1
|
Fanaee S, Austin W, Filiaggi M, Adibnia V. External Bleeding and Advanced Biomacromolecules for Hemostasis. Biomacromolecules 2024. [PMID: 39463174 DOI: 10.1021/acs.biomac.4c00952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Hemorrhage is a significant medical problem that has been an active area of research over the past few decades. The human body has a complex response to bleeding that leads to blood clot formation and hemostasis. Many biomaterials based on various biomacromolecules have been developed to either accelerate or improve the body's natural response to bleeding. This review examines the mechanisms of hemostasis, types of bleeding, and the in vitro or in vivo models and techniques used to study bleeding and hemostatic materials. It provides a detailed overview of the diverse hemostatic materials, including those that are highly absorbent, wet adhesives, and those that accelerate the biochemical cascade of blood clotting. These materials are currently marketed, under preclinical testing, or being researched. In exploring the latest advancements in hemostatic technologies, this paper highlights the potential of these materials to significantly improve bleeding control in clinical and emergency situations.
Collapse
Affiliation(s)
- Sajjad Fanaee
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - William Austin
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Mark Filiaggi
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Vahid Adibnia
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Chemistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
2
|
Shayor AA, Kabir ME, Rifath MSA, Rashid AB, Oh KW. A Synergistic Overview between Microfluidics and Numerical Research for Vascular Flow and Pathological Investigations. SENSORS (BASEL, SWITZERLAND) 2024; 24:5872. [PMID: 39338617 PMCID: PMC11435959 DOI: 10.3390/s24185872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/20/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024]
Abstract
Vascular diseases are widespread, and sometimes such life-threatening medical disorders cause abnormal blood flow, blood particle damage, changes to flow dynamics, restricted blood flow, and other adverse effects. The study of vascular flow is crucial in clinical practice because it can shed light on the causes of stenosis, aneurysm, blood cancer, and many other such diseases, and guide the development of novel treatments and interventions. Microfluidics and computational fluid dynamics (CFDs) are two of the most promising new tools for investigating these phenomena. When compared to conventional experimental methods, microfluidics offers many benefits, including lower costs, smaller sample quantities, and increased control over fluid flow and parameters. In this paper, we address the strengths and weaknesses of computational and experimental approaches utilizing microfluidic devices to investigate the rheological properties of blood, the forces of action causing diseases related to cardiology, provide an overview of the models and methodologies of experiments, and the fabrication of devices utilized in these types of research, and portray the results achieved and their applications. We also discuss how these results can inform clinical practice and where future research should go. Overall, it provides insights into why a combination of both CFDs, and experimental methods can give even more detailed information on disease mechanisms recreated on a microfluidic platform, replicating the original biological system and aiding in developing the device or chip itself.
Collapse
Affiliation(s)
- Ahmed Abrar Shayor
- Department of Mechanical Engineering, Khulna University of Engineering & Technology, Khulna 9203, Bangladesh
| | - Md Emamul Kabir
- Department of Mechanical Engineering, Khulna University of Engineering & Technology, Khulna 9203, Bangladesh
- Sensors and MicroActuators Learning Lab (SMALL), Department of Electrical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - Md Sartaj Ahamed Rifath
- Department of Mechanical Engineering, Khulna University of Engineering & Technology, Khulna 9203, Bangladesh
| | - Adib Bin Rashid
- Department of Industrial and Production Engineering, Military Institute of Science and Technology, Dhaka 1216, Bangladesh
| | - Kwang W Oh
- Sensors and MicroActuators Learning Lab (SMALL), Department of Electrical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
3
|
Dasgupta I, Rangineni DP, Abdelsaid H, Ma Y, Bhushan A. Tiny Organs, Big Impact: How Microfluidic Organ-on-Chip Technology Is Revolutionizing Mucosal Tissues and Vasculature. Bioengineering (Basel) 2024; 11:476. [PMID: 38790343 PMCID: PMC11117503 DOI: 10.3390/bioengineering11050476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-chip (OOC) technology has gained importance for biomedical studies and drug development. This technology involves microfluidic devices that mimic the structure and function of specific human organs or tissues. OOCs are a promising alternative to traditional cell-based models and animals, as they provide a more representative experimental model of human physiology. By creating a microenvironment that closely resembles in vivo conditions, OOC platforms enable the study of intricate interactions between different cells as well as a better understanding of the underlying mechanisms pertaining to diseases. OOCs can be integrated with other technologies, such as sensors and imaging systems to monitor real-time responses and gather extensive data on tissue behavior. Despite these advances, OOCs for many organs are in their initial stages of development, with several challenges yet to be overcome. These include improving the complexity and maturity of these cellular models, enhancing their reproducibility, standardization, and scaling them up for high-throughput uses. Nonetheless, OOCs hold great promise in advancing biomedical research, drug discovery, and personalized medicine, benefiting human health and well-being. Here, we review several recent OOCs that attempt to overcome some of these challenges. These OOCs with unique applications can be engineered to model organ systems such as the stomach, cornea, blood vessels, and mouth, allowing for analyses and investigations under more realistic conditions. With this, these models can lead to the discovery of potential therapeutic interventions. In this review, we express the significance of the relationship between mucosal tissues and vasculature in organ-on-chip (OOC) systems. This interconnection mirrors the intricate physiological interactions observed in the human body, making it crucial for achieving accurate and meaningful representations of biological processes within OOC models. Vasculature delivers essential nutrients and oxygen to mucosal tissues, ensuring their proper function and survival. This exchange is critical for maintaining the health and integrity of mucosal barriers. This review will discuss the OOCs used to represent the mucosal architecture and vasculature, and it can encourage us to think of ways in which the integration of both can better mimic the complexities of biological systems and gain deeper insights into various physiological and pathological processes. This will help to facilitate the development of more accurate predictive models, which are invaluable for advancing our understanding of disease mechanisms and developing novel therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA; (I.D.); (D.P.R.); (H.A.); (Y.M.)
| |
Collapse
|
4
|
Provenzale I, Solari FA, Schönichen C, Brouns SLN, Fernández DI, Kuijpers MJE, van der Meijden PEJ, Gibbins JM, Sickmann A, Jones C, Heemskerk JWM. Endothelium-mediated regulation of platelet activation: Involvement of multiple protein kinases. FASEB J 2024; 38:e23468. [PMID: 38334433 DOI: 10.1096/fj.202300360rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
The endothelial regulation of platelet activity is incompletely understood. Here we describe novel approaches to find molecular pathways implicated on the platelet-endothelium interaction. Using high-shear whole-blood microfluidics, employing coagulant or non-coagulant conditions at physiological temperature, we observed that the presence of human umbilical vein endothelial cells (HUVEC) strongly suppressed platelet adhesion and activation, via the collagen receptor glycoprotein VI (GPVI) and the PAR receptors for thrombin. Real-time monitoring of the cytosolic Ca2+ rises in the platelets indicated no major improvement of inhibition by prostacyclin or nitric oxide. Similarly under stasis, exposure of isolated platelets to HUVEC reduced the Ca2+ responses by collagen-related peptide (CRP-XL, GPVI agonist) and thrombin (PAR agonist). We then analyzed the label-free phosphoproteome of platelets (three donors), exposed to HUVEC, CRP-XL, and/or thrombin. High-resolution mass spectrometry gave 5463 phosphopeptides, corresponding to 1472 proteins, with good correlation between biological and technical replicates (R > .86). Stringent filtering steps revealed 26 regulatory pathways (Reactome) and 143 regulated kinase substrates (PhosphoSitePlus), giving a set of protein phosphorylation sites that was differentially (44) or similarly (110) regulated by HUVEC or agonist exposure. The differential regulation was confirmed by stable-isotope analysis of platelets from two additional donors. Substrate analysis indicated major roles of poorly studied protein kinase classes (MAPK, CDK, DYRK, STK, PKC members). Collectively, these results reveal a resetting of the protein phosphorylation profile in platelets exposed to endothelium or to conventional agonists and to endothelium-promoted activity of a multi-kinase network, beyond classical prostacyclin and nitric oxide actors, that may contribute to platelet inhibition.
Collapse
Affiliation(s)
- Isabella Provenzale
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Claudia Schönichen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Thrombosis and Haemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sanne L N Brouns
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Delia I Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Chris Jones
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Synapse Research Institute Maastricht, Maastricht, The Netherlands
| |
Collapse
|
5
|
Sakurai Y, Hardy ET, Lam WA. Hemostasis-on-a-chip / incorporating the endothelium in microfluidic models of bleeding. Platelets 2023; 34:2185453. [PMID: 36872890 DOI: 10.1080/09537104.2023.2185453] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Currently, point-of-care assays for human platelet function and coagulation are used to assess bleeding risks and drug testing, but they lack intact endothelium, a critical component of the human vascular system. Within these assays, the assessment of bleeding risk is typically indicated by the lack of or reduced platelet function and coagulation without true evaluation of hemostasis. Hemostasis is defined as the cessation of bleeding. Additionally, animal models of hemostasis also, by definition, lack human endothelium, which may limit their clinical relevance. This review discusses the current state-of-the-art of hemostasis-on-a-chip, specifically, human cell-based microfluidic models that incorporate endothelial cells, which function as physiologically relevant in vitro models of bleeding. These assays recapitulate the entire process of vascular injury, bleeding, and hemostasis, and provide real-time, direct observation, thereby serving as research-enabling tools that enhance our understanding of hemostasis and also as novel drug discovery platforms.
Collapse
Affiliation(s)
- Yumiko Sakurai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Elaissa T Hardy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, GA, USA
| | - Wilbur A Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, GA, USA
| |
Collapse
|
6
|
Yadav SK, Park S, Lee YM, Hurh S, Kim D, Min S, Kim S, Yan JJ, Kang BC, Kim S, Yang J, Jeong JC. Application of microphysiologic system to assess neutrophil extracellular trap in xenotransplantation. J Immunol Methods 2023; 521:113537. [PMID: 37598787 DOI: 10.1016/j.jim.2023.113537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Transplantation of organs, cells, or tissues from one species to another, known as xenotransplantation, has the potential to alleviate organ donor shortages and enhance the success of organ transplantation. However, the possibility of immunological rejection by the recipient is one of the biggest difficulties associated with xenotransplantation. The creation of neutrophil extracellular traps (NETs), also known as NETosis, is hypothesized as a mechanism of rejection. Innovations in microfluidics and co-culturing techniques have provided access to several classes of microengineered model systems in experimental models, connecting animal research and traditional in vitro methods such as organoids, microphysiological systems, and organs-on-chip. To achieve this goal, we established a perfusable 3D Xeno vessel chip using a porcine aortic endothelial cell line and examined how NETs grow when isolated human and primate neutrophils were used. Neutrophils from both humans and monkeys displayed the usual NETosis phases, including nuclear decondensation, enlargement, and rounding of DNA, occupying the entire cytoplasm, and discharge of fragmented DNA after cell membrane rupture. Using confocal fluorescence imaging of DNA and citrullinated histone colocalization and DNA histone complex formation in supernatants from xeno vessel chips, we confirmed NETs generation by human and monkey neutrophils when cocultured in a xeno-vessel chip.
Collapse
Affiliation(s)
- Suchen Kumar Yadav
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Seokwoo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Yun-Mi Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Sunghoon Hurh
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.
| | | | - Sangil Min
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea.
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Ji-Jing Yan
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| | - Byeong-Cheol Kang
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea.
| | | | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| | - Jong Cheol Jeong
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| |
Collapse
|
7
|
Hearn JI, Gardiner EE. Research and Clinical Approaches to Assess Platelet Function in Flowing Blood. Arterioscler Thromb Vasc Biol 2023; 43:1775-1783. [PMID: 37615110 DOI: 10.1161/atvbaha.123.317048] [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: 08/25/2023]
Abstract
Platelet adhesion and activation is fundamental to the formation of a hemostatic response to limit loss of blood and instigate wound repair to seal a site of vascular injury. The process of platelet aggregate formation is supported by the coagulation system driving injury-proximal formation of thrombin, which converts fibrinogen to insoluble fibrin. This highly coordinated series of molecular and membranous events must be routinely achieved in flowing blood, at vascular fluid shear rates that place significant strain on molecular and cellular interactions. Platelets have long been recognized to be able to slow down and adhere to sites of vascular injury and then activate and recruit more platelets that forge and strengthen adhesive ties with the vascular wall under these conditions. It has been a major challenge for the Platelet Research Community to construct experimental conditions that allow precise definition of the molecular steps occurring under flow. This brief review will discuss work to date from our group, as well as others that has furthered our understanding of platelet function in flowing blood.
Collapse
Affiliation(s)
- James I Hearn
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
8
|
Golomingi M, Kohler J, Lamers C, Pouw RB, Ricklin D, Dobó J, Gál P, Pál G, Kiss B, Dopler A, Schmidt CQ, Hardy ET, Lam W, Schroeder V. Complement inhibition can decrease the haemostatic response in a microvascular bleeding model at multiple levels. Front Immunol 2023; 14:1226832. [PMID: 37771595 PMCID: PMC10525698 DOI: 10.3389/fimmu.2023.1226832] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
Background Haemostasis is a crucial process by which the body stops bleeding. It is achieved by the formation of a platelet plug, which is strengthened by formation of a fibrin mesh mediated by the coagulation cascade. In proinflammatory and prothrombotic conditions, multiple interactions of the complement system and the coagulation cascade are known to aggravate thromboinflammatory processes and increase the risk of arterial and venous thrombosis. Whether those interactions also play a relevant role during the physiological process of haemostasis is not yet completely understood. The aim of this study was to investigate the potential role of complement components and activation during the haemostatic response to mechanical vessel injury. Methods We used a microvascular bleeding model that simulates a blood vessel, featuring human endothelial cells, perfusion with fresh human whole blood, and an inducible mechanical injury to the vessel. We studied the effects of complement inhibitors against components of the lectin (MASP-1, MASP-2), classical (C1s), alternative (FD) and common pathways (C3, C5), as well as a novel triple fusion inhibitor of all three complement pathways (TriFu). Effects on clot formation were analysed by recording of fibrin deposition and the platelet activation marker CD62P at the injury site in real time using a confocal microscope. Results With the inhibitors targeting MASP-2 or C1s, no significant reduction of fibrin formation was observed, while platelet activation was significantly reduced in the presence of the FD inhibitor. Both common pathway inhibitors targeting C3 or C5, respectively, were associated with a substantial reduction of fibrin formation, and platelet activation was also reduced in the presence of the C3 inhibitor. Triple inhibition of all three activation pathways at the C3-convertase level by TriFu reduced both fibrin formation and platelet activation. When several complement inhibitors were directly compared in two individual donors, TriFu and the inhibitors of MASP-1 and C3 had the strongest effects on clot formation. Conclusion The observed impact of complement inhibition on reducing fibrin clot formation and platelet activation suggests a role of the complement system in haemostasis, with modulators of complement initiation, amplification or effector functions showing distinct profiles. While the interactions between complement and coagulation might have evolved to support haemostasis and protect against bleeding in case of vessel injury, they can turn harmful in pathological conditions when aggravating thromboinflammation and promoting thrombosis.
Collapse
Affiliation(s)
- Murielle Golomingi
- Experimental Haemostasis Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jessie Kohler
- Experimental Haemostasis Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Christina Lamers
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Richard B. Pouw
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Daniel Ricklin
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Bence Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Arthur Dopler
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Christoph Q. Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Elaissa Trybus Hardy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Wilbur Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Verena Schroeder
- Experimental Haemostasis Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
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
|
10
|
Jia X, Hua C, Yang F, Li X, Zhao P, Zhou F, Lu Y, Liang H, Xing M, Lyu G. Hydrophobic aerogel-modified hemostatic gauze with thermal management performance. Bioact Mater 2023; 26:142-158. [PMID: 36911208 PMCID: PMC9996136 DOI: 10.1016/j.bioactmat.2023.02.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Current hemostatic agents or dressings are not efficient under extremely hot and cold environments due to deterioration of active ingredients, water evaporation and ice crystal growth. To address these challenges, we engineered a biocompatible hemostatic system with thermoregulatory properties for harsh conditions by combining the asymmetric wetting nano-silica aerogel coated-gauze (AWNSA@G) with a layer-by-layer (LBL) structure. Our AWNSA@G was a dressing with a tunable wettability prepared by spraying the hydrophobic nano-silica aerogel onto the gauze from different distances. The hemostatic time and blood loss of the AWNSA@G were 5.1 and 6.9 times lower than normal gauze in rat's injured femoral artery model. Moreover, the modified gauze was torn off after hemostasis without rebleeding, approximately 23.8 times of peak peeling force lower than normal gauze. For the LBL structure, consisting of the nano-silica aerogel layer and a n-octadecane phase change material layer, in both hot (70 °C) and cold (-27 °C) environments, exhibited dual-functional thermal management and maintained a stable internal temperature. We further verified our composite presented superior blood coagulation effect in extreme environments due to the LBL structure, the pro-coagulant properties of nano-silica aerogel and unidirectional fluid pumping of AWNSA@G. Our work, therefore, shows great hemostasis potential under normal and extreme temperature environments.
Collapse
Affiliation(s)
- Xiaoli Jia
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Chao Hua
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China
| | - Fengbo Yang
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaoxiao Li
- Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Peng Zhao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Feifan Zhou
- Medical School of Nantong University, Nantong, 226019, China
| | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Hao Liang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Guozhong Lyu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China.,Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
11
|
Barrett L, Curry N, Abu-Hanna J. Experimental Models of Traumatic Injuries: Do They Capture the Coagulopathy and Underlying Endotheliopathy Induced by Human Trauma? Int J Mol Sci 2023; 24:11174. [PMID: 37446351 PMCID: PMC10343021 DOI: 10.3390/ijms241311174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Trauma-induced coagulopathy (TIC) is a major cause of morbidity and mortality in patients with traumatic injury. It describes the spectrum of coagulation abnormalities that occur because of the trauma itself and the body's response to the trauma. These coagulation abnormalities range from hypocoagulability and hyperfibrinolysis, resulting in potentially fatal bleeding, in the early stages of trauma to hypercoagulability, leading to widespread clot formation, in the later stages. Pathological changes in the vascular endothelium and its regulation of haemostasis, a phenomenon known as the endotheliopathy of trauma (EoT), are thought to underlie TIC. Our understanding of EoT and its contribution to TIC remains in its infancy largely due to the scarcity of experimental research. This review discusses the mechanisms employed by the vascular endothelium to regulate haemostasis and their dysregulation following traumatic injury before providing an overview of the available experimental in vitro and in vivo models of trauma and their applicability for the study of the EoT and its contribution to TIC.
Collapse
Affiliation(s)
- Liam Barrett
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK;
- Emergency Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nicola Curry
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LD, UK
| | - Jeries Abu-Hanna
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
| |
Collapse
|
12
|
Angelidakis E, Chen S, Zhang S, Wan Z, Kamm RD, Shelton SE. Impact of Fibrinogen, Fibrin Thrombi, and Thrombin on Cancer Cell Extravasation Using In Vitro Microvascular Networks. Adv Healthc Mater 2023; 12:e2202984. [PMID: 37119127 PMCID: PMC10524192 DOI: 10.1002/adhm.202202984] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/25/2023] [Indexed: 04/30/2023]
Abstract
A bidirectional association exists between metastatic dissemination and the hypercoagulable state associated with many types of cancer. As such, clinical studies have provided evidence that markers associated with elevated levels of coagulation and fibrinolysis correlate with decreased patient survival. However, elucidating the mechanisms underpinning the effects of different components of the coagulation system on metastasis formation is challenging both in animal models and 2D models lacking the complex cellular interactions necessary to model both thrombosis and metastasis. Here, an in vitro, 3D, microvascular model for observing the formation of fibrin thrombi is described, which is in turn used to study how different aspects of the hypercoagulable state associated with cancer affect the endothelium. Using this platform, cancer cells expressing ICAM-1 are shown to form a fibrinogen-dependent bridge and transmigrate through the endothelium more effectively. Cancer cells are also demonstrated to interact with fibrin thrombi, using them to adhere, spread, and enhance their extravasation efficiency. Finally, thrombin is also shown to enhance cancer cell extravasation. This system presents a physiologically relevant model of fibrin clot formation in the human microvasculature, enabling in-depth investigation of the cellular interactions between cancer cells and the coagulation system affecting cancer cell extravasation.
Collapse
Affiliation(s)
- Emmanouil Angelidakis
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sophia Chen
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Shun Zhang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Zhengpeng Wan
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah E. Shelton
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medical OncologyDana Farber Cancer InstituteBostonMA02215USA
| |
Collapse
|
13
|
Xu W, Zhang M, Du W, Ling G, Yuan Y, Zhang P. Engineering a naturally-derived wound dressing based on bio-ionic liquid conjugation. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.112055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
14
|
Zheng Y, Wu J, Zhu Y, Wu C. Inorganic-based biomaterials for rapid hemostasis and wound healing. Chem Sci 2022; 14:29-53. [PMID: 36605747 PMCID: PMC9769395 DOI: 10.1039/d2sc04962g] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 12/02/2022] Open
Abstract
The challenge for the treatment of severe traumas poses an urgent clinical need for the development of biomaterials to achieve rapid hemostasis and wound healing. In the past few decades, active inorganic components and their derived composites have become potential clinical products owing to their excellent performances in the process of hemorrhage control and tissue repair. In this review, we provide a current overview of the development of inorganic-based biomaterials used for hemostasis and wound healing. We highlight the methods and strategies for the design of inorganic-based biomaterials, including 3D printing, freeze-drying, electrospinning and vacuum filtration. Importantly, inorganic-based biomaterials for rapid hemostasis and wound healing are presented, and we divide them into several categories according to different chemistry and forms and further discuss their properties, therapeutic mechanisms and applications. Finally, the conclusions and future prospects are suggested for the development of novel inorganic-based biomaterials in the field of rapid hemostasis and wound healing.
Collapse
Affiliation(s)
- Yi Zheng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences No. 1295 Dingxi Road Shanghai 200050 People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences No. 19(A) Yuquan Road Beijing 100049 People's Republic of China
| | - Jinfu Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences No. 1295 Dingxi Road Shanghai 200050 People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences No. 19(A) Yuquan Road Beijing 100049 People's Republic of China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences No. 1295 Dingxi Road Shanghai 200050 People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences No. 19(A) Yuquan Road Beijing 100049 People's Republic of China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences No. 1295 Dingxi Road Shanghai 200050 People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences No. 19(A) Yuquan Road Beijing 100049 People's Republic of China
| |
Collapse
|
15
|
Ganguly K, Espinal MM, Dutta SD, Patel DK, Patil TV, Luthfikasari R, Lim* KT. Multifunctional 3D platforms for rapid hemostasis and wound healing: Structural and functional prospects at biointerfaces. Int J Bioprint 2022; 9:648. [PMID: 36844240 PMCID: PMC9947489 DOI: 10.18063/ijb.v9i1.648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/29/2022] [Indexed: 12/05/2022] Open
Abstract
354Fabrication of multifunctional hemostats is indispensable against chronic blood loss and accelerated wound healing. Various hemostatic materials that aid wound repair or rapid tissue regeneration has been developed in the last 5 years. This review provides an overview of the three-dimensional (3D) hemostatic platforms designed through the latest technologies like electrospinning, 3D printing, and lithography, solely or in combination, for application in rapid wound healing. We critically discuss the pivotal role of micro/nano-3D topography and biomaterial properties in mediating rapid blood clots and healing at the hemostat-biointerface. We also highlight the advantages and limitations of the designed 3D hemostats. We anticipate that this review will guide the fabrication of smart hemostats of the future for tissue engineering applications.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Maria Mercedes Espinal
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Dinesh K. Patel
- Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Rachmi Luthfikasari
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ki-Taek Lim*
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
16
|
Musick JO, Fibben KS, Lam WA. Hyperviscosity syndromes; hemorheology for physicians and the use of microfluidic devices. Curr Opin Hematol 2022; 29:290-296. [PMID: 35916537 PMCID: PMC9547821 DOI: 10.1097/moh.0000000000000735] [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: 11/26/2022]
Abstract
PURPOSE OF REVIEW Hyperviscosity syndromes can lead to significant morbidity and mortality. Existing methods to measure microcirculatory rheology are not readily available and limited in relevance and accuracy at this level. In this review, we review selected hyperviscosity syndromes and the advancement of their knowledge using microfluidic platforms. RECENT FINDINGS Viscosity changes drastically at the microvascular level as the physical properties of the cells themselves become the major determinants of resistance to blood flow. Current, outdated viscosity measurements only quantify whole blood or serum. Changes in blood composition, cell number, or the physical properties themselves lead to increased blood viscosity. Given the significant morbidity and mortality from hyperviscosity syndromes, new biophysical tools are needed and being developed to study microvascular biophysical and hemodynamic conditions at this microvascular level to help predict those at risk and guide therapeutic treatment. SUMMARY The use of 'lab-on-a-chip' technology continues to rise to relevance with point of care, personalized testing and medicine as customizable microfluidic platforms enable independent control of many in vivo factors and are a powerful tool to study microcirculatory hemorheology.
Collapse
Affiliation(s)
- Jamie O. Musick
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Kirby S. Fibben
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Wilbur A. Lam
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| |
Collapse
|
17
|
Chen L, Yu L, Liu Y, Xu H, Ma L, Tian P, Zhu J, Wang F, Yi K, Xiao H, Zhou F, Yang Y, Cheng Y, Bai L, Wang F, Zhu Y. Space-time-regulated imaging analyzer for smart coagulation diagnosis. Cell Rep Med 2022; 3:100765. [PMID: 36206751 PMCID: PMC9589004 DOI: 10.1016/j.xcrm.2022.100765] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/26/2022] [Accepted: 09/14/2022] [Indexed: 11/07/2022]
Abstract
The development of intelligent blood coagulation diagnoses is awaited to meet the current need for large clinical time-sensitive caseloads due to its efficient and automated diagnoses. Herein, a method is reported and validated to realize it through artificial intelligence (AI)-assisted optical clotting biophysics (OCB) properties identification. The image differential calculation is used for precise acquisition of OCB properties with elimination of initial differences, and the strategy of space-time regulation allows on-demand space time OCB properties identification and enables diverse blood function diagnoses. The integrated applications of smartphones and cloud computing offer a user-friendly automated analysis for accurate and convenient diagnoses. The prospective assays of clinical cases (n = 41) show that the system realizes 97.6%, 95.1%, and 100% accuracy for coagulation factors, fibrinogen function, and comprehensive blood coagulation diagnoses, respectively. This method should enable more low-cost and convenient diagnoses and provide a path for potential diagnostic-markers finding. An ultraportable optofluidic analyzer empowers convenient coagulation diagnoses The system enables optical clotting biophysics (OCB) properties acquisition and process Coagulation function diagnoses uses intelligent OCB properties identification Space-time regulation of OCB properties endow it capability to diverse diagnoses
Collapse
Affiliation(s)
- Longfei Chen
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China; Renmin Hospital, Wuhan University, Wuhan 430060, China; Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| | - Le Yu
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China; Renmin Hospital, Wuhan University, Wuhan 430060, China; Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| | - Yantong Liu
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China; Renmin Hospital, Wuhan University, Wuhan 430060, China; Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| | - Hongshan Xu
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China
| | - Linlu Ma
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Pengfu Tian
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China
| | - Jiaomeng Zhu
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China
| | - Fang Wang
- 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, Wuhan University, Wuhan 430071, China
| | - Hui Xiao
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yi Yang
- Key Laboratory of Artificial Micro- and Nano- Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China; Renmin Hospital, Wuhan University, Wuhan 430060, China; Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China.
| | | | - Long Bai
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yimin Zhu
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China
| |
Collapse
|
18
|
Golomingi M, Kohler J, Jenny L, Hardy ET, Dobó J, Gál P, Pál G, Kiss B, Lam WA, Schroeder V. Complement lectin pathway components MBL and MASP-1 promote haemostasis upon vessel injury in a microvascular bleeding model. Front Immunol 2022; 13:948190. [PMID: 36032172 PMCID: PMC9412763 DOI: 10.3389/fimmu.2022.948190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundComplement lectin pathway components, in particular mannan-binding lectin (MBL) and MBL-associated serine proteases (MASPs) have been shown to interact with coagulation factors and contribute to clot formation. Here we investigated the role of MBL and MASP-1 in the haemostatic response following mechanical vessel injury in a human microfluidic bleeding model.MethodsWe studied haemostasis in a microvascular bleeding model in the presence of human endothelial cells and human whole blood under flow conditions. We monitored incorporation of proteins into the clot with fluorescently labelled antibodies and studied their effects on clot formation, platelet activation, and bleeding time with specific inhibitors. Platelet activation was also studied by flow cytometry.ResultsUpon vessel injury, MBL accumulated at the injury site in a well-defined wall-like structure. MBL showed partial colocalisation with fibrin, and strong colocalisation with von Willebrand factor and (activated) platelets. Flow cytometry ruled out direct binding of MBL to platelets, but confirmed a PAR4- and thrombin-dependent platelet-activating function of MASP-1. Inhibiting MBL during haemostasis reduced platelet activation, while inhibiting MASP-1 reduced platelet activation, fibrin deposition and prolonged bleeding time.ConclusionWe show in a microvascular human bleeding model that MBL and MASP-1 have important roles in the haemostatic response triggered by mechanical vessel injury: MBL recognises the injury site, while MASP-1 increases fibrin formation, platelet activation and shortens bleeding time. While the complement lectin pathway may be harmful in the context of pathological thrombosis, it appears to be beneficial during the physiological coagulation response by supporting the crucial haemostatic system.
Collapse
Affiliation(s)
- Murielle Golomingi
- Experimental Haemostasis Group, Department for BioMedical Research, DBMR, University of Bern, Bern, Switzerland
| | - Jessie Kohler
- Experimental Haemostasis Group, Department for BioMedical Research, DBMR, University of Bern, Bern, Switzerland
| | - Lorenz Jenny
- Experimental Haemostasis Group, Department for BioMedical Research, DBMR, University of Bern, Bern, Switzerland
| | - Elaissa T. Hardy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Bence Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Verena Schroeder
- Experimental Haemostasis Group, Department for BioMedical Research, DBMR, University of Bern, Bern, Switzerland
- *Correspondence: Verena Schroeder,
| |
Collapse
|
19
|
Membrane curvature and PS localize coagulation proteins to filopodia and retraction fibers of endothelial cells. Blood Adv 2022; 7:60-72. [PMID: 35849711 PMCID: PMC9827038 DOI: 10.1182/bloodadvances.2021006870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Prior reports indicate that the convex membrane curvature of phosphatidylserine (PS)-containing vesicles enhances formation of binding sites for factor Va and lactadherin. Yet, the relationship of convex curvature to localization of these proteins on cells remains unknown. We developed a membrane topology model, using phospholipid bilayers supported by nano-etched silica substrates, to further explore the relationship between curvature and localization of coagulation proteins. Ridge convexity corresponded to maximal curvature of physiologic membranes (radii of 10 or 30 nm) and the troughs had a variable concave curvature. The benchmark PS probe lactadherin exhibited strong differential binding to the ridges, on membranes with 4% to 15% PS. Factor Va, with a PS-binding motif homologous to lactadherin, also bound selectively to the ridges. Bound factor Va supported coincident binding of factor Xa, localizing prothrombinase complexes to the ridges. Endothelial cells responded to prothrombotic stressors and stimuli (staurosporine, tumor necrosis factor-α [TNF- α]) by retracting cell margins and forming filaments and filopodia. These had a high positive curvature similar to supported membrane ridges and selectively bound lactadherin. Likewise, the retraction filaments and filopodia bound factor Va and supported assembly of prothrombinase, whereas the cell body did not. The perfusion of plasma over TNF-α-stimulated endothelia in culture dishes and engineered 3-dimensional microvessels led to fibrin deposition at cell margins, inhibited by lactadherin, without clotting of bulk plasma. Our results indicate that stressed or stimulated endothelial cells support prothrombinase activity localized to convex topological features at cell margins. These findings may relate to perivascular fibrin deposition in sepsis and inflammation.
Collapse
|
20
|
Chen L, Zheng Y, Liu Y, Tian P, Yu L, Bai L, Zhou F, Yang Y, Cheng Y, Wang F, Zheng L, Jiang F, Zhu Y. Microfluidic-based in vitro thrombosis model for studying microplastics toxicity. LAB ON A CHIP 2022; 22:1344-1353. [PMID: 35179168 DOI: 10.1039/d1lc00989c] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The potential impact of microplastics (MPs) on health has caused great concern, and a toxicology platform that realistically reproduces the system behaviour is urgently needed to further explore and validate MP-related health issues. Herein, we introduce an optically assisted thrombus platform to reveal the interaction of MPs with the vascular system. The risk of accumulation has also been evaluated using a mouse model, and the effect of MPs on the properties of the thrombus are validated via in vitro experiments. The microfluidic system is endothelialized, and the regional tissue injury-induced thrombosis is then realized through optical irradiation. Whole blood is perfused with MPs, and the invasion process visualized and recorded. The mouse model shows a cumulative risk in the blood with continuous exposure to MPs (P-value < 0.0001). The on-chip results show that MP invasion leads to decreased binding of fibrin to platelets (P-value < 0.0001), which is consistent with the results of the in vitro experiments, and shows a high risk of thrombus shedding in real blood flow compared with normal thrombus. This work provides a new method to further reveal MP-related health risks.
Collapse
Affiliation(s)
- Longfei Chen
- 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
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Yantong Liu
- 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
| | - Pengfu Tian
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China.
| | - Le Yu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, China.
| | - Long Bai
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yi Yang
- 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
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Li Zheng
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266061, China
| | - Fenghua Jiang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266061, China
| | - Yimin Zhu
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China
| |
Collapse
|
21
|
Berry J, Harper MT. Protease-activated receptor antagonists prevent thrombosis when dual antiplatelet therapy is insufficient in an occlusive thrombosis microfluidic model. Res Pract Thromb Haemost 2022; 6:e12703. [PMID: 35434469 PMCID: PMC9001860 DOI: 10.1002/rth2.12703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/11/2022] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Background Platelet activation and arterial thrombosis on a ruptured atherosclerotic plaque is a major cause of myocardial infarction. Dual antiplatelet therapy (DAPT), the combination of platelet aggregation inhibitors, aspirin and a P2Y12 antagonist, is used to prevent arterial thrombosis. However, many people continue to have arterial thrombosis and myocardial infarction despite DAPT, indicating that additional therapies are required where DAPT is insufficient. Objectives To determine whether antagonists of protease-activated receptors (PARs) can prevent occlusive thrombosis under conditions where DAPT is insufficient. Methods We used human whole blood in a microfluidic model of occlusive thrombosis to compare conditions under which DAPT is effective to those under which DAPT was not. Cangrelor (a P2Y12 antagonist) and aspirin were used to mimic DAPT. We then investigated whether the PAR1 antagonist vorapaxar or the PAR4 antagonist BMS 986120, alone or in combination with DAPT, prevented occlusive thrombosis. Results and Conclusions A ruptured plaque exposes collagen fibers and is often rich in tissue factor, triggering activation of platelets and coagulation. Occlusive thrombi formed on type I collagen in the presence or absence of tissue factor (TF). However, although DAPT prevented occlusive thrombosis in the absence of TF, DAPT had little effect when TF was also present. Under these conditions, PAR antagonism was also ineffective. However, occlusive thrombosis was prevented by combining DAPT with PAR antagonism. These data demonstrate that PAR antagonists may be a useful addition to DAPT in some patients and further demonstrate the utility of in vitro models of occlusive thrombosis.
Collapse
Affiliation(s)
- Jess Berry
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
22
|
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298:101530. [PMID: 34953859 PMCID: PMC8784641 DOI: 10.1016/j.jbc.2021.101530] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Various forms of fibrosis, comprising tissue thickening and scarring, are involved in 40% of deaths across the world. Since the discovery of scarless functional healing in fetuses prior to a certain stage of development, scientists have attempted to replicate scarless wound healing in adults with little success. While the extracellular matrix (ECM), fibroblasts, and inflammatory mediators have been historically investigated as separate branches of biology, it has become increasingly necessary to consider them as parts of a complex and tightly regulated system that becomes dysregulated in fibrosis. With this new paradigm, revisiting fetal scarless wound healing provides a unique opportunity to better understand how this highly regulated system operates mechanistically. In the following review, we navigate the four stages of wound healing (hemostasis, inflammation, repair, and remodeling) against the backdrop of adult versus fetal wound healing, while also exploring the relationships between the ECM, effector cells, and signaling molecules. We conclude by singling out recent findings that offer promising leads to alter the dynamics between the ECM, fibroblasts, and inflammation to promote scarless healing. One factor that promises to be significant is fibroblast heterogeneity and how certain fibroblast subpopulations might be predisposed to scarless healing. Altogether, reconsidering fetal wound healing by examining the interplay of the various factors contributing to fibrosis provides new research directions that will hopefully help us better understand and address fibroproliferative diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic sclerosis, progressive kidney disease, and cardiovascular fibrosis.
Collapse
Affiliation(s)
- Leandro Moretti
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jack Stalfort
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas Harrison Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
23
|
Wang X, Dang Q, Liu C, Chang G, Song H, Xu Q, Ma Y, Li B, Zhang B, Cha D. Antibacterial porous sponge fabricated with capric acid-grafted chitosan and oxidized dextran as a novel hemostatic dressing. Carbohydr Polym 2022; 277:118782. [PMID: 34893218 DOI: 10.1016/j.carbpol.2021.118782] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/22/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
This work aims to fabricate multifunctional hemostatic sponges (C-ODs). Porous C-ODs were first constructed by using capric acid-modified chitosan (CSCA) and oxidized dextrans (ODs) with different oxidation degrees. Batches of experiments showed that (i) CSCA (33.39% of grafting degree), ODs, and C-ODs (100-200 μm in pore size) were synthesized, evidenced by FT-IR, 1H NMR, elemental analysis, hydroxylamine hydrochloride titration, and SEM results; (ii) among C-ODs, C-OD2 had appropriate porosity (85.0%), swelling (20 times its dry weight), absorption, water retention, water vapor transmission, and mechanical properties; (iii) C-OD2 possessed low toxicity (relative cell viability > 86%), low hemolysis rate (0.65%), suitable tissue adhesion (4.74 kPa), and strong antibacterial efficacy (five strains); and (iv) C-OD2's dynamic blood clotting was within 30 s. In three animal injury models, C-OD2's hemostasis time and blood loss were fairly lower than commercial gelatin sponge. Totally, C-OD2 might serve as an ideal hemostatic dressing.
Collapse
Affiliation(s)
- Xiaoyu Wang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Qifeng Dang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Chengsheng Liu
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China.
| | - Guozhu Chang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Hao Song
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Qing Xu
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Yue Ma
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Boyuan Li
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Bonian Zhang
- Qingdao Aorun Biotechnology Co., Ltd., Room 602, Century Mansion, 39 Donghaixi Road, Qingdao 266071, PR China
| | - Dongsu Cha
- The Graduate School of Biotechnology, Korea University, Seoul 136-701, South Korea
| |
Collapse
|
24
|
Abstract
This chapter describes the development of a poly(dimethylsiloxane)-based microfluidic platform that is able to holistically assess and visualize the entire hemostatic process in vitro. The microfluidic platform includes (1) integration of intact endothelium, (2) physiological flow conditions, (3) controlled mechanical injury to study global hemostatic potential of a patient's whole blood samples in a microvascular model and for dissecting pathophysiologic mechanisms of diseases.
Collapse
Affiliation(s)
- Elaissa Trybus Hardy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Yumiko Sakurai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wilbur A Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA. .,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, GA, USA. .,Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
25
|
Xu Y, Yu G, Nie R, Wu Z. Microfluidic systems toward blood hemostasis monitoring and thrombosis diagnosis: From design principles to micro/nano fabrication technologies. VIEW 2022. [DOI: 10.1002/viw.20200183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yi Xu
- Soft Intelligence Lab State Key Laboratory of Digital Manufacturing Equipment and Technology School of Mechanical Science and Engineering Huazhong University of Science and Technology Wuhan China
| | - Guang Yu
- Experimental Medicine Center Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ruqiong Nie
- Department of Cardiology Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou China
| | - Zhigang Wu
- Soft Intelligence Lab State Key Laboratory of Digital Manufacturing Equipment and Technology School of Mechanical Science and Engineering Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
26
|
Zhang Y, Ramasundara SDZ, Preketes-Tardiani RE, Cheng V, Lu H, Ju LA. Emerging Microfluidic Approaches for Platelet Mechanobiology and Interplay With Circulatory Systems. Front Cardiovasc Med 2021; 8:766513. [PMID: 34901226 PMCID: PMC8655735 DOI: 10.3389/fcvm.2021.766513] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/15/2021] [Indexed: 12/29/2022] Open
Abstract
Understanding how platelets can sense and respond to hemodynamic forces in disturbed blood flow and complexed vasculature is crucial to the development of more effective and safer antithrombotic therapeutics. By incorporating diverse structural and functional designs, microfluidic technologies have emerged to mimic microvascular anatomies and hemodynamic microenvironments, which open the floodgates for fascinating platelet mechanobiology investigations. The latest endothelialized microfluidics can even recapitulate the crosstalk between platelets and the circulatory system, including the vessel walls and plasma proteins such as von Willebrand factor. Hereby, we highlight these exciting microfluidic applications to platelet mechanobiology and platelet–circulatory system interplay as implicated in thrombosis. Last but not least, we discuss the need for microfluidic standardization and summarize the commercially available microfluidic platforms for researchers to obtain reproducible and consistent results in the field.
Collapse
Affiliation(s)
- Yingqi Zhang
- 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
| | - Savindi De Zoysa Ramasundara
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia.,Heart Research Institute, Newtown, NSW, Australia.,School of Medicine, The University of Notre Dame Sydney, Darlinghurst, NSW, Australia
| | - Renee Ellen Preketes-Tardiani
- 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
| | - Vivian Cheng
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia
| | - Hongxu Lu
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW, Australia.,Faculty of Science, Institute for Biomedical Materials and Devices, The University of Technology Sydney, Ultimo, 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
|
27
|
Diamond SL, Rossi JM. Point of care whole blood microfluidics for detecting and managing thrombotic and bleeding risks. LAB ON A CHIP 2021; 21:3667-3674. [PMID: 34476426 PMCID: PMC8478847 DOI: 10.1039/d1lc00465d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Point-of-care diagnostics of platelet and coagulation function present demanding challenges. Current clinical diagnostics often use centrifuged plasmas or platelets and frozen plasma standards, recombinant protein standards, or even venoms. Almost all commercialized tests of blood do not recreate the in vivo hemodynamics where platelets accumulate to high densities and thrombin is generated from a procoagulant surface. Despite numerous drugs that target platelets, insufficient coagulation, or excess coagulation, POC blood testing is essentially limited to viscoelastic methods that provide a clotting time, clot strength, and clot lysis, while used mostly in trauma centers with specialized capabilities. Microfluidics now allows small volumes of whole blood (<1 mL) to be tested under venous or arterial shear rates with multi-color readouts to follow platelet function, thrombin generation, fibrin production, and clot stability. Injection molded chips containing pre-patterned fibrillar collagen and lipidated tissue factor can be stored dry for 6 months at 4C, thus allowing rapid blood testing on single-use disposable chips. Using only a small imaging microscope and micropump, these microfluidic devices can detect platelet inhibitors, direct oral anticoagulants (DOACs) and their reversal agents. POC microfluidics are ideal for neonatal surgical applications that involve small blood samples, rapid DOAC testing in stroke or bleeding or emergency surgery situations with patients presenting high risk cofactors for either bleeding or thrombosis.
Collapse
Affiliation(s)
- Scott L Diamond
- Institute for Medicine and Engineering, Department of Chemical and Biomolecular Engineering, University of Pennsylvania, 1024 Vagelos Research Laboratory, Philadelphia, PA 19104, USA.
| | - Jason M Rossi
- Institute for Medicine and Engineering, Department of Chemical and Biomolecular Engineering, University of Pennsylvania, 1024 Vagelos Research Laboratory, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Mathur T, Flanagan JM, Jain A. Tripartite collaboration of blood-derived endothelial cells, next generation RNA sequencing and bioengineered vessel-chip may distinguish vasculopathy and thrombosis among sickle cell disease patients. Bioeng Transl Med 2021; 6:e10211. [PMID: 34589594 PMCID: PMC8459595 DOI: 10.1002/btm2.10211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/24/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Sickle cell disease (SCD) is the most prevalent inherited blood disorder in the world. But the clinical manifestations of the disease are highly variable. In particular, it is currently difficult to predict the adverse outcomes within patients with SCD, such as, vasculopathy, thrombosis, and stroke. Therefore, for most effective and timely interventions, a predictive analytic strategy is desirable. In this study, we evaluate the endothelial and prothrombotic characteristics of blood outgrowth endothelial cells (BOECs) generated from blood samples of SCD patients with known differences in clinical severity of the disease. We present a method to evaluate patient-specific vaso-occlusive risk by combining novel RNA-seq and organ-on-chip approaches. Through differential gene expression (DGE) and pathway analysis we find that BOECs from SCD patients exhibit an activated state through cell adhesion molecule (CAM) and cytokine signaling pathways among many others. In agreement with clinical symptoms of patients, DGE analyses reveal that patient with severe SCD had a greater extent of endothelial activation compared to patient with milder symptoms. This difference is confirmed by performing qRT-PCR of endothelial adhesion markers like E-selectin, P-selectin, tissue factor, and Von Willebrand factor. Finally, the differential regulation of the proinflammatory phenotype is confirmed through platelet adhesion readouts in our BOEC vessel-chip. Taken together, we hypothesize that these easily blood-derived endothelial cells evaluated through RNA-seq and organ-on-chips may serve as a biotechnique to predict vaso-occlusive episodes in SCD patients and will ultimately allow better therapeutic interventions.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Jonathan M. Flanagan
- Department of Pediatrics, Section of Hematology‐OncologyBaylor College of MedicineHoustonTexasUSA
| | - Abhishek Jain
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
- Department of Medical PhysiologyCollege of Medicine, Texas A&M Health Science CenterBryanTexasUSA
| |
Collapse
|
29
|
Abstract
Microengineering advances have enabled the development of perfusable, endothelialized models of the microvasculature that recapitulate the unique biological and biophysical conditions of the microcirculation in vivo. Indeed, at that size scale (<100 μm)-where blood no longer behaves as a simple continuum fluid; blood cells approximate the size of the vessels themselves; and complex interactions among blood cells, plasma molecules, and the endothelium constantly ensue-vascularized microfluidics are ideal tools to investigate these microvascular phenomena. Moreover, perfusable, endothelialized microfluidics offer unique opportunities for investigating microvascular diseases by enabling systematic dissection of both the blood and vascular components of the pathophysiology at hand. We review (a) the state of the art in microvascular devices and (b) the myriad of microvascular diseases and pressing challenges. The engineering community has unique opportunities to innovate with new microvascular devices and to partner with biomedical researchers to usher in a new era of understanding and discovery of microvascular diseases.
Collapse
Affiliation(s)
- David R Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA; ,
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA; ,
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
30
|
Llenas M, Paoli R, Feiner-Gracia N, Albertazzi L, Samitier J, Caballero D. Versatile Vessel-on-a-Chip Platform for Studying Key Features of Blood Vascular Tumors. Bioengineering (Basel) 2021; 8:bioengineering8060081. [PMID: 34207754 PMCID: PMC8226980 DOI: 10.3390/bioengineering8060081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
Tumor vessel-on-a-chip systems have attracted the interest of the cancer research community due to their ability to accurately recapitulate the multiple dynamic events of the metastatic cascade. Vessel-on-a-chip microfluidic platforms have been less utilized for investigating the distinctive features and functional heterogeneities of tumor-derived vascular networks. In particular, vascular tumors are characterized by the massive formation of thrombi and severe bleeding, a rare and life-threatening situation for which there are yet no clear therapeutic guidelines. This is mainly due to the lack of technological platforms capable of reproducing these characteristic traits of the pathology in a simple and well-controlled manner. Herein, we report the fabrication of a versatile tumor vessel-on-a-chip platform to reproduce, investigate, and characterize the massive formation of thrombi and hemorrhage on-chip in a fast and easy manner. Despite its simplicity, this method offers multiple advantages to recapitulate the pathophysiological events of vascular tumors, and therefore, may find useful applications in the field of vascular-related diseases, while at the same time being an alternative to more complex approaches.
Collapse
Affiliation(s)
- Marina Llenas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
| | - Roberto Paoli
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Natalia Feiner-Gracia
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Biomedical Engineering and Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
- Correspondence: (L.A.); (J.S.); (D.C.)
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (L.A.); (J.S.); (D.C.)
| | - David Caballero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (L.A.); (J.S.); (D.C.)
| |
Collapse
|
31
|
Liu Y, Yu K, Shang S, Xie R, Lu F, Bao R, Lan G, Hu E. Chestnut-like macro-acanthosphere triggered hemostasis: a featured mechanism based on puncturing red blood cells. NANOSCALE 2021; 13:9843-9852. [PMID: 34032253 DOI: 10.1039/d1nr01148k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Acute hemorrhage that occurs after trauma is a life-threatening condition. Hence, to halt massive bleeding, there is a critical need to develop a suitable therapy. In this study, we developed self-propelling chestnut-like particles (Pro-MAS) comprising a macro-acanthosphere (MAS) coated with calcium carbonate and protonated tranexamic acid to puncture red blood cells (RBCs) and thus activate hemostasis. In vitro assessments revealed that Pro-MAS was biocompatible, biodegradable, and nontoxic; furthermore, it was capable of puncturing RBCs to release procoagulants and activate platelet aggregation for hemostasis. Animal tests showed that self-propelling Pro-MAS effectively traveled through blood flow to the deep ends of wounds; hemorrhage was controlled within 90 s and 4 min in the injured liver and bleeding femoral artery, respectively. Compared with a commercial hemostat, superior hemostasis was achieved with Pro-MAS, which could be ascribed to its functional and structural features. Overall, traveling Pro-MAS possessed sufficient impact force to puncture RBCs and sufficient momentum to reach the targeted bleeding sites. The present study demonstrated the ability of a novel platform, self-propelling MAS particles, to trigger hemostasis by puncturing RBCs. To the best of our knowledge, this is the first trial in which the release of endogenous procoagulants is promoted without the addition of exogenous procoagulants for severe hemorrhage control.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
von Willebrand factor binding to myosin assists in coagulation. Blood Adv 2021; 4:174-180. [PMID: 31935285 DOI: 10.1182/bloodadvances.2019000533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022] Open
Abstract
von Willebrand factor (VWF) binds to platelets and collagen as a means of facilitating coagulation at sites of injury. Recent evidence has shown that myosin can serve as a surface for thrombin generation and binds to activated factor V and factor X. We studied whether VWF can also bind myosin as a means of bringing factor VIII (FVIII) to sites of clot formation. A myosin-binding assay was developed using skeletal muscle myosin to measure VWF binding, and plasma-derived and recombinant VWF containing molecular disruptions at key VWF sites were tested. Competition assays were performed using anti-VWF antibodies. FVIII binding to myosin was measured using a chromogenic FVIII substrate. Thrombin generation was measured using a fluorogenic substrate with and without myosin. Wild-type recombinant VWF and human plasma VWF from healthy controls bound myosin, whereas plasma lacking VWF exhibited no detectable myosin binding. Binding was multimer dependent and blocked by anti-VWF A1 domain antibodies or A1 domain VWF variants. The specific residues involved in myosin binding were similar, but not identical, to those required for collagen IV binding. FVIII did not bind myosin directly, but FVIII activity was detected when VWF and FVIII were bound to myosin. Myosin enhanced thrombin generation in platelet-poor plasma, although no difference was detected with the addition of myosin to platelet-rich plasma. Myosin may help to facilitate delivery of FVIII to sites of injury and indirectly accelerate thrombin generation by providing a surface for VWF binding in the setting of trauma and myosin exposure.
Collapse
|
33
|
Lim J, Ching H, Yoon JK, Jeon NL, Kim Y. Microvascularized tumor organoids-on-chips: advancing preclinical drug screening with pathophysiological relevance. NANO CONVERGENCE 2021; 8:12. [PMID: 33846849 PMCID: PMC8042002 DOI: 10.1186/s40580-021-00261-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/17/2021] [Indexed: 05/06/2023]
Abstract
Recent developments of organoids engineering and organ-on-a-chip microfluidic technologies have enabled the recapitulation of the major functions and architectures of microscale human tissue, including tumor pathophysiology. Nevertheless, there remain challenges in recapitulating the complexity and heterogeneity of tumor microenvironment. The integration of these engineering technologies suggests a potential strategy to overcome the limitations in reconstituting the perfusable microvascular system of large-scale tumors conserving their key functional features. Here, we review the recent progress of in vitro tumor-on-a-chip microfluidic technologies, focusing on the reconstruction of microvascularized organoid models to suggest a better platform for personalized cancer medicine.
Collapse
Affiliation(s)
- Jungeun Lim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
| | - Hanna Ching
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeong-Kee Yoon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Noo Li Jeon
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - YongTae Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
34
|
Poventud-Fuentes I, Kwon KW, Seo J, Tomaiuolo M, Stalker TJ, Brass LF, Huh D. A Human Vascular Injury-on-a-Chip Model of Hemostasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2004889. [PMID: 33150735 PMCID: PMC8049960 DOI: 10.1002/smll.202004889] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/28/2020] [Indexed: 05/02/2023]
Abstract
Hemostasis is an innate protective mechanism that plays a central role in maintaining the homeostasis of the vascular system during vascular injury. Studying this essential physiological process is often challenged by the difficulty of modeling and probing the complex dynamics of hemostatic responses in the native context of human blood vessels. To address this major challenge, this paper describes a microengineering approach for in vitro modeling of hemostasis. This microphysiological model replicates the living endothelium, multilayered microarchitecture, and procoagulant activity of human blood vessels, and is also equipped with a microneedle that is actuated with spatial precision to simulate penetrating vascular injuries. The system recapitulates key features of the hemostatic response to acute vascular injury as observed in vivo, including i) thrombin-driven accumulation of platelets and fibrin, ii) formation of a platelet- and fibrin-rich hemostatic plug that halts blood loss, and iii) matrix deformation driven by platelet contraction for wound closure. Moreover, the potential use of this model for drug testing applications is demonstrated by evaluating the effects of anticoagulants and antiplatelet agents that are in current clinical use. The vascular injury-on-a-chip may serve as an enabling platform for preclinical investigation of hematological disorders and emerging therapeutic approaches against them.
Collapse
Affiliation(s)
| | - Keon Woo Kwon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maurizio Tomaiuolo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Timothy J Stalker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lawrence F Brass
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
35
|
Caruso C, Lam WA. Point-of-Care Diagnostic Assays and Novel Preclinical Technologies for Hemostasis and Thrombosis. Semin Thromb Hemost 2021; 47:120-128. [PMID: 33636744 DOI: 10.1055/s-0041-1723798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hemostasis is a complex wound-healing process involving numerous mechanical and biochemical mechanisms and influenced by many factors including platelets, coagulation factors, and endothelial components. Slight alterations in these mechanisms can lead to either prothrombotic or bleeding consequences, and such hemostatic imbalances can lead to significant clinical consequences with resultant morbidity and mortality. An ideal hemostasis assay would not only address all the unique processes involved in clot formation and resolution but also take place under flow conditions to account for endothelial involvement. Global assays do exist; however, these assays are not flow based. Flow-based assays have been limited secondary to their large blood volume requirements and low throughput, limiting potential clinical applications. Microfluidic-based assays address the aforementioned limitations of both global and flow-based assays by utilizing standardized devices that require low blood volumes, offer reproducible analysis, and have functionality under a range of shear stresses and flow conditions. While still largely confined to the preclinical space, here we aim to discuss these novel technologies and potential clinical implications, particularly in comparison to the current, commercially available point-of-care assays.
Collapse
Affiliation(s)
- Christina Caruso
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Wilbur A Lam
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia.,Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
36
|
Mangin PH, Neeves KB, Lam WA, Cosemans JMEM, Korin N, Kerrigan SW, Panteleev MA. In vitro flow-based assay: From simple toward more sophisticated models for mimicking hemostasis and thrombosis. J Thromb Haemost 2021; 19:582-587. [PMID: 34396675 DOI: 10.1111/jth.15143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
In vitro flow-based assays are widely used to investigate the role of platelets and coagulation in hemostasis and thrombosis. Their main advantage over other assays relies on the fact that they integrate blood flow that regulates many aspects of platelet function, including adhesion, activation, and aggregation. Blood flow is also central in the regulation of coagulation through its ability to modulate the local concentrations of coagulation factors within and around thrombi. The most broadly used assay to study thrombus formation consists in perfusing whole blood over immobilized fibrillar collagen through a single channel, which helps to reproduce thrombus formation as it occurs in vivo after vascular injury, with platelets adhering, becoming activated, and forming a mural thrombus. This process can also be studied under conditions of thrombin generation, notably by recalcifying blood collected in sodium citrate. In this manuscript, we briefly discuss the advantages and limits of this broadly used "in vitro thrombus formation model." The main emphasis is on the description of the most recent developments regarding design of new flow models and new techniques, and how these may advance the landscape of in vitro studies into the formation of physiological or pathophysiological thrombi. Challenges linked to mimicking the formation of a hemostatic plug in a healthy vessel or a thrombus in diseased arteries and the complexity of reproducing the various aspects of venous thrombosis are discussed. Future directions are proposed to improve the physiological or pathophysiological relevance of current flow-based assays.
Collapse
Affiliation(s)
- Pierre H Mangin
- INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Université de Strasbourg, Strasbourg, France
| | - Keith B Neeves
- Departments of Bioengineering and Pediatrics, Hemophilia and Thrombosis Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Wilbur A Lam
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, GA, USA
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Netanel Korin
- Department of Biomedical Engineering Technion, Israel Institute of Technology, Haifa, Israel
| | - Steven W Kerrigan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| |
Collapse
|
37
|
Thromboinflammation Model-on-A-Chip by Whole Blood Microfluidics on Fixed Human Endothelium. Diagnostics (Basel) 2021; 11:diagnostics11020203. [PMID: 33573079 PMCID: PMC7911484 DOI: 10.3390/diagnostics11020203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/23/2020] [Accepted: 01/26/2021] [Indexed: 01/15/2023] Open
Abstract
Microfluidic devices have an established role in the study of platelets and coagulation factors in thrombosis, with potential diagnostic applications. However, few microfluidic devices have assessed the contribution of neutrophils to thrombus formation, despite increasing knowledge of neutrophils’ importance in cardiovascular thrombosis. We describe a thromboinflammation model which uses straight channels, lined with fixed human umbilical vein endothelial cells, after treatment with tumour necrosis factor-alpha. Re-calcified whole blood is perfused over the endothelium at venous and arterial shear rate. Neutrophil adhesion, platelet and fibrin thrombus formation, is measured over time by the addition of fluorescent antibodies to a whole blood sample. Fixed endothelium retains surface expression of adhesion molecules ICAM-1 and E-Selectin. Neutrophils adhere preferentially to platelet thrombi on the endothelium. Inhibitors of neutrophil adhesion and anti-inflammatory agents, such as isoquercetin, decrease neutrophil adhesion. Our model offers the advantage of the use of (1) fixed endothelium, (2) whole blood, instead of isolated neutrophils, and (3) a small amount of blood (1 mL). The characteristics of this thromboinflammation model provide the potential for further development for drug screening and point-of-care applications.
Collapse
|
38
|
Buchegger B, Tanzer A, Posch S, Gabriel C, Klar TA, Jacak J. STED lithography in microfluidics for 3D thrombocyte aggregation testing. J Nanobiotechnology 2021; 19:23. [PMID: 33461577 PMCID: PMC7814651 DOI: 10.1186/s12951-020-00762-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 12/24/2020] [Indexed: 11/10/2022] Open
Abstract
Three-dimensional photopolymerization techniques such as multiphoton polymerization lithography (MPL) and stimulated emission depletion (STED) lithography are powerful tools for fabricating structures in the sub-µm range. Combining these techniques with microfluidics enables us to broaden the range of their applications. In this study, we show a microfluidic device enhanced with MPL structures carrying STED-lithographically written nanoanchors that promote binding of the von Willebrand factor (vWF). The density of vWF is adjusted by varying the number of the nanoanchors on the 3D structures. This allows us to study the impact of the density of vWF on the activation of thrombocytes. The activation of the thrombocytes seems to decrease with the density of vWF on the 3D scaffolds inside the microfluidic channels.
Collapse
Affiliation(s)
- Bianca Buchegger
- Institute of Applied Physics and Linz Institute of Technology (LIT), Johannes Kepler University Linz, Altenberger Straße 69, 4040, Linz, Austria.,University of Applied Sciences, Upper Austria School of Medical Engineering and Applied Social Sciences, Garnisonstraße 21, 4020, Linz, Austria
| | - Alexander Tanzer
- Institute of Applied Physics and Linz Institute of Technology (LIT), Johannes Kepler University Linz, Altenberger Straße 69, 4040, Linz, Austria
| | - Sandra Posch
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstraße 40, 4020, Linz, Austria
| | - Christian Gabriel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1200, Vienna, Austria
| | - Thomas A Klar
- Institute of Applied Physics and Linz Institute of Technology (LIT), Johannes Kepler University Linz, Altenberger Straße 69, 4040, Linz, Austria
| | - Jaroslaw Jacak
- University of Applied Sciences, Upper Austria School of Medical Engineering and Applied Social Sciences, Garnisonstraße 21, 4020, Linz, Austria.
| |
Collapse
|
39
|
Decuzzi P, Peer D, Di Mascolo D, Palange AL, Manghnani PN, Moghimi SM, Farhangrazi ZS, Howard KA, Rosenblum D, Liang T, Chen Z, Wang Z, Zhu JJ, Gu Z, Korin N, Letourneur D, Chauvierre C, van der Meel R, Kiessling F, Lammers T. Roadmap on nanomedicine. NANOTECHNOLOGY 2021; 32:012001. [PMID: 33043901 PMCID: PMC7612035 DOI: 10.1088/1361-6528/abaadb] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Since the launch of the Alliance for Nanotechnology in Cancer by the National Cancer Institute in late 2004, several similar initiatives have been promoted all over the globe with the intention of advancing the diagnosis, treatment and prevention of cancer in the wake of nanoscience and nanotechnology. All this has encouraged scientists with diverse backgrounds to team up with one another, learn from each other, and generate new knowledge at the interface between engineering, physics, chemistry and biomedical sciences. Importantly, this new knowledge has been wisely channeled towards the development of novel diagnostic, imaging and therapeutic nanosystems, many of which are currently at different stages of clinical development. This roadmap collects eight brief articles elaborating on the interaction of nanomedicines with human biology; the biomedical and clinical applications of nanomedicines; and the importance of patient stratification in the development of future nanomedicines. The first article reports on the role of geometry and mechanical properties in nanomedicine rational design; the second articulates on the interaction of nanomedicines with cells of the immune system; and the third deals with exploiting endogenous molecules, such as albumin, to carry therapeutic agents. The second group of articles highlights the successful application of nanomedicines in the treatment of cancer with the optimal delivery of nucleic acids, diabetes with the sustained and controlled release of insulin, stroke by using thrombolytic particles, and atherosclerosis with the development of targeted nanoparticles. Finally, the last contribution comments on how nanomedicine and theranostics could play a pivotal role in the development of personalized medicines. As this roadmap cannot cover the massive extent of development of nanomedicine over the past 15 years, only a few major achievements are highlighted as the field progressively matures from the initial hype to the consolidation phase.
Collapse
Affiliation(s)
- Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
- Corresponding authors: and
| | - Dan Peer
- Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering
- Center for Nanoscience and Nanotechnology
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 6997801, Israel
- Corresponding authors: and
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Purnima Naresh Manghnani
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - S. Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | | | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center, Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Daniel Rosenblum
- Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering
- Center for Nanoscience and Nanotechnology
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tingxizi Liang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhaowei Chen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Netanel Korin
- Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Didier Letourneur
- Université de Paris, Université Paris 13, INSERM 1148, LVTS, Hôpital Bichat, F-75018 Paris, France
| | - Cédric Chauvierre
- Université de Paris, Université Paris 13, INSERM 1148, LVTS, Hôpital Bichat, F-75018 Paris, France
| | - Roy van der Meel
- Laboratory of Chemical Biology, Dept. of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Dept. of Targeted Therapeutics, University of Twente, Enschede, The Netherlands
- Dept. of Pharmaceutics, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
40
|
Kholmukhamedov A. Procoagulant Platelets. Platelets 2020. [DOI: 10.5772/intechopen.92638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There are two well-known subpopulations of activated platelets: pro-aggregatory and procoagulant. Procoagulant platelets represent a subpopulation of activated platelets, which are morphologically and functionally distinct from pro-aggregatory ones. Although various names have been used to describe these platelets in the literature (CoaT, CoaTed, highly activated, ballooned, capped, etc.), there is a consensus on their phenotypic features including exposure of high levels of phosphatidylserine (PSer) on the surface; decreased aggregatory and adhesive properties; support of active tenase and prothrombinase complexes; maximal generation by co-stimulation of glycoprotein VI (GPVI) and protease-activated receptors (PAR). In this chapter, morphologic and functional features of procoagulant platelets, as well as the mechanisms of their formation, will be discussed.
Collapse
|
41
|
Endothelial damage and a thin intercellular fibrin network promote haemorrhage in acute promyelocytic leukaemia. EBioMedicine 2020; 60:102992. [PMID: 32949998 PMCID: PMC7501057 DOI: 10.1016/j.ebiom.2020.102992] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/16/2020] [Accepted: 08/24/2020] [Indexed: 01/10/2023] Open
Abstract
Background The role of vascular endothelium in acute promyelocytic leukaemia (APL) remains unknown. We aimed to investigate the mechanisms by which APL cells interact with endothelial cells (ECs) and to further explore how the endothelium affects bleeding as well as therapeutic interventions. Method APL cells and an original APL cell line, NB4 cells, were used for experiments. The effects of leukaemic cells on ECs were analyzed in vitro and in vivo. Moreover, the endothelial barrier function and procoagulant activity were detected. An APL mouse model was established for in vivo studies. Findings APL cells interacted with ECs via ICAM-1 and VCAM-1 receptors to disrupt endothelial integrity. This binding activated MLCK signaling, resulting in the trans-endothelial passage of protein and red blood cells (RBCs). Combined treatment with asiatic acid or anti-adhesion receptor antibody inhibited the response of ECs to APL cells, thereby preventing APL-associated haemorrhage in vitro and in vivo. Activated ECs exhibited a procoagulant phenotype after phosphatidylserine exposure. Plasma from APL patients formed a thin fibrin network between procoagulant ECs, and this intercellular fibrin decreased the passage of albumin and RBCs. Ex vivo addition of fibrinogen further enhanced this barrier function in a dose-dependent manner. Interpretation Endothelial damage induced by leukaemic cell adherence promotes haemorrhaging in APL. Stabilization of ECs, decreasing adhesion receptor expression, and increasing fibrinogen transfusion levels may be a new therapeutic avenue to alleviate this fatal bleeding complication. Funding National Science Foundation of China (81670128, 81873433).
Collapse
|
42
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
43
|
Deal HE, Brown AC, Daniele MA. Microphysiological systems for the modeling of wound healing and evaluation of pro-healing therapies. J Mater Chem B 2020; 8:7062-7075. [PMID: 32756718 PMCID: PMC7460719 DOI: 10.1039/d0tb00544d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Wound healing is a multivariate process involving the coordinated response of numerous proteins and cell types. Accordingly, biomedical research has seen an increased adoption of the use of in vitro wound healing assays with complexity beyond that offered by traditional well-plate constructs. These microphysiological systems (MPS) seek to recapitulate one or more physiological features of the in vivo microenvironment, while retaining the analytical capacity of more reductionist assays. Design efforts to achieve relevant wound healing physiology include the use of dynamic perfusion over static culture, the incorporation of multiple cell types, the arrangement of cells in three dimensions, the addition of biomechanically and biochemically relevant hydrogels, and combinations thereof. This review provides a brief overview of the wound healing process and in vivo assays, and we critically review the current state of MPS and supporting technologies for modelling and studying wound healing. We distinguish between MPS that seek to inform a particular phase of wound healing, and constructs that have the potential to inform multiple phases of wound healing. This distinction is a product of whether analysis of a particular process is prioritized, or a particular physiology is prioritized, during design. Material selection is emphasized throughout, and relevant fabrication techniques discussed.
Collapse
Affiliation(s)
- Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA and Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA
| |
Collapse
|
44
|
Lakshmanan HHS, Pore AA, Kohs TCL, Yazar F, Thompson RM, Jurney PL, Maddala J, Olson SR, Shatzel JJ, Vanapalli SA, McCarty OJT. Design of a Microfluidic Bleeding Chip to Evaluate Antithrombotic Agents for Use in COVID-19 Patients. Cell Mol Bioeng 2020; 13:331-339. [PMID: 32837586 PMCID: PMC7408976 DOI: 10.1007/s12195-020-00644-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Interventions that could prevent thrombosis, clinical decompensation, and respiratory compromise in patients with novel coronavirus disease (COVID-19) are key to decrease mortality rate. Studies show that profound cytokine release and excessive activation of blood coagulation appear to be key drivers of COVID-19 associated mortality. Since limited in vitro methods exist for assessing the effects of anticoagulants on hemostasis, the development of novel therapies to safely prevent thrombosis in COVID-19 patients relies on preclinical animal models and early phase human trials. Herein we present the design of a microfluidic “bleeding chip” to evaluate the effects of antithrombotic therapies on hemostatic plug formation in vitro. Methods The design of the microfluidic device consists of two orthogonal channels: an inlet that serves as a model blood vessel, and a bleeding channel to model hemostatic plug formation at sites of compromised endothelial barrier function. This is achieved by placing a series of 3 pillars spaced 10 μm apart at the intersection of the two channels. The pillars and bleeding channel are coated with the extracellular matrix protein collagen. Results Perfusion of human whole blood through the microfluidic bleeding chip led to initial platelet adhesion and aggregation at the pillars followed by hemostatic plug formation and occlusion of the bleeding channel. Conclusions Safe and effective mitigating agents are needed for treatment and prevention of thrombotic complications in COVID-19 patients. This simple microfluidic device holds potential to be developed into a tool for assessing the effects of anticoagulant therapy on hemostasis.
Collapse
Affiliation(s)
- Hari Hara Sudhan Lakshmanan
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Adity A Pore
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX USA
| | - Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Feyza Yazar
- Department of Biomedical Engineering, San José State University, San Jose, CA USA
| | - Rachel M Thompson
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Patrick L Jurney
- Department of Biomedical Engineering, San José State University, San Jose, CA USA
| | - Jeevan Maddala
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Sven R Olson
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| |
Collapse
|
45
|
Hu X, Li Y, Li J, Chen H. Effects of altered blood flow induced by the muscle pump on thrombosis in a microfluidic venous valve model. LAB ON A CHIP 2020; 20:2473-2481. [PMID: 32543635 DOI: 10.1039/d0lc00287a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Deep vein thrombosis (DVT) often occurs in the lower limb veins of bedridden patients and greatly reduces the quality of life. The altered blood flow in venous valves induced by the insufficient efficacy of the muscle pump is commonly considered as a main factor. However, it is still a great challenge to observe the altered blood flow in real time, and its role in the formation of thrombi is poorly understood. Here we make a microfluidic venous valve model with flexible leaflets in a deformable channel that can mimic the motion of valves and the compression of vessels by muscle contraction, and identify the stasis and intermittent reflux in the valve pocket generated by the muscle pump. A thrombus forms in the stasis flow, while the intermittent reflux removes the fibrin and inhibits the growth of the thrombus. A flexible microfluidic device that can mimic the motion of valves and the contraction of vessels would have wide applications in the research on cardiovascular diseases.
Collapse
Affiliation(s)
- Xiangyu Hu
- State Key Laboratory of Tribology, Mechanical Engineering Department, Tsinghua University, Beijing, 100084, China.
| | | | | | | |
Collapse
|
46
|
Oshinowo O, Lambert T, Sakurai Y, Copeland R, Hansen CE, Lam WA, Myers DR. Getting a good view: in vitro imaging of platelets under flow. Platelets 2020; 31:570-579. [PMID: 32106734 PMCID: PMC7332395 DOI: 10.1080/09537104.2020.1732320] [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: 11/01/2019] [Revised: 01/06/2020] [Accepted: 02/12/2020] [Indexed: 01/19/2023]
Abstract
As the anucleate cells responsible for hemostasis and thrombosis, platelets are exposed to a myriad of biophysical and biochemical stimuli within vasculature and heterogeneous blood clots. Highly controlled, reductionist in vitro imaging studies have been instrumental in providing a detailed and quantitative understanding of platelet biology and behavior, and have helped elucidate some surprising functions of platelets. In this review, we highlight the tools and approaches that enable visualization of platelets in conjunction with precise control over the local biofluidic and biochemical microenvironment. We also discuss next generation tools that add further control over microenvironment cell stiffness or enable visualization of the interactions between platelets and endothelial cells. Throughout the review, we include pragmatic knowledge on imaging systems, experimental conditions, and approaches that have proved to be useful to our in vitro imaging studies of platelets under flow.
Collapse
Affiliation(s)
- Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - Tamara Lambert
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - Yumiko Sakurai
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - Renee Copeland
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - Caroline E. Hansen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - Wilbur A. Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| | - David R. Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Children’s Healthcare of Atlanta, Emory University School of Medicine, Emory University, Atlanta, Georgia
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
47
|
Brouns SL, Provenzale I, van Geffen JP, van der Meijden PE, Heemskerk JW. Localized endothelial-based control of platelet aggregation and coagulation under flow: A proof-of-principle vessel-on-a-chip study. J Thromb Haemost 2020; 18:931-941. [PMID: 31863548 PMCID: PMC7187151 DOI: 10.1111/jth.14719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND In the intact vessel wall, endothelial cells form a barrier between the blood and the remaining vascular structures, serving to maintain blood fluidity and preventing platelet activation and fibrin clot formation. The spatiotemporal space of this inhibition is largely unknown. OBJECTIVE To assess the local inhibitory roles of a discontinuous endothelium, we developed a vessel-on-a-chip model, consisting of a microfluidic chamber coated with the thrombogenic collagen and tissue factor (TF), and covered with patches of human endothelial cells. By flow perfusion of human blood and plasma, the heterogeneous formation of platelet aggregates and fibrin clots was monitored by multicolor fluorescence microscopy. RESULTS On collagen/TF coatings, a coverage of 40% to 60% of human umbilical vein endothelial cells resulted in a strong overall delay in platelet deposition and fibrin fiber formation under flow. Fibrin formation colocalized with the deposited platelets, and was restricted to regions in between endothelial cells, thus pointing to immediate local suppression of the clotting process. Fibrin kinetics were enhanced by treatment of the cells with heparinase III, partially disrupting the glycocalyx, and to a lesser degree by antagonism of the endothelial thrombomodulin. Co-coating of purified thrombomodulin and collagen had a similar coagulation-suppressing effect as endothelial thrombomodulin. CONCLUSIONS In this vessel-on-a-chip system with patches of endothelial cells on thrombogenic surfaces, the coagulant activity under flow is regulated by: (a) the residual exposure of trigger (collagen/TF), (b) the endothelial glycocalyx, and (c) to a lesser degree the endothelial thrombomodulin.
Collapse
Affiliation(s)
- Sanne L.N. Brouns
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Isabella Provenzale
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Johanna P. van Geffen
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Paola E.J. van der Meijden
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Johan W.M. Heemskerk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
48
|
Puff pastry-like chitosan/konjac glucomannan matrix with thrombin-occupied microporous starch particles as a composite for hemostasis. Carbohydr Polym 2020; 232:115814. [DOI: 10.1016/j.carbpol.2019.115814] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/24/2019] [Accepted: 12/29/2019] [Indexed: 11/21/2022]
|
49
|
The effect of platelet storage temperature on haemostatic, immune, and endothelial function: potential for personalised medicine. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2020; 17:321-330. [PMID: 31385802 DOI: 10.2450/2019.0095-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Reports from both adult and paediatric populations indicate that approximately two-thirds of platelet transfusions are used prophylactically to prevent bleeding, while the remaining one-third are used therapeutically to manage active bleeding. These two indications, prophylactic and therapeutic, serve two very distinct purposes and therefore will have two different functional requirements. In addition, disease aetiology in a given patient may require platelets with different functional characteristics. These characteristics can be derived from the various manufacturing methods used in platelet product production, including collection methods, processing methods, and storage options. The iterative combinations of manufacturing methods can result in a number of unique platelet products with different efficacy and safety profiles, which could potentially be used to benefit patient populations by meeting diverse clinical needs. In particular, cold storage of platelet products causes many biochemical and functional changes, of which the most notable characterised to date include increased haemostatic activity and altered expression of molecules inherent to platelet:leucocyte interactions. The in vivo consequences, both short- and long-term, of these molecular and cellular cold-storage-induced changes have yet to be clearly defined. Elucidation of these mechanisms would potentially reveal unique biologies that could be harnessed to provide more targeted therapies. To this end, in this new era of personalised medicine, perhaps there is an opportunity to provide individual patients with platelet products that are tailored to their clinical condition and the specific indication for transfusion.
Collapse
|
50
|
Yu X, Panckeri KA, Ivanciu L, Camire RM, Coxon CH, Cuker A, Diamond SL. Microfluidic hemophilia models using blood from healthy donors. Res Pract Thromb Haemost 2020; 4:54-63. [PMID: 31989085 PMCID: PMC6971334 DOI: 10.1002/rth2.12286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Microfluidic clotting assays permit drug action studies for hemophilia therapeutics under flow. However, limited availability of patient samples and Inter-donor variability limit the application of such assays, especially with many patients on prophylaxis. OBJECTIVE To develop approaches to phenocopy hemophilia using modified healthy blood in microfluidic assays. METHODS Corn trypsin inhibitor (4 µg/mL)-treated healthy blood was dosed with either anti-factor VIII (FVIII; hemophilia A model) or a recombinant factor IX (FIX) missense variant (FIX-V181T; hemophilia B model). Treated blood was perfused at 100 s-1 wall shear rate over collagen/tissue factor (TF) or collagen/factor XIa (FXIa). RESULTS Anti-FVIII partially blocked fibrin production on collagen/TF, but completely blocked fibrin production on collagen/FXIa, a phenotype reversed with 1 µmol/L bispecific antibody (emicizumab), which binds FIXa and factor X. As expected, emicizumab had no significant effect on healthy blood (no anti-FVIII present) perfused over collagen/FXIa. The efficacy of emicizumab in anti-FVIII-treated healthy blood phenocopied the action of emicizumab in the blood of a patient with hemophilia A perfused over collagen/FXIa. Interestingly, a patient-derived FVIII-neutralizing antibody reduced fibrin production when added to healthy blood perfused over collagen/FXIa. For low TF surfaces, reFIX-V181T (50 µg/mL) fully blocked platelet and fibrin deposition, a phenotype fully reversed with anti-TFPI. CONCLUSION Two new microfluidic hemophilia A and B models demonstrate the potency of anti-TF pathway inhibitor, emicizumab, and a patient-derived inhibitory antibody. Using collagen/FXIa-coated surfaces resulted in reliable and highly sensitive hemophilia models.
Collapse
Affiliation(s)
- Xinren Yu
- Department of Chemical and Biomolecular EngineeringInstitute for Medicine and EngineeringUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Karen A. Panckeri
- Penn Comprehensive Hemophilia and Thrombosis ProgramHospital of the University of PennsylvaniaPhiladelphiaPennsylvania
| | - Lacramioara Ivanciu
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
- Division of HematologyDepartment of PediatricsPerelman School of MedicineThe University of PennsylvaniaPhiladelphiaPennsylvania
| | - Rodney M. Camire
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
- Division of HematologyDepartment of PediatricsPerelman School of MedicineThe University of PennsylvaniaPhiladelphiaPennsylvania
| | - Carmen H. Coxon
- National Institute for Biological Standards and ControlPotters BarUK
| | - Adam Cuker
- Penn Comprehensive Hemophilia and Thrombosis ProgramHospital of the University of PennsylvaniaPhiladelphiaPennsylvania
| | - Scott L. Diamond
- Department of Chemical and Biomolecular EngineeringInstitute for Medicine and EngineeringUniversity of PennsylvaniaPhiladelphiaPennsylvania
| |
Collapse
|