natarajan A, Liu B, Zhang Y, Li K, Rushdi MN, Chen W, Lou J, Krogsgaard M, Zhu C. Catch bond models may explain how force amplifies TCR signaling and antigen discrimination. Nat Commun 2023; 14:2616. [PMID: 37147290 PMCID: PMC10163261 DOI: 10.1038/s41467-023-38267-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 04/24/2023] [Indexed: 05/07/2023] Open
Abstract
The TCR integrates forces in its triggering process upon interaction with pMHC. Force elicits TCR catch-slip bonds with strong pMHCs but slip-only bonds with weak pMHCs. We develop two models and apply them to analyze 55 datasets, demonstrating the models' ability to quantitatively integrate and classify a broad range of bond behaviors and biological activities. Comparing to a generic two-state model, our models can distinguish class I from class II MHCs and correlate their structural parameters with the TCR/pMHC's potency to trigger T cell activation. The models are tested by mutagenesis using an MHC and a TCR mutated to alter conformation changes. The extensive comparisons between theory and experiment provide model validation and testable hypothesis regarding specific conformational changes that control bond profiles, thereby suggesting structural mechanisms for the inner workings of the TCR mechanosensing machinery and plausible explanations of why and how force may amplify TCR signaling and antigen discrimination.
Collapse
Affiliation(s)
- Hyun-Kyu Choi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Peiwen Cong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Chenghao Ge
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA, 91320, USA
| | - Aswin Natarajan
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Baoyu Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Yong Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaitao Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Muaz Nik Rushdi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Medtronic CO., Minneapolis, MN, 55432, USA
| | - Wei Chen
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jizhong Lou
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
25
|
Aggarwal A, Jennings CL, Manning E, Cameron SJ. Platelets at the Vessel Wall in Non-Thrombotic Disease. Circ Res 2023; 132:775-790. [PMID: 36927182 PMCID: PMC10027394 DOI: 10.1161/circresaha.122.321566] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
Platelets are small, anucleate entities that bud from megakaryocytes in the bone marrow. Among circulating cells, platelets are the most abundant cell, traditionally involved in regulating the balance between thrombosis (the terminal event of platelet activation) and hemostasis (a protective response to tissue injury). Although platelets lack the precise cellular control offered by nucleate cells, they are in fact very dynamic cells, enriched in preformed RNA that allows them the capability of de novo protein synthesis which alters the platelet phenotype and responses in physiological and pathological events. Antiplatelet medications have significantly reduced the morbidity and mortality for patients afflicted with thrombotic diseases, including stroke and myocardial infarction. However, it has become apparent in the last few years that platelets play a critical role beyond thrombosis and hemostasis. For example, platelet-derived proteins by constitutive and regulated exocytosis can be found in the plasma and may educate distant tissue including blood vessels. First, platelets are enriched in inflammatory and anti-inflammatory molecules that may regulate vascular remodeling. Second, platelet-derived microparticles released into the circulation can be acquired by vascular endothelial cells through the process of endocytosis. Third, platelets are highly enriched in mitochondria that may contribute to the local reactive oxygen species pool and remodel phospholipids in the plasma membrane of blood vessels. Lastly, platelets are enriched in proteins and phosphoproteins which can be secreted independent of stimulation by surface receptor agonists in conditions of disturbed blood flow. This so-called biomechanical platelet activation occurs in regions of pathologically narrowed (atherosclerotic) or dilated (aneurysmal) vessels. Emerging evidence suggests platelets may regulate the process of angiogenesis and blood flow to tumors as well as education of distant organs for the purposes of allograft health following transplantation. This review will illustrate the potential of platelets to remodel blood vessels in various diseases with a focus on the aforementioned mechanisms.
Collapse
Affiliation(s)
- Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
| | - Courtney L. Jennings
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
| | - Emily Manning
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Scott J. Cameron
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
- Heart Vascular and Thoracic Institute, Department of Cardiovascular Medicine, Section of Vascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Hematology, Taussig Cancer Center, Cleveland, Ohio
| |
Collapse
|
26
|
Rosado AM, Zhang Y, Choi HK, Chen Y, Ehrlich SM, Jin F, Grakoui A, Evavold BD, Zhu C. Memory in repetitive protein–protein interaction series. APL Bioeng 2023. [DOI: 10.1063/5.0130805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Interactions between proteins coordinate biological processes in an organism and may impact its responses to changing environments and diseases through feedback systems. Feedback systems function by using changes in the past to influence behaviors in the future, which we refer to here as memory. Here, we summarized several observations made, ideas conceptualized, and mathematical models developed for quantitatively analyzing memory effects in repetitive protein–protein interactions (PPIs). Specifically, we consider how proteins on the cell or in isolation retain information about prior interactions to impact current interactions. The micropipette, biomembrane force probe, and atomic force microscopic techniques were used to repeatedly assay PPIs. The resulting time series were analyzed by a previous and two new models to extract three memory indices of short (seconds), intermediate (minutes), and long (hours) timescales. We found that interactions of cell membrane, but not soluble, T cell receptor (TCR) with peptide-major histocompatibility complex (pMHC) exhibits short-term memory that impacts on-rate, but not off-rate of the binding kinetics. Peptide dissociation from MHC resulted in intermediate- and long-term memories in TCR–pMHC interactions. However, we observed no changes in kinetic parameters by repetitive measurements on living cells over intermediate timescales using stable pMHCs. Parameters quantifying memory effects in PPIs could provide additional information regarding biological mechanisms. The methods developed herein also provide tools for future research.
Collapse
Affiliation(s)
- Aaron M. Rosado
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Yan Zhang
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Hyun-Kyu Choi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Yunfeng Chen
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Samuel M. Ehrlich
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Fengzhi Jin
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Arash Grakoui
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Brian D. Evavold
- Department of Immunology and Microbiology, Emory University School of Medicine, Atlanta, Georgia 30332 USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
27
|
Tong D, Soley N, Kolasangiani R, Schwartz MA, Bidone TC. Integrin α IIbβ 3 intermediates: From molecular dynamics to adhesion assembly. Biophys J 2023; 122:533-543. [PMID: 36566352 PMCID: PMC9941721 DOI: 10.1016/j.bpj.2022.12.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/14/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
The platelet integrin αIIbβ3 undergoes long-range conformational transitions associated with its functional conversion from inactive (low-affinity) to active (high-affinity) during hemostasis. Although new conformations that are intermediate between the well-characterized bent and extended states have been identified, their molecular dynamic properties and functions in the assembly of adhesions remain largely unexplored. In this study, we evaluated the properties of intermediate conformations of integrin αIIbβ3 and characterized their effects on the assembly of adhesions by combining all-atom simulations, principal component analysis, and mesoscale modeling. Our results show that in the low-affinity, bent conformation, the integrin ectodomain tends to pivot around the legs; in intermediate conformations, the headpiece becomes partially extended, away from the lower legs. In the fully open, active state, αIIbβ3 is flexible, and the motions between headpiece and lower legs are accompanied by fluctuations of the transmembrane helices. At the mesoscale, bent integrins form only unstable adhesions, but intermediate or open conformations stabilize the adhesions. These studies reveal a mechanism by which small variations in ligand binding affinity and enhancement of the ligand-bound lifetime in the presence of actin retrograde flow stabilize αIIbβ3 integrin adhesions.
Collapse
Affiliation(s)
- Dudu Tong
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Nidhi Soley
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Reza Kolasangiani
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut; Department of Cell Biology, Yale University, New Haven, Connecticut; Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, Connecticut
| | - Tamara C Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah; Department of Biochemistry, University of Utah, Salt Lake City, Utah; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
28
|
Qin R, An C, Chen W. Physical-Chemical Regulation of Membrane Receptors Dynamics in Viral Invasion and Immune Defense. J Mol Biol 2023; 435:167800. [PMID: 36007627 PMCID: PMC9394170 DOI: 10.1016/j.jmb.2022.167800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 02/04/2023]
Abstract
Mechanical cues dynamically regulate membrane receptors functions to trigger various physiological and pathological processes from viral invasion to immune defense. These cues mainly include various types of dynamic mechanical forces and the spatial confinement of plasma membrane. However, the molecular mechanisms of how they couple with biochemical cues in regulating membrane receptors functions still remain mysterious. Here, we review recent advances in methodologies of single-molecule biomechanical techniques and in novel biomechanical regulatory mechanisms of critical ligand recognition of viral and immune receptors including SARS-CoV-2 spike protein, T cell receptor (TCR) and other co-stimulatory immune receptors. Furthermore, we provide our perspectives of the general principle of how force-dependent kinetics determine the dynamic functions of membrane receptors and of biomechanical-mechanism-driven SARS-CoV-2 neutralizing antibody design and TCR engineering for T-cell-based therapies.
Collapse
Affiliation(s)
- Rui Qin
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chenyi An
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory for Modern Optical Instrumentation Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
29
|
Belyaev AV, Kushchenko YK. Biomechanical activation of blood platelets via adhesion to von Willebrand factor studied with mesoscopic simulations. Biomech Model Mechanobiol 2023; 22:785-808. [PMID: 36627458 PMCID: PMC9838538 DOI: 10.1007/s10237-022-01681-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023]
Abstract
Platelet adhesion and activation are essential initial processes of arterial and microvascular hemostasis, where high hydrodynamic forces from the bloodflow impede coagulation. The process relies on von Willebrand factor (VWF)-a linear multimeric protein of blood plasma plays a pivotal role in mechanochemical regulation of shear-induced platelet aggregation (SIPA). Adhesive interactions between VWF and glycoprotein receptors GPIb are crucial for platelet recruitment under high shear stress in fluid. Recent advances in experimental studies revealed that mechanical tension on the extracellular part of GPIb may trigger a cascade of biochemical reactions in platelets leading to activation of integrins [Formula: see text] (also known as GPIIb/IIIa) and strengthening of the adhesion. The present paper is aimed at investigation of this process by three-dimensional computer simulations of platelet adhesion to surface-grafted VWF multimers in pressure-driven flow of platelet-rich plasma. The simulations demonstrate that GPIb-mediated mechanotransduction is a feasible way of platelet activation and stabilization of platelet aggregates under high shear stress. Quantitative understanding of mechanochemical processes involved in SIPA would potentially promote the discovery of new anti-platelet medication and the development of multiscale numerical models of platelet thrombosis and hemostasis.
Collapse
Affiliation(s)
- Aleksey V. Belyaev
- grid.14476.300000 0001 2342 9668Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskiye Gory, Moscow, Russia 119991
| | - Yulia K. Kushchenko
- grid.14476.300000 0001 2342 9668Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskiye Gory, Moscow, Russia 119991
| |
Collapse
|
30
|
Chen Y, Kong F, Li Z, Ju LA, Zhu C. Force-regulated spontaneous conformational changes of integrins α 5 β 1 and α V β 3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.523308. [PMID: 36712101 PMCID: PMC9881988 DOI: 10.1101/2023.01.09.523308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Force can modulate the properties and functions of macromolecules by inducing conformational changes, such as coiling/uncoiling, zipping/unzipping, and folding/unfolding. Here we compared force-modulated bending/unbending of two purified integrin ectodomains, α 5 β 1 and α V β 3 , using single-molecule approaches. Similar to previously characterized mechano-sensitive macromolecules, the conformation of α 5 β 1 is determined by a threshold head-to-tail tension, suggesting a canonical energy landscape with a deep energy well that traps the integrin in the bent state until sufficient force tilts the energy landscape to accelerate transition to the extended state. By comparison, α V β 3 exhibits bi-stability even without force and can spontaneously transition between the bent and extended conformations in a wide range of forces without energy supplies. Molecular dynamics simulations revealed consecutive formation and disruption of 7 hydrogen bonds during α V β 3 bending and unbending, respectively. Accordingly, we constructed an energy landscape with hexa-stable intermediate states to break down the energy barrier separating the bent and extended states into smaller ones, making it possible for the thermal agitation energy to overcome them sequentially and to be accumulated and converted into mechanical work required for α V β 3 to bend against force. Our study elucidates the different inner workings of α 5 β 1 and α V β 3 at the sub-molecular level, sheds lights on how their respectively functions are facilitated by their distinctive mechano-sensitivities, helps understand their signal initiation processes, and provides critical concepts and useful design principles for engineering of protein-based biomechanical nanomachines.
Collapse
|
31
|
Wang Y, Jin J, Wang HJ, Ju LA. Acoustic Force-Based Cell-Matrix Avidity Measurement in High Throughput. BIOSENSORS 2023; 13:95. [PMID: 36671930 PMCID: PMC9855465 DOI: 10.3390/bios13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Cancer cells interacting with the extracellular matrix (ECM) in the tumor microenvironment is pivotal for tumorigenesis, invasion, and metastasis. Cell-ECM adhesion has been intensively studied in cancer biology in the past decades to understand the molecular mechanisms underlying the adhesion events and extracellular mechanosensing, as well as develop therapeutic strategies targeting the cell adhesion molecules. Many methods have been established to measure the cell-ECM adhesion strength and correlate it with the metastatic potential of certain cancer types. However, those approaches are either low throughput, not quantitative, or with poor sensitivity and reproducibility. Herein, we developed a novel acoustic force spectroscopy based method to quantify the cell-ECM adhesion strength during adhesion maturation process using the emerging z-Movi® technology. This can be served as a fast, simple, and high-throughput platform for functional assessment of cell adhesion molecules in a highly predictive and reproducible manner.
Collapse
Affiliation(s)
- Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Jasmine Jin
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Haoqing Jerry Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
32
|
Integrin Conformational Dynamics and Mechanotransduction. Cells 2022; 11:cells11223584. [PMID: 36429013 PMCID: PMC9688440 DOI: 10.3390/cells11223584] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The function of the integrin family of receptors as central mediators of cell-extracellular matrix (ECM) and cell-cell adhesion requires a remarkable convergence of interactions and influences. Integrins must be anchored to the cytoskeleton and bound to extracellular ligands in order to provide firm adhesion, with force transmission across this linkage conferring tissue integrity. Integrin affinity to ligands is highly regulated by cell signaling pathways, altering affinity constants by 1000-fold or more, via a series of long-range conformational transitions. In this review, we first summarize basic, well-known features of integrin conformational states and then focus on new information concerning the impact of mechanical forces on these states and interstate transitions. We also discuss how these effects may impact mechansensitive cell functions and identify unanswered questions for future studies.
Collapse
|
33
|
Dupuy A, Ju LA, Chiu J, Passam FH. Mechano-Redox Control of Integrins in Thromboinflammation. Antioxid Redox Signal 2022; 37:1072-1093. [PMID: 35044225 DOI: 10.1089/ars.2021.0265] [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] [Indexed: 11/12/2022]
Abstract
Significance: How mechanical forces and biochemical cues are coupled remains a miracle for many biological processes. Integrins, well-known adhesion receptors, sense changes in mechanical forces and reduction-oxidation reactions (redox) in their environment to mediate their adhesive function. The coupling of mechanical and redox function is a new area of investigation. Disturbance of normal mechanical forces and the redox balance occurs in thromboinflammatory conditions; atherosclerotic plaques create changes to the mechanical forces in the circulation. Diabetes induces redox changes in the circulation by the production of reactive oxygen species and vascular inflammation. Recent Advances: Integrins sense changes in the blood flow shear stress at the level of focal adhesions and respond to flow and traction forces by increased signaling. Talin, the integrin-actin linker, is a traction force sensor and adaptor. Oxidation and reduction of integrin disulfide bonds regulate their adhesion. A conserved disulfide bond in integrin αlpha IIb beta 3 (αIIbβ3) is directly reduced by the thiol oxidoreductase endoplasmic reticulum protein 5 (ERp5) under shear stress. Critical Issues: The coordination of mechano-redox events between the extracellular and intracellular compartments is an active area of investigation. Another fundamental issue is to determine the spatiotemporal arrangement of key regulators of integrins' mechanical and redox interactions. How thromboinflammatory conditions lead to mechanoredox uncoupling is relatively unexplored. Future Directions: Integrated approaches, involving disulfide bond biochemistry, microfluidic assays, and dynamic force spectroscopy, will aid in showing that cell adhesion constitutes a crossroad of mechano- and redox biology, within the same molecule, the integrin. Antioxid. Redox Signal. 37, 1072-1093.
Collapse
Affiliation(s)
- Alexander Dupuy
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, Australia.,Heart Research Institute, Newtown, Australia
| | - Lining Arnold Ju
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia.,Heart Research Institute, Newtown, Australia.,School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, Australia
| | - Joyce Chiu
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia.,ACRF Centenary Cancer Research Centre, The Centenary Institute, Camperdown, Australia
| | - Freda H Passam
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, Australia.,Heart Research Institute, Newtown, Australia
| |
Collapse
|
34
|
Li L, Hu J, Różycki B, Ji J, Song F. Interplay of receptor-ligand binding and lipid domain formation during cell adhesion. Front Mol Biosci 2022; 9:1019477. [PMID: 36203878 PMCID: PMC9531914 DOI: 10.3389/fmolb.2022.1019477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022] Open
Abstract
Cell adhesion involved in biological processes such as cell migration, immune responses, and cancer metastasis, is mediated by the specific binding of receptor and ligand proteins. Some of these proteins exhibit affinity for nanoscale lipid clusters in cell membranes. A key question is how these nanoscale lipid clusters influence and react to the receptor-ligand binding during cell adhesion. In this article, we review recent computational studies that shed new light on the interplay of the receptor-ligand binding and the formation of lipid domains in adhering membranes. These studies indicate that the receptor-ligand binding promotes coalescence of lipid clusters into mesoscale domains, which, in turn, enhances both the affinity and cooperativity of the receptor-ligand binding in cell-cell adhesion with mobile ligands. In contrast, in the case of cell-extracellular matrix adhesion with immobile ligands, the receptor-ligand binding and the lipid cluster coalescence can be correlated or anti-correlated, depending strongly on the ligand distribution. These findings deepen our understanding of correlations between cell adhesion and membrane heterogeneities.
Collapse
Affiliation(s)
- Long Li
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Jinglei Hu
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Bartosz Różycki
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Jing Ji
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Fan Song
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
35
|
Zhang Y, Ehrlich SM, Zhu C, Du X. Signaling mechanisms of the platelet glycoprotein Ib-IX complex. Platelets 2022; 33:823-832. [PMID: 35615944 PMCID: PMC9378482 DOI: 10.1080/09537104.2022.2071852] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/03/2022] [Accepted: 04/23/2022] [Indexed: 12/14/2022]
Abstract
The glycoprotein Ib-IX (GPIb-IX) complex mediates initial platelet adhesion to von Willebrand factor (VWF) immobilized on subendothelial matrix and endothelial surfaces, and transmits VWF binding-induced signals to stimulate platelet activation. GPIb-IX also functions as part of a mechanosensor to convert mechanical force received via VWF binding into intracellular signals, thereby greatly enhancing platelet activation. Thrombin binding to GPIb-IX initiates GPIb-IX signaling cooperatively with protease-activated receptors to synergistically stimulate the platelet response to low-dose thrombin. GPIb-IX signaling may also occur following the binding of other GPIb-IX ligands such as leukocyte integrin αMβ2 and red cell-derived semaphorin 7A, contributing to thrombo-inflammation. GPIb-IX signaling requires the interaction between the cytoplasmic domains of GPIb-IX and 14-3-3 protein and is mediated through Src family kinases, the Rho family of small GTPases, phosphoinositide 3-kinase-Akt-cGMP-mitogen-activated protein kinase, and LIM kinase 1 signaling pathways, leading to calcium mobilization, integrin activation, and granule secretion. This review summarizes the current understanding of GPIb-IX signaling.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago,Chicago, Illinois, USA
| | - Samuel M Ehrlich
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Cheng Zhu
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Xiaoping Du
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago,Chicago, Illinois, USA
| |
Collapse
|
36
|
Guo S, Zhang S, Chen K, Chen X, Hu F. Effects of diagnostic ultrasound with cRGD-microbubbles on simultaneous detection and treatment of atherosclerotic plaque in ApoE−/− mice. Front Cardiovasc Med 2022; 9:946557. [PMID: 35935617 PMCID: PMC9354833 DOI: 10.3389/fcvm.2022.946557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Atherosclerotic vulnerable plaque is the leading cause of acute fatal cardiovascular events. Thus, early rapid identification and appropriate treatment of atherosclerotic plaque maybe can prevent fatal cardiovascular events. However, few non–invasive molecular imaging techniques are currently available for the simultaneous detection and targeted treatment of atherosclerotic plaques. We hypothesized that diagnostic ultrasound (DU) combined with cyclic Arg-Gly-Asp-modified microbubbles (MBR) could provide targeted imaging and dissolution of activated platelets to identify advanced atherosclerotic plaques and improve plaque instability. Methods Three mouse models, apolipoprotein E-deficient mice on a hypercholesterolemic diet (HCD) or normal chow diet and wild-type mice on an HCD were used. The most appropriate ultrasonic mechanical index (MI) was determined based on the expression of GP IIb/IIIa in sham, DU alone and DUMBR-treated groups at MI values of 0.5, 1.5, and 1.9. The video intensity (VI) values, activated platelets and plaque instability were analyzed by ultrasound molecular imaging, scanning electron microscopy and histopathological methods. Results We found that the VI values of ultrasound molecular imaging of MBR were positively correlated with plaque GP IIb/IIIa expression, vulnerability index and necrotic center / fiber cap ratio. 24 h after treatment at different MIs, compared with those of the other groups, both the VI values and GP IIb/IIIa expression were significantly reduced in MI 1.5 and MI 1.9 DUMBR-treated groups. The plaque vulnerability index and necrotic center / fiber cap ratio were significantly decreased in MI 1.5-treated group, which may be due to targeted dissolution of activated platelets, with a reduction in von Willebrand factor expression. Conclusion DUMBR targeting GP IIb/IIIa receptors could rapidly detect advanced atherosclerotic plaques and simultaneously give targeted therapy by dissolving activated and aggregated platelets. This technology may represent a novel approach for the simultaneous identification and treatment of atherosclerotic plaques.
Collapse
|
37
|
Xiao W, Chen W, Wang Y, Zhang C, Zhang X, Zhang S, Wu W. Recombinant DTβ4-inspired porous 3D vascular graft enhanced antithrombogenicity and recruited circulating CD93 +/CD34 + cells for endothelialization. SCIENCE ADVANCES 2022; 8:eabn1958. [PMID: 35857526 PMCID: PMC9278867 DOI: 10.1126/sciadv.abn1958] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/27/2022] [Indexed: 05/31/2023]
Abstract
Matching material degradation with host remodeling, including endothelialization and muscular remodeling, is important to vascular regeneration. We fabricated 3D PGS-PCL vascular grafts, which presented tunable polymer components, porosity, mechanical strength, and degrading rate. Furthermore, highly porous structures enabled 3D patterning of conjugated heparin-binding peptide, dimeric thymosin β4 (DTβ4), which played key roles in antiplatelets, fibrinogenesis inhibition, and recruiting circulating progenitor cells, thereafter contributed to high patency rate, and unprecedentedly acquired carotid arterial regeneration in rabbit model. Through single-cell RNA sequencing analysis and cell tracing studies, a subset of endothelial progenitor cells, myeloid-derived CD93+/CD34+ cells, was identified as the main contributor to final endothelium regeneration. To conclude, DTβ4-inspired porous 3DVGs present adjustable physical properties, superior anticoagulating, and re-endothelializing potentials, which leads to the regeneration of small-caliber artery, thus offering a promising tool for vessel replacement in clinical applications.
Collapse
Affiliation(s)
- Weiwei Xiao
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wanli Chen
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Yinggang Wang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Xinchi Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Siqian Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wei Wu
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
38
|
GPIIb/IIIa-GPVI-commanded platelet patrol. Blood 2022; 140:81-83. [PMID: 35834285 DOI: 10.1182/blood.2022016697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 01/16/2023] Open
|
39
|
An C, Wang X, Song F, Hu J, Li L. Insights into intercellular receptor-ligand binding kinetics in cell communication. Front Bioeng Biotechnol 2022; 10:953353. [PMID: 35837553 PMCID: PMC9273785 DOI: 10.3389/fbioe.2022.953353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/09/2022] [Indexed: 01/14/2023] Open
Abstract
Cell-cell communication is crucial for cells to sense, respond and adapt to environmental cues and stimuli. The intercellular communication process, which involves multiple length scales, is mediated by the specific binding of membrane-anchored receptors and ligands. Gaining insight into two-dimensional receptor-ligand binding kinetics is of great significance for understanding numerous physiological and pathological processes, and stimulating new strategies in drug design and discovery. To this end, extensive studies have been performed to illuminate the underlying mechanisms that control intercellular receptor-ligand binding kinetics via experiment, theoretical analysis and numerical simulation. It has been well established that the cellular microenvironment where the receptor-ligand interaction occurs plays a vital role. In this review, we focus on the advances regarding the regulatory effects of three factors including 1) protein-membrane interaction, 2) biomechanical force, and 3) bioelectric microenvironment to summarize the relevant experimental observations, underlying mechanisms, as well as their biomedical significances and applications. Meanwhile, we introduce modeling methods together with experiment technologies developed for dealing with issues at different scales. We also outline future directions to advance the field and highlight that building up systematic understandings for the coupling effects of these regulatory factors can greatly help pharmaceutical development.
Collapse
Affiliation(s)
- Chenyi An
- School of Biology and Engineering, Guizhou Medical University, Guiyang, China
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, China
| | - Fan Song
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jinglei Hu
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Coumarin Derivatives Inhibit ADP-Induced Platelet Activation and Aggregation. Molecules 2022; 27:molecules27134054. [PMID: 35807298 PMCID: PMC9268609 DOI: 10.3390/molecules27134054] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Coumarin was first discovered in Tonka bean and then widely in other plants. Coumarin has an anticoagulant effect, and its derivative, warfarin, is a vitamin K analogue that inhibits the synthesis of clotting factors and is more widely used in the clinical treatment of endovascular embolism. At present, many artificial chemical synthesis methods can be used to modify the structure of coumarin to develop many effective drugs with low toxicity. In this study, we investigated the effects of six coumarin derivatives on the platelet aggregation induced by adenosine diphosphate (ADP). We found that the six coumarin derivatives inhibited the active form of GPIIb/IIIa on platelets and hence inhibit platelet aggregation. We found that 7-hydroxy-3-phenyl 4H-chromen-4-one (7-hydroxyflavone) had the most severe effect. In addition, we further analyzed the downstream signal transduction of the ADP receptor, including the release of calcium ions and the regulation of cAMP, which were inhibited by the six coumarin derivatives selected in this study. These results suggest that coumarin derivatives inhibit coagulation by inhibiting the synthesis of coagulation factors and they may also inhibit platelet aggregation.
Collapse
|
41
|
The interface between biochemical signaling and cell mechanics shapes T lymphocyte migration and activation. Eur J Cell Biol 2022; 101:151236. [DOI: 10.1016/j.ejcb.2022.151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 11/18/2022] Open
|
42
|
Liu ZL, Bresette C, Aidun CK, Ku DN. SIPA in 10 milliseconds: VWF tentacles agglomerate and capture platelets under high shear. Blood Adv 2022; 6:2453-2465. [PMID: 34933342 PMCID: PMC9043924 DOI: 10.1182/bloodadvances.2021005692] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/27/2021] [Indexed: 11/22/2022] Open
Abstract
Shear-induced platelet aggregation (SIPA) occurs under elevated shear rates (10 000 s-1) found in stenotic coronary and carotid arteries. The pathologically high shear environment can lead to occlusive thrombosis by SIPA from the interaction of nonactivated platelets and von Willebrand factor (VWF) via glycoprotein Ib-A1 binding. This process under high shear rates is difficult to visualize experimentally with concurrent molecular- and cellular-resolutions. To understand this fast bonding, we employ a validated multiscale in silico model incorporating measured molecular kinetics and a thrombosis-on-a-chip device to delineate the flow-mediated biophysics of VWF and platelets assembly into mural microthrombi. We show that SIPA begins with VWF elongation, followed by agglomeration of platelets in the flow by soluble VWF entanglement before mural capture of the agglomerate by immobilized VWF. The entire SIPA process occurs on the order of 10 milliseconds with the agglomerate traveling a lag distance of a few hundred microns before capture, matching in vitro results. Increasing soluble VWF concentration by ∼20 times in silico leads to a ∼2 to 3 times increase in SIPA rates, matching the increase in occlusion rates found in vitro. The morphology of mural aggregates is primarily controlled by VWF molecular weight (length), where normal-length VWF leads to cluster or elongated aggregates and ultra-long VWF leads to loose aggregates seen by others' experiments. Finally, we present phase diagrams of SIPA, which provides biomechanistic rationales for a variety of thrombotic and hemostatic events in terms of platelet agglomeration and capture.
Collapse
Affiliation(s)
- Zixiang Leonardo Liu
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| | | | - Cyrus K. Aidun
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - David N. Ku
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
43
|
Morrell CN, Mix D, Aggarwal A, Bhandari R, Godwin M, Owens Iii AP, Lyden SP, Doyle A, Krauel K, Rondina MT, Mohan A, Lowenstein CJ, Shim S, Stauffer S, Josyula VP, Ture SK, Yule DI, Wagner Iii LE, Ashton JM, Elbadawi A, Cameron SJ. Platelet olfactory receptor activation limits platelet reactivity and growth of aortic aneurysms. J Clin Invest 2022; 132:152373. [PMID: 35324479 PMCID: PMC9057618 DOI: 10.1172/jci152373] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 03/16/2022] [Indexed: 11/28/2022] Open
Abstract
As blood transitions from steady laminar flow (S-flow) in healthy arteries to disturbed flow (D-flow) in aneurysmal arteries, platelets are subjected to external forces. Biomechanical platelet activation is incompletely understood and is a potential mechanism behind antiplatelet medication resistance. Although it has been demonstrated that antiplatelet drugs suppress the growth of abdominal aortic aneurysms (AAA) in patients, we found that a certain degree of platelet reactivity persisted in spite of aspirin therapy, urging us to consider additional antiplatelet therapeutic targets. Transcriptomic profiling of platelets from patients with AAA revealed upregulation of a signal transduction pathway common to olfactory receptors, and this was explored as a mediator of AAA progression. Healthy platelets subjected to D-flow ex vivo, platelets from patients with AAA, and platelets in murine models of AAA demonstrated increased membrane olfactory receptor 2L13 (OR2L13) expression. A drug screen identified a molecule activating platelet OR2L13, which limited both biochemical and biomechanical platelet activation as well as AAA growth. This observation was further supported by selective deletion of the OR2L13 ortholog in a murine model of AAA that accelerated aortic aneurysm growth and rupture. These studies revealed that olfactory receptors regulate platelet activation in AAA and aneurysmal progression through platelet-derived mediators of aortic remodeling.
Collapse
Affiliation(s)
- Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - Doran Mix
- Department of Surgery, Division of Vascular Surgery, University of Rochester School of Medicine, Rochester, United States of America
| | - Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Rohan Bhandari
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Matthew Godwin
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - A Phillip Owens Iii
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, United States of America
| | - Sean P Lyden
- Department of Vascular Surgery, Cleveland Clinic, Cleveland, United States of America
| | - Adam Doyle
- Department of Surgery, Division of Vascular Surgery, University of Rochester School of Medicine, Rochester, United States of America
| | - Krystin Krauel
- Department of Molecular Medicine, University of Utah, Salt Lake City, United States of America
| | - Matthew T Rondina
- Department of Internal Medicine, University of Utah, Salt Lake City, United States of America
| | - Amy Mohan
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - Charles J Lowenstein
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, United States of America
| | - Sharon Shim
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Shaun Stauffer
- Center for Therapeutics Discovery, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Vara Prasad Josyula
- Center for Therapeutics Discovery, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Sara K Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, United States of America
| | - Larry E Wagner Iii
- Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, United States of America
| | - John M Ashton
- Department of Biomedical Genetics, University of Rochester School of Medicine, Rochester, United States of America
| | - Ayman Elbadawi
- Department of Cardiovascular Medicine, University of Texas Medical Branch, Galveston, United States of America
| | - Scott J Cameron
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| |
Collapse
|
44
|
Su S, Ling Y, Fang Y, Wu J. Force-enhanced biophysical connectivity of platelet β3 integrin signaling through Talin is predicted by steered molecular dynamics simulations. Sci Rep 2022; 12:4605. [PMID: 35301368 PMCID: PMC8931153 DOI: 10.1038/s41598-022-08554-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 02/28/2022] [Indexed: 01/01/2023] Open
Abstract
Platelet β3-integrin signaling through Talin is crucial in platelet transmembrane signaling, activation, adhesion, spreading and aggregation, and remains unclear in mechano-microenvironments. In order to examine Talin-β3 integrin biophysical connectivity, a series of “ramp-clamp” steered molecular dynamics (SMD) simulations were performed on complex of F3 domain of Talin and cytoplasmic tail of β3 integrin to imitate different force-loads in platelet. Pull-induced allostery of the hydrophobic pocket in F3 domain might markedly enhance complex rupture-force (> 150pN) and slow down breakage of the complex; the complex should mechano-stable for its conformational conservation under loads (≤ 80pN); increasing force below 60pN would decrease the complex dissociation probability, and force-induced extension of β5 strand on Talin and binding site residues, ASP740 and ALA742 as well as Asn744, on β3-integrin were responsible for the force-enhanced linkage of the Talin-β3 integrin. Force might enhance biophysical connectivity of β3-integrin signaling through Talin by a catch bond mechanism, which be mediated by the force-induced allostery of complex at clamped stage. This work provides a novel insight into the force-regulated transmembrane β3-integrin signaling and its molecular basis for platelet activation, and exhibited a potential power of the present computer strategy in predicting mechanical regulation on ligand-receptor interaction under loads.
Collapse
Affiliation(s)
- Shuixiu Su
- Institute of Biomechanics/School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yingchen Ling
- Institute of Biomechanics/School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Ying Fang
- Institute of Biomechanics/School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China.
| | - Jianhua Wu
- Institute of Biomechanics/School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
45
|
Wang H, Obeidy P, Wang Z, Zhao Y, Wang Y, Su QP, Cox CD, Ju LA. Fluorescence-coupled micropipette aspiration assay to examine calcium mobilization caused by red blood cell mechanosensing. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2022; 51:135-146. [PMID: 35286429 PMCID: PMC8964638 DOI: 10.1007/s00249-022-01595-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/16/2022]
Abstract
Mechanical stimuli such as tension, compression, and shear stress play critical roles in the physiological functions of red blood cells (RBCs) and their homeostasis, ATP release, and rheological properties. Intracellular calcium (Ca2+) mobilization reflects RBC mechanosensing as they transverse the complex vasculature. Emerging studies have demonstrated the presence of mechanosensitive Ca2+ permeable ion channels and their function has been implicated in the regulation of RBC volume and deformability. However, how these mechanoreceptors trigger Ca2+ influx and subsequent cellular responses are still unclear. Here, we introduce a fluorescence-coupled micropipette aspiration assay to examine RBC mechanosensing at the single-cell level. To achieve a wide range of cell aspirations, we implemented and compared two negative pressure adjusting apparatuses: a homemade water manometer (- 2.94 to 0 mmH2O) and a pneumatic high-speed pressure clamp (- 25 to 0 mmHg). To visualize Ca2+ influx, RBCs were pre-loaded with an intensiometric probe Cal-520 AM, then imaged under a confocal microscope with concurrent bright-field and fluorescent imaging at acquisition rates of 10 frames per second. Remarkably, we observed the related changes in intracellular Ca2+ levels immediately after aspirating individual RBCs in a pressure-dependent manner. The RBC aspirated by the water manometer only displayed 1.1-fold increase in fluorescence intensity, whereas the RBC aspirated by the pneumatic clamp showed up to threefold increase. These results demonstrated the water manometer as a gentle tool for cell manipulation with minimal pre-activation, while the high-speed pneumatic clamp as a much stronger pressure actuator to examine cell mechanosensing directly. Together, this multimodal platform enables us to precisely control aspiration and membrane tension, and subsequently correlate this with intracellular calcium concentration dynamics in a robust and reproducible manner.
Collapse
Affiliation(s)
- Haoqing Wang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.,Heart Research Institute, Newtown, NSW, 2042, Australia
| | - Peyman Obeidy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Zihao Wang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.,School of Aerospace, Mechanical and Mechatronic Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Yunduo Zhao
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.,Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
| | - Yao Wang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.,Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Qian Peter Su
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.,Heart Research Institute, Newtown, NSW, 2042, Australia.,School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia.,Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia. .,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia. .,Heart Research Institute, Newtown, NSW, 2042, Australia.
| |
Collapse
|
46
|
Wang H, Zhou F, Guo Y, Ju LA. Micropipette-based biomechanical nanotools on living cells. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2022; 51:119-133. [PMID: 35171346 PMCID: PMC8964576 DOI: 10.1007/s00249-021-01587-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Mechanobiology is an emerging field at the interface of biology and mechanics, investigating the roles of mechanical forces within biomolecules, organelles, cells, and tissues. As a highlight, the recent advances of micropipette-based aspiration assays and dynamic force spectroscopies such as biomembrane force probe (BFP) provide unprecedented mechanobiological insights with excellent live-cell compatibility. In their classic applications, these assays measure force-dependent ligand-receptor-binding kinetics, protein conformational changes, and cellular mechanical properties such as cortical tension and stiffness. In recent years, when combined with advanced microscopies in high spatial and temporal resolutions, these biomechanical nanotools enable characterization of receptor-mediated cell mechanosensing and subsequent organelle behaviors at single-cellular and molecular level. In this review, we summarize the latest developments of these assays for live-cell mechanobiology studies. We also provide perspectives on their future upgrades with multimodal integration and high-throughput capability.
Collapse
Affiliation(s)
- Haoqing Wang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia.,Heart Research Institute, Newtown, NSW, Australia
| | - Fang Zhou
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia
| | - Yuze Guo
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia. .,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia. .,Heart Research Institute, Newtown, NSW, Australia.
| |
Collapse
|
47
|
Zhang Y, Jiang F, Chen Y, Ju LA. Platelet Mechanobiology Inspired Microdevices: From Hematological Function Tests to Disease and Drug Screening. Front Pharmacol 2022; 12:779753. [PMID: 35126120 PMCID: PMC8811026 DOI: 10.3389/fphar.2021.779753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/28/2021] [Indexed: 12/30/2022] Open
Abstract
Platelet function tests are essential to profile platelet dysfunction and dysregulation in hemostasis and thrombosis. Clinically they provide critical guidance to the patient management and therapeutic evaluation. Recently, the biomechanical effects induced by hemodynamic and contractile forces on platelet functions attracted increasing attention. Unfortunately, the existing platelet function tests on the market do not sufficiently incorporate the topical platelet mechanobiology at play. Besides, they are often expensive and bulky systems that require large sample volumes and long processing time. To this end, numerous novel microfluidic technologies emerge to mimic vascular anatomies, incorporate hemodynamic parameters and recapitulate platelet mechanobiology. These miniaturized and cost-efficient microfluidic devices shed light on high-throughput, rapid and scalable platelet function testing, hematological disorder profiling and antiplatelet drug screening. Moreover, the existing antiplatelet drugs often have suboptimal efficacy while incurring several adverse bleeding side effects on certain individuals. Encouraged by a few microfluidic systems that are successfully commercialized and applied to clinical practices, the microfluidics that incorporate platelet mechanobiology hold great potential as handy, efficient, and inexpensive point-of-care tools for patient monitoring and therapeutic evaluation. Hereby, we first summarize the conventional and commercially available platelet function tests. Then we highlight the recent advances of platelet mechanobiology inspired microfluidic technologies. Last but not least, we discuss their future potential of microfluidics as point-of-care tools for platelet function test and antiplatelet drug screening.
Collapse
Affiliation(s)
- Yingqi Zhang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
| | - Fengtao Jiang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Yunfeng Chen
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- The Department of Pathology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
- *Correspondence: Lining Arnold Ju,
| |
Collapse
|
48
|
Li M, Wang L, Tang D, Zhao G, Ni Z, Gu N, Yang F. Hemodynamic Mimic Shear Stress for Platelet Membrane Nanobubbles Preparation and Integrin α IIbβ 3 Conformation Regulation. NANO LETTERS 2022; 22:271-279. [PMID: 34894698 DOI: 10.1021/acs.nanolett.1c03731] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Platelet (PLT) membrane biomimetic nanomaterials have become promising theranostic platforms due to their good biocompatibility and effectiveness. However, in order to achieve precise regulation of cell membrane components, novel controllable construction approaches need to be developed. Inspired by the interaction mechanism among platelet production, activation, and dynamic biomechanical signals in blood circulation, here a platelet nanobubbles (PNBs) with reassembled platelet membrane with ideal echogenicity was fabricated using an adjustable pressure-induced shear stress method. The results demonstrate that the high shear stress during PNBs fabrication led to the enrichment of platelet membrane lipid rafts and proteins, as well as their reassembly on the gas-liquid interface. More importantly, the conformation of platelet integrin αIIbβ3 was transformed into a shear stress-induced intermediate affinity state, which gives PNBs enhanced adhesion ability to the vascular endothelial injury. Taken together, these PNBs have great application potential in the specifically targeted ultrasound diagnosis of vascular endothelial injury.
Collapse
Affiliation(s)
- Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing 210009, China
| | - Liang Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Dalin Tang
- Mathematical Sciences Department, Worcester Polytechnic Institute, Worcester, Massachusetts 01609-2280, United States
| | - Gutian Zhao
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing 210009, China
| | - Zhonghua Ni
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing 210009, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
49
|
Hosseini E, Solouki A, Haghshenas M, Ghasemzadeh M, Schoenwaelder SM. Agitation-dependent biomechanical forces modulate GPVI receptor expression and platelet adhesion capacity during storage. Thromb J 2022; 20:3. [PMID: 35022046 PMCID: PMC8756730 DOI: 10.1186/s12959-021-00359-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/09/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Continuous agitation during storage slows down the platelet storage lesions. However, in special circumstances, manual-mixing can be alternatively used to store products for short time periods without compromising platelet quality. Based on this finding, and given the role of shear stress in modulating receptor expression, we were interested in comparing the levels of platelet adhesion receptor, GPVI and platelet adhesion capacity under each storage condition. METHODS Platelet concentrates (PCs) were divided into three groups: continuously-agitated PCs (CAG-PCs) with or without PP2 (Src kinase inhibitor) and manually-mixed PCs (MM-PCs). Platelet count/MPV, swirling, GPVI and P-selectin expression, GPVI shedding, platelet adhesion/spreading to collagen were examined during 5 days of storage. RESULTS While MM- and CAG-PCs showed similar levels of P-selectin expression, GPVI expression was significantly elevated in MM-PCs with lower GPVI shedding/expression ratios, enhanced platelet adhesion/spreading and swirling in manually-mixed PCs. Of note, CAG-PCs treated with PP2 also demonstrated lower P-selectin expression and GPVI shedding, higher GPVI expression and attenuated swirling and spreading capability. CONCLUSION Given the comparable platelet activation state in MM and CAG-PCs as indicated by P-selectin expression, enhanced platelet adhesion/spreading in MM-PCs, along with relatively higher GPVI expression here, supports previous studies demonstrating a role for biomechanical forces in modulating GPVI-dependent function. Thus, lower GPVI expression in CAG-PCs may be due to shear forces induced by agitation, which keeps this receptor down-regulated while also attenuating platelet adhesion/spreading capacities during storage. Low platelet function in PP2-CAG-PCs also highlights the importance of Src-kinases threshold activity in maintaining platelets quality.
Collapse
Affiliation(s)
- Ehteramolsadat Hosseini
- Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Amin Solouki
- Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Masood Haghshenas
- Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| | - Simone M Schoenwaelder
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia.,Heart Research Institute, Newtown, NSW, Australia
| |
Collapse
|
50
|
Ju LA, Kossmann S, Zhao YC, Moldovan L, Zhang Y, De Zoysa Ramasundara S, Zhou F, Lu H, Alwis I, Schoenwaelder SM, Yuan Y, Jackson SP. Microfluidic post method for 3-dimensional modeling of platelet–leukocyte interactions. Analyst 2022; 147:1222-1235. [DOI: 10.1039/d2an00270a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
These studies demonstrate the versatility and relevance of a novel ‘platelet post’ model to examine the adhesive interactions between platelets and neutrophils under 3D disturbed flow conditions relevant to thromboinflammation.
Collapse
Affiliation(s)
- Lining Arnold Ju
- Heart Research Institute, Newtown, NSW, 2042, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Sabine Kossmann
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yunduo Charles Zhao
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Laura Moldovan
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yingqi Zhang
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Savindi De Zoysa Ramasundara
- Heart Research Institute, Newtown, NSW, 2042, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Imala Alwis
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Simone M. Schoenwaelder
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yuping Yuan
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Shaun P. Jackson
- Heart Research Institute, Newtown, NSW, 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, California 92037, USA
| |
Collapse
|