1
|
Fardian-Melamed N, Skripka A, Ursprung B, Lee C, Darlington TP, Teitelboim A, Qi X, Wang M, Gerton JM, Cohen BE, Chan EM, Schuck PJ. Infrared nanosensors of piconewton to micronewton forces. Nature 2025; 637:70-75. [PMID: 39743607 DOI: 10.1038/s41586-024-08221-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 10/15/2024] [Indexed: 01/04/2025]
Abstract
Mechanical force is an essential feature for many physical and biological processes1-7, and remote measurement of mechanical signals with high sensitivity and spatial resolution is needed for diverse applications, including robotics8, biophysics9,10, energy storage11 and medicine12,13. Nanoscale luminescent force sensors excel at measuring piconewton forces, whereas larger sensors have proven powerful in probing micronewton forces14-16. However, large gaps remain in the force magnitudes that can be probed remotely from subsurface or interfacial sites, and no individual, non-invasive sensor is capable of measuring over the large dynamic range needed to understand many systems14,17. Here we demonstrate Tm3+-doped avalanching-nanoparticle18 force sensors that can be addressed remotely by deeply penetrating near-infrared light and can detect piconewton to micronewton forces with a dynamic range spanning more than four orders of magnitude. Using atomic force microscopy coupled with single-nanoparticle optical spectroscopy, we characterize the mechanical sensitivity of the photon-avalanching process and reveal its exceptional force responsiveness. By manipulating the Tm3+ concentrations and energy transfer within the nanosensors, we demonstrate different optical force-sensing modalities, including mechanobrightening and mechanochromism. The adaptability of these nanoscale optical force sensors, along with their multiscale-sensing capability, enable operation in the dynamic and versatile environments present in real-world, complex structures spanning biological organisms to nanoelectromechanical systems.
Collapse
Affiliation(s)
| | - Artiom Skripka
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Nanomaterials for Bioimaging Group, Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autόnoma de Madrid, Madrid, Spain
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Benedikt Ursprung
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Changhwan Lee
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Thomas P Darlington
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Ayelet Teitelboim
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Xiao Qi
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Maoji Wang
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Jordan M Gerton
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Bruce E Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Emory M Chan
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - P James Schuck
- Department of Mechanical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Wang S, Wang Z, Shen Z, Zhang M, Jin D, Zheng K, Liu X, Chai M, Wang Z, Chi A, Ostrovidov S, Wu H, Shao D, Liu G, Wu K, Leong KW, Shi X. Magnetic soft microrobots for erectile dysfunction therapy. Proc Natl Acad Sci U S A 2024; 121:e2407809121. [PMID: 39556757 PMCID: PMC11626158 DOI: 10.1073/pnas.2407809121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024] Open
Abstract
Erectile dysfunction (ED) is a major threat to male fertility and quality of life, and mesenchymal stromal cells (MSCs) are a promising therapeutic option. However, therapeutic outcomes are compromised by low MSC retention and survival rates in corpus cavernosum tissue. Here, we developed an innovative magnetic soft microrobot comprising an ultrasoft hydrogel microsphere embedded with a magnetic nanoparticle chain for MSC delivery. This design also features phenylboronic acid groups for scavenging reactive oxygen species (ROS). With a Young's modulus of less than 1 kPa, the ultrasoft microrobot adapts its shape within narrow blood vessels, ensuring a uniform distribution of MSCs within the corpus cavernosum. Our findings showed that compared with traditional MSC injections, the MSC delivery microrobot (MSC-Rob) significantly enhanced MSC retention and survival. In both rat and beagle ED models, MSC-Rob treatment accelerated the repair of corpus cavernosum tissue and restored erectile function. Single-cell RNA sequencing (scRNA-seq) revealed that MSC-Rob treatment facilitates nerve and blood vessel regeneration in the corpus cavernosum by increasing the presence of regenerative macrophages. Overall, our MSC-Rob not only advances the clinical application of MSCs for ED therapy but also broadens the scope of microrobots for other cell therapies.
Collapse
Affiliation(s)
- Shuting Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou510640, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou510006, P. R. China
| | - Zhenqing Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, P. R. China
| | - Zongshan Shen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou510055, P. R. China
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, P. R. China
| | - Dongdong Jin
- School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen518071, P. R. China
| | - Ke Zheng
- School of Materials Science and Engineering, Dongguan University of Technology, Dongguan523808, P. R. China
| | - Xuemin Liu
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Muyuan Chai
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Zhenxing Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Ani Chi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Serge Ostrovidov
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku113-8510, Tokyo, Japan
| | - Hongkai Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong999077, P. R. China
| | - Dan Shao
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou511442, P. R. China
- Department of Biomedical Engineering Columbia University, New York, NY10032
| | - Guihua Liu
- Reproductive Centre The Sixth Affiliated Hospital Sun Yat-sen University, Guangzhou510655, P. R. China
| | - Kai Wu
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Kam W. Leong
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou510640, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou510006, P. R. China
| |
Collapse
|
3
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
4
|
Islam MM, Gaska I, Oshinowo O, Otumala A, Shekhar S, Au Yong N, Myers DR. Single-pericyte nanomechanics measured by contraction cytometry. APL Bioeng 2024; 8:036109. [PMID: 39131206 PMCID: PMC11316606 DOI: 10.1063/5.0213761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Pericytes line the microvasculature throughout the body and play a key role in regulating blood flow by constricting and dilating vessels. However, the biophysical mechanisms through which pericytes transduce microenvironmental chemical and mechanical cues to mediate vessel diameter, thereby impacting oxygen and nutrient delivery, remain largely unknown. This knowledge gap is clinically relevant as numerous diseases are associated with the aberrant contraction of pericytes, which are unusually susceptible to injury. Here, we report the development of a high-throughput hydrogel-based pericyte contraction cytometer that quantifies single-cell contraction forces from murine and human pericytes in different microvascular microenvironments and in the presence of competing vasoconstricting and vasodilating stimuli. We further show that murine pericyte survival in hypoxia is mediated by the mechanical microenvironment and that, paradoxically, pre-treating pericytes to reduce contraction increases hypoxic cell death. Moreover, using the contraction cytometer as a drug-screening tool, we found that cofilin-1 could be applied extracellularly to release murine pericytes from hypoxia-induced contractile rigor mortis and, therefore, may represent a novel approach for mitigating the long-lasting decrease in blood flow that occurs after hypoxic injury.
Collapse
Affiliation(s)
| | - Ignas Gaska
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | | | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | - David R. Myers
- Author to whom correspondence should be addressed:. Tel.: 404-727-0401
| |
Collapse
|
5
|
Leartprapun N, Zeng Z, Hajjarian Z, Bossuyt V, Nadkarni SK. Laser speckle rheological microscopy reveals wideband viscoelastic spectra of biological tissues. SCIENCE ADVANCES 2024; 10:eadl1586. [PMID: 38718128 PMCID: PMC11078189 DOI: 10.1126/sciadv.adl1586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Viscoelastic transformation of tissue drives aberrant cellular functions and is an early biomarker of disease pathogenesis. Tissues scale a range of viscoelastic moduli, from biofluids to bone. Moreover, viscoelastic behavior is governed by the frequency at which tissue is probed, yielding distinct viscous and elastic responses modulated over a wide frequency band. Existing tools do not quantify wideband viscoelastic spectra in tissues, leaving a vast knowledge gap. We present wideband laser speckle rheological microscopy (WB-SHEAR) that reveals elastic and viscous response over sub-megahertz frequencies previously not investigated in tissue. WB-SHEAR uses an optical, noncontact approach to quantify wideband viscoelastic spectra in specimens spanning a range of moduli from low-viscosity fibrin to highly elastic bone. Via laser scanning, micromechanical imaging is enabled to access wideband viscoelastic spectra in heterogeneous tumor specimens with high spatial resolution (25 micrometers). The ability to interrogate the viscoelastic landscape of diverse biospecimens could transform our understanding of mechanobiological processes in various diseases.
Collapse
Affiliation(s)
- Nichaluk Leartprapun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ziqian Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Zeinab Hajjarian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Seemantini K. Nadkarni
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Gholivand A, Korculanin O, Dahlhoff K, Babaki M, Dickscheid T, Lettinga MP. Effect of in-plane and out-of-plane bifurcated microfluidic channels on the flow of aggregating red blood cells. LAB ON A CHIP 2024; 24:2317-2326. [PMID: 38545688 DOI: 10.1039/d4lc00151f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The blood flow through our microvascular system is a renowned difficult process to understand because the complex flow behavior of blood is intertwined with the complex geometry it has to flow through. Conventional 2D microfluidics has provided important insights, but progress is hampered by the limitation of 2-D confinement. Here we use selective laser-induced etching to excavate non-planar 3-D microfluidic channels in glass that consist of two generations of bifurcations, heading towards more physiological geometries. We identify a cross-talk between the first and second bifurcation only when both bifurcations are in the same plane, as observed in 2D microfluidics. Contrarily, the flow in the branch where the second bifurcation is perpendicular to the first is hardly affected by the initial distortion. This difference in flow behavior is only observed when red blood cells are aggregated, due to the presence of dextran, and disappears by increasing the distance between both generations of bifurcations. Thus, 3-D structures scramble in-plane flow distortions, exemplifying the importance of experimenting with truly 3D microfluidic designs in order to understand complex physiological flow behavior.
Collapse
Affiliation(s)
- Amirreza Gholivand
- Biomacromolecular Systems and Processes (IBI-4), Research Centre Jülich, 52425 Jülich, Germany.
- Laboratory for Soft Matter and Biophysics, KU Leuven, B-3001 Leuven, Belgium
| | - Olivera Korculanin
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons (ER-C-3 Structural Biology), Research Centre Jülich, 52425 Jülich, Germany
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany
| | - Knut Dahlhoff
- Central Institute of Engineering, Electronics and Analytics (ZEA-1), Research Centre Jülich, 52425 Jülich, Germany
| | - Mehrnaz Babaki
- Biomacromolecular Systems and Processes (IBI-4), Research Centre Jülich, 52425 Jülich, Germany.
- Laboratory for Soft Matter and Biophysics, KU Leuven, B-3001 Leuven, Belgium
| | - Timo Dickscheid
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany
- Institute of Computer Science, Heinrich Heine University Düsseldorf, Germany
- Helmholtz AI, Research Centre Jülich, 52425 Jülich, Germany
| | - Minne Paul Lettinga
- Biomacromolecular Systems and Processes (IBI-4), Research Centre Jülich, 52425 Jülich, Germany.
- Laboratory for Soft Matter and Biophysics, KU Leuven, B-3001 Leuven, Belgium
| |
Collapse
|
7
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
8
|
Borges J, Zeng J, Liu XQ, Chang H, Monge C, Garot C, Ren K, Machillot P, Vrana NE, Lavalle P, Akagi T, Matsusaki M, Ji J, Akashi M, Mano JF, Gribova V, Picart C. Recent Developments in Layer-by-Layer Assembly for Drug Delivery and Tissue Engineering Applications. Adv Healthc Mater 2024; 13:e2302713. [PMID: 38116714 PMCID: PMC11469081 DOI: 10.1002/adhm.202302713] [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: 08/17/2023] [Revised: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Surfaces with biological functionalities are of great interest for biomaterials, tissue engineering, biophysics, and for controlling biological processes. The layer-by-layer (LbL) assembly is a highly versatile methodology introduced 30 years ago, which consists of assembling complementary polyelectrolytes or biomolecules in a stepwise manner to form thin self-assembled films. In view of its simplicity, compatibility with biological molecules, and adaptability to any kind of supporting material carrier, this technology has undergone major developments over the past decades. Specific applications have emerged in different biomedical fields owing to the possibility to load or immobilize biomolecules with preserved bioactivity, to use an extremely broad range of biomolecules and supporting carriers, and to modify the film's mechanical properties via crosslinking. In this review, the focus is on the recent developments regarding LbL films formed as 2D or 3D objects for applications in drug delivery and tissue engineering. Possible applications in the fields of vaccinology, 3D biomimetic tissue models, as well as bone and cardiovascular tissue engineering are highlighted. In addition, the most recent technological developments in the field of film construction, such as high-content liquid handling or machine learning, which are expected to open new perspectives in the future developments of LbL, are presented.
Collapse
Grants
- GA259370 ERC "BIOMIM"
- GA692924 ERC "BioactiveCoatings"
- GA790435 ERC "Regenerbone"
- ANR-17-CE13-022 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR-18-CE17-0016 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- 192974 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR-20-CE19-022 BIOFISS Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR22-CE19-0024 SAFEST Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- DOS0062033/0 FUI-BPI France
- 883370 European Research Council "REBORN"
- 2020.00758.CEECIND Portuguese Foundation for Science and Technology
- UIDB/50011/2020,UIDP/50011/2020,LA/P/0006/2020 FCT/MCTES (PIDDAC)
- 751061 European Union's Horizon 2020 "PolyVac"
- 11623 Sidaction
- 20H00665 JSPS Grant-in-Aid for Scientific Research
- 3981662 BPI France Aide Deep Tech programme
- ECTZ60600 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- 101079482 HORIZON EUROPE Framework Programme "SUPRALIFE"
- 101058554 Horizon Europe EIC Accelerator "SPARTHACUS"
- Sidaction
- Agence Nationale de Recherches sur le Sida et les Hépatites Virales
Collapse
Affiliation(s)
- João Borges
- CICECO – Aveiro Institute of MaterialsDepartment of ChemistryUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Jinfeng Zeng
- Division of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Xi Qiu Liu
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hao Chang
- Hangzhou Institute of MedicineChinese Academy of SciencesHangzhouZhejiang310022China
| | - Claire Monge
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI)UMR5305 CNRS/Universite Claude Bernard Lyon 17 Passage du VercorsLyon69367France
| | - Charlotte Garot
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| | - Ke‐feng Ren
- Department of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Paul Machillot
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| | - Nihal E. Vrana
- SPARTHA Medical1 Rue Eugène BoeckelStrasbourg67000France
| | - Philippe Lavalle
- SPARTHA Medical1 Rue Eugène BoeckelStrasbourg67000France
- Institut National de la Santé et de la Recherche MédicaleInserm UMR_S 1121 Biomaterials and BioengineeringCentre de Recherche en Biomédecine de Strasbourg1 rue Eugène BoeckelStrasbourg67000France
- Université de StrasbourgFaculté de Chirurgie Dentaire1 place de l'HôpitalStrasbourg67000France
| | - Takami Akagi
- Building Block Science Joint Research ChairGraduate School of Frontier BiosciencesOsaka University1–3 YamadaokaSuitaOsaka565–0871Japan
| | - Michiya Matsusaki
- Division of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Jian Ji
- Department of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Mitsuru Akashi
- Building Block Science Joint Research ChairGraduate School of Frontier BiosciencesOsaka University1–3 YamadaokaSuitaOsaka565–0871Japan
| | - João F. Mano
- CICECO – Aveiro Institute of MaterialsDepartment of ChemistryUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Varvara Gribova
- Institut National de la Santé et de la Recherche MédicaleInserm UMR_S 1121 Biomaterials and BioengineeringCentre de Recherche en Biomédecine de Strasbourg1 rue Eugène BoeckelStrasbourg67000France
- Université de StrasbourgFaculté de Chirurgie Dentaire1 place de l'HôpitalStrasbourg67000France
| | - Catherine Picart
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| |
Collapse
|
9
|
Martinez-Torres C, Grimbergen J, Koopman J, Koenderink GH. Interplay of fibrinogen α EC globular domains and factor XIIIa cross-linking dictates the extensibility and strain stiffening of fibrin networks. J Thromb Haemost 2024; 22:715-726. [PMID: 37940047 DOI: 10.1016/j.jtha.2023.10.025] [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: 05/12/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Fibrinogen is a plasma protein forming the fibrin scaffold of blood clots. Its mechanical properties therefore affect the risk of bleeding as well as thrombosis. There has been much recent interest in the biophysical mechanisms controlling fibrin mechanics; however, the role of molecular heterogeneity of the circulating fibrinogen in determining clot mechanical function remains poorly characterized. OBJECTIVES By comparing 2 fibrinogen variants where the only difference is the Aα-chain length, with one variant having a globular domain at its C-terminus, this study aimed to reveal how the molecular structure impacts the structure and mechanics of fibrin networks. METHODS We characterized the mechanical response to large shear for networks formed from 2 recombinant fibrinogen variants: the most prevalent variant in circulation with a molecular weight of 340 kDa (recombinant human fibrinogen [rFib] 340) and a minor variant with a molecular weight of 420 kDa (rFib420). RESULTS We show that the elastic properties of the 2 variants are identical when fibrin is cross-linked with factor XIIIa but differ strongly in its absence. Uncross-linked rFib420 networks are softer and up to 3-fold more extensible than rFib340 networks. Electron microscopy imaging showed that the 2 variants formed networks with a comparable structure, except at 4 mg/mL, where rFib420 formed denser networks. CONCLUSION We propose that the αEC domains of rFib420 increase the extensibility of uncross-linked fibrin networks by promoting protofibril sliding, which is blocked by FXIIIa cross-linking. Our findings can help explain the functional role of different circulating fibrinogen variants in blood clot mechanics and tissue repair.
Collapse
Affiliation(s)
- Cristina Martinez-Torres
- AMOLF, Amsterdam, The Netherlands; Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands
| | | | | | - Gijsje H Koenderink
- AMOLF, Amsterdam, The Netherlands; Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
10
|
B Fortela DL, Mikolajczyk AP, Carnes MR, Sharp W, Revellame E, Hernandez R, Holmes WE, Zappi ME. Predicting molecular docking of per- and polyfluoroalkyl substances to blood protein using generative artificial intelligence algorithm DiffDock. Biotechniques 2024; 76:14-26. [PMID: 37947020 DOI: 10.2144/btn-2023-0070] [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: 11/12/2023] Open
Abstract
This study computationally evaluates the molecular docking affinity of various perfluoroalkyl and polyfluoroalkyl substances (PFAs) towards blood proteins using a generative machine-learning algorithm, DiffDock, specialized in protein-ligand blind-docking learning and prediction. Concerns about the chemical pathways and accumulation of PFAs in the environment and eventually in the human body has been rising due to empirical findings that levels of PFAs in human blood has been rising. DiffDock may offer a fast approach in determining the fate and potential molecular pathways of PFAs in human body.
Collapse
Affiliation(s)
- Dhan Lord B Fortela
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| | - Ashley P Mikolajczyk
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| | - Miranda R Carnes
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
| | - Wayne Sharp
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
- Department of Civil Engineering, University of Louisiana, Lafayette, LA 70504, USA
| | - Emmanuel Revellame
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| | - Rafael Hernandez
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| | - William E Holmes
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| | - Mark E Zappi
- Department of Chemical Engineering, University of Louisiana, Lafayette, LA 70504, USA
- Energy Institute of Louisiana, University of Louisiana, Lafayette, LA 70504, USA
| |
Collapse
|
11
|
Robinson SD, de Boisanger J, Pearl FMG, Critchley G, Rosenfelder N, Giamas G. A brain metastasis liquid biopsy: Where are we now? Neurooncol Adv 2024; 6:vdae066. [PMID: 38770219 PMCID: PMC11102938 DOI: 10.1093/noajnl/vdae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Brain metastases remain a challenging and feared complication for patients with cancer and research in this area has lagged behind research into metastases to other organs. Due to their location and the risks associated with neurosurgical biopsies, the biology underpinning brain metastases response to treatment and evolution over time remains poorly understood. Liquid biopsies are proposed to overcome many of the limitations present with tissue biopsies, providing a better representation of tumor heterogeneity, facilitating repeated sampling, and providing a noninvasive assessment of tumor biology. Several different liquid biopsy approaches have been investigated including circulating tumor cells, circulating tumor DNA, extracellular vesicles, and tumor-educated platelets; however, these have generally been less effective in assessing brain metastases compared to metastases to other organs requiring improved techniques to investigate these approaches, studies combining different liquid biopsy approaches and/or novel liquid biopsy approaches. Through this review, we highlight the current state of the art and define key unanswered questions related to brain metastases liquid biopsies.
Collapse
Affiliation(s)
- Stephen David Robinson
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - James de Boisanger
- Neuro-Oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Frances M G Pearl
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Giles Critchley
- Department of Neurosurgery, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Section of Neurosurgery, Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Nicola Rosenfelder
- Neuro-Oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| |
Collapse
|
12
|
Jackson ML, Bond AR, George SJ. Mechanobiology of the endothelium in vascular health and disease: in vitro shear stress models. Cardiovasc Drugs Ther 2023; 37:997-1010. [PMID: 36190667 PMCID: PMC10516801 DOI: 10.1007/s10557-022-07385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
Abstract
In recent years, there has been growing evidence that vascular pathologies arise in sites experiencing an altered haemodynamic environment. Fluid shear stress (FSS) is an important contributor to vascular homeostasis and regulates endothelial cell (EC) gene expression, morphology, and behaviour through specialised mechanosensitive signalling pathways. The presence of an altered FSS profile is a pathological characteristic of many vascular diseases, with the most established example being the preferential localisation of atherosclerotic plaque development. However, the precise haemodynamic contributions to other vascular pathologies including coronary artery vein graft failure remains poorly defined. To evaluate potential novel therapeutics for the treatment of vascular diseases via targeting EC behaviour, it is important to undertake in vitro experiments using appropriate culture conditions, particularly FSS. There are a wide range of in vitro models used to study the effect of FSS on the cultured endothelium, each with the ability to generate FSS flow profiles through which the investigator can control haemodynamic parameters including flow magnitude and directionality. An important consideration for selection of an appropriate model of FSS exposure is the FSS profile that the model can generate, in comparison to the physiological and pathophysiological haemodynamic environment of the vessel of interest. A resource bringing together the haemodynamic environment characteristic of atherosclerosis pathology and the flow profiles generated by in vitro methods of applying FSS would be beneficial to researchers when selecting the appropriate model for their research. Consequently, here we summarise the widely used methods of exposing cultured endothelium to FSS, the flow profile they generate and their advantages and limitations in investigating the pathological contribution of altered FSS to vascular disease and evaluating novel therapeutic targets for the treatment and prevention of vascular disease.
Collapse
Affiliation(s)
- Molly L. Jackson
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| | - Andrew Richard Bond
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| | - Sarah Jane George
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| |
Collapse
|
13
|
Michael C, Pancaldi F, Britton S, Kim OV, Peshkova AD, Vo K, Xu Z, Litvinov RI, Weisel JW, Alber M. Combined computational modeling and experimental study of the biomechanical mechanisms of platelet-driven contraction of fibrin clots. Commun Biol 2023; 6:869. [PMID: 37620422 PMCID: PMC10449797 DOI: 10.1038/s42003-023-05240-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
While blood clot formation has been relatively well studied, little is known about the mechanisms underlying the subsequent structural and mechanical clot remodeling called contraction or retraction. Impairment of the clot contraction process is associated with both life-threatening bleeding and thrombotic conditions, such as ischemic stroke, venous thromboembolism, and others. Recently, blood clot contraction was observed to be hindered in patients with COVID-19. A three-dimensional multiscale computational model is developed and used to quantify biomechanical mechanisms of the kinetics of clot contraction driven by platelet-fibrin pulling interactions. These results provide important biological insights into contraction of platelet filopodia, the mechanically active thin protrusions of the plasma membrane, described previously as performing mostly a sensory function. The biomechanical mechanisms and modeling approach described can potentially apply to studying other systems in which cells are embedded in a filamentous network and exert forces on the extracellular matrix modulated by the substrate stiffness.
Collapse
Affiliation(s)
- Christian Michael
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Francesco Pancaldi
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Samuel Britton
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Oleg V Kim
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biomedical Engineering and Mechanics, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Alina D Peshkova
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Khoi Vo
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Zhiliang Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| | - Mark Alber
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA.
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA.
- Department of Bioengineering, University of California Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
14
|
George A, Akbaridoust F, Zainal Abidin NA, Nesbitt WS, Marusic I. Characterisation of hydrodynamic trapping in microfluidic cross-slot devices for high strain rate applications. LAB ON A CHIP 2023. [PMID: 37305977 DOI: 10.1039/d3lc00256j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hydrodynamic trapping of a particle or cluster of particles based on contact and non-contact approaches has brought prominent insights to micro-nano scale applications. Of the non-contact methods, image-based real-time control in cross-slot microfluidic devices is one of the most promising potential platform for single cellular assays. Here, we report results from experiments conducted in two cross-slot microfluidic channels of different widths, with varying real-time delay of the control algorithm and different magnification. Sustained trapping of 5 μm diameter particles was achieved with high strain rates, of order 102 s-1, higher than in any previous studies. Our experiments show that the maximum attainable strain rate is a function of the real-time delay of the control algorithm and the particle resolution (pixel/μm). Therefore, we anticipate that with further reduced time delays and enhanced particle resolution, considerably higher strain rates can be attained, opening the platform to single cellular assay studies where very high strain rates are required.
Collapse
Affiliation(s)
- Aravind George
- Department of Mechanical Engineering, University of Melbourne, VIC 3010, Australia.
| | - Farzan Akbaridoust
- Department of Mechanical Engineering, University of Melbourne, VIC 3010, Australia.
| | - Nurul A Zainal Abidin
- The Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Warwick S Nesbitt
- The Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Ivan Marusic
- Department of Mechanical Engineering, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
15
|
Leartprapun N, Zeng Z, Hajjarian Z, Bossuyt V, Nadkarni SK. Speckle rheological spectroscopy reveals wideband viscoelastic spectra of biological tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544037. [PMID: 37333220 PMCID: PMC10274797 DOI: 10.1101/2023.06.08.544037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Mechanical transformation of tissue is not merely a symptom but a decisive driver in pathological processes. Comprising intricate network of cells, fibrillar proteins, and interstitial fluid, tissues exhibit distinct solid-(elastic) and liquid-like (viscous) behaviours that span a wide band of frequencies. Yet, characterization of wideband viscoelastic behaviour in whole tissue has not been investigated, leaving a vast knowledge gap in the higher frequency range that is linked to fundamental intracellular processes and microstructural dynamics. Here, we present wideband Speckle rHEologicAl spectRoScopy (SHEARS) to address this need. We demonstrate, for the first time, analysis of frequency-dependent elastic and viscous moduli up to the sub-MHz regime in biomimetic scaffolds and tissue specimens of blood clots, breast tumours, and bone. By capturing previously inaccessible viscoelastic behaviour across the wide frequency spectrum, our approach provides distinct and comprehensive mechanical signatures of tissues that may provide new mechanobiological insights and inform novel disease prognostication.
Collapse
Affiliation(s)
- Nichaluk Leartprapun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Ziqian Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Zeinab Hajjarian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Seemantini K. Nadkarni
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
16
|
Heran W, Xin L, Qi G, Xiongfei Z. Vascularized organ bioprinting: From strategy to paradigm. Cell Prolif 2023; 56:e13453. [PMID: 36929675 DOI: 10.1111/cpr.13453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Over the past two decades, bioprinting has become a popular research topic worldwide, as it is the most promising approach for manufacturing vascularized organ in vitro. However, transitioning bioprinting from simple tissue models to real biomedical applications is still a challenge due to the lack of interdisciplinary theoretical knowledge and perfect multitechnology integration. This review examines the goals of vasculature manufacturing and proposes the objectives in three stages. We then outline a bidirectional manufacturing strategy consisting of top-down reproduction (bioprinting) and bottom-up regeneration (cellular behaviour). We also provide an in-depth analysis of the views from the four aspects of design, ink, printing, and culture. Furthermore, we present the 'constructing-comprehension cycle' research paradigm in Strategic Priority Research Program and the 'math-model-based batch insights generator' research paradigm for the future, which have the potential to revolutionize the biomedical field.
Collapse
Affiliation(s)
- Wang Heran
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liu Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gu Qi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng Xiongfei
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
| |
Collapse
|
17
|
Gou K, Hu JJ, Baek S. Mechanical characterization of human umbilical arteries by thick-walled models: Enhanced vascular compliance by removing an abluminal lining. J Mech Behav Biomed Mater 2023; 142:105811. [PMID: 37028123 DOI: 10.1016/j.jmbbm.2023.105811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/19/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023]
Abstract
The decellularized human umbilical artery (HUA) is considered as a promising option for small-diameter, tissue-engineered vascular grafts (TEVGs). Our previous study showed that the HUA bears a thin, watertight lining on its outermost abluminal surface. Removal of this abluminal lining layer improves efficacy of the perfusion-assisted decellularization of the HUA and increases its compliance. As stress across the wall is believed to affect growth and remodeling of the TEVG, it is imperative to mechanically characterize the HUA using thick-walled models. Combining inflation experiments and computational methods, we investigate the mechanical properties of the HUA before and after the abluminal lining removal to examine the HUA's wall mechanics. The inflation tests of five HUAs were performed to obtain the mechanical and geometrical response of the vessel wall before and after the lining layer removal. Using nonlinear hyperelastic models, the same responses are obtained computationally using the thick-walled models. The experimental data are incorporated into the computational models to estimate the mechanical and orientation parameters of the fibers and isotropic matrix of different layers in the HUAs. The parameter fitting of both thick-walled models (before and after the abluminal lining removal) results in most of the R-squared values for measuring the goodness of fitting to be over 0.90 for all samples. The compliance of the HUA increases from a mean value of 2.60% per 100 mmHg before the removal of the lining to a mean value of 4.21% per 100 mmHg after the removal. The results reveal that, although the abluminal lining is thin, it is stiff and capable of supporting majority of the high luminal pressure, and that the inner layer is far less stressed than the abluminal lining. Computational simulations also show that removal of the abluminal lining increases the circumferential wall stress by up to 280 kPa under the in vivo luminal pressure. The integrated computational and experimental approaches provide more accurate estimates of the material behaviors of HUAs employed in grafts and, in turn, the study enhances our understanding of interactions between the graft and the native vessel on vascular growth and remodeling.
Collapse
Affiliation(s)
- Kun Gou
- Department of Mathematical, Physical, and Engineering Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA.
| | - Jin-Jia Hu
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan.
| | - Seungik Baek
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
18
|
Wei J, Li M. Interplay of Fluid Mechanics and Matrix Stiffness in Tuning the Mechanical Behaviors of Single Cells Probed by Atomic Force Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1309-1319. [PMID: 36633932 DOI: 10.1021/acs.langmuir.2c03137] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
It is well known that both fluid mechanics and matrix stiffness present within the cellular microenvironments play an essential role in the physiological and pathological processes of cells. However, so far, knowledge of the interplay of fluid mechanics and matrix stiffness in tuning the mechanical behaviors of single cells is still extremely limited. Particularly, atomic force microscopy (AFM) is now an important and standard tool for characterizing the mechanical properties of single living cells. Nevertheless, studies of utilizing AFM to detect cellular mechanics are commonly performed in static medium conditions, which are unable to access the effects of fluidic media on cellular behaviors. Here, by integrating AFM with a fluidic cell medium device and hydrogel technology, the combined effects of fluid mechanics and matrix stiffness on cell mechanics were investigated. A fluidic medium device with tunable fluid mechanics was established to simulate the shear flow effects, and hydrogels were used to fabricate substrates with different stiffnesses for cell growth. Especially, the cantilever of the AFM probe was modified with a microsphere to indent cells for probing cell mechanics. Based on the established experimental platform, the elastic and viscous properties of single living cells grown on substrates with tunable matrix stiffness under fluidic microenvironments were quantitatively measured, and the remarkable alterations in the mechanical properties of cells were unraveled. The subcellular structure changes of cells in fluidic microenvironments were observed by fluorescence microscopy. Further, AFM morphological imaging was used to image living cells grown in fluidic medium conditions, and significant changes in the surface structure and roughness of cells were observed. The study provides a novel way to investigate the synergistic effects of fluid mechanics and matrix stiffness on the behaviors of single cells, which will benefit unveiling the underlying mechanical cues involved the interactions between microenvironments and cells.
Collapse
Affiliation(s)
- Jiajia Wei
- State Key Laboratory of Robotics, Chinese Academy of Sciences, Shenyang Institute of Automation, Shenyang110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang110169, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Mi Li
- State Key Laboratory of Robotics, Chinese Academy of Sciences, Shenyang Institute of Automation, Shenyang110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang110169, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
19
|
Deng P, Zhao M, Zhang X, Qin J. A Transwell-Based Vascularized Model to Investigate the Effect of Interstitial Flow on Vasculogenesis. Bioengineering (Basel) 2022; 9:668. [PMID: 36354579 PMCID: PMC9687519 DOI: 10.3390/bioengineering9110668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 09/08/2024] Open
Abstract
Interstitial flow plays a significant role in vascular system development, mainly including angiogenesis and vasculogenesis. However, compared to angiogenesis, the effect of interstitial flow on vasculogenesis is less explored. Current in vitro models for investigating the effect of interstitial flow on vasculogenesis heavily rely on microfluidic chips, which require microfluidic expertise and facilities, and may not be accessible to biological labs. Here, we proposed a facile approach to building perfusable vascular networks through the self-assembly of endothelial cells in a modified transwell format and investigated the effect of interstitial flow on vasculogenesis. We found that the effect of interstitial flow on vasculogenesis was closely related to the existence of VEGF and fibroblasts in the developed model: (1) In the presence of fibroblasts, interstitial flow (within the range of 0.1-0.6 μm/s) facilitated the perfusability of the engineered vasculatures. Additional VEGF in the culture medium further worked synergically with interstitial flow to develop longer, wider, denser, and more perfusable vasculatures than static counterparts; (2) In the absence of fibroblasts, vasculatures underwent severe regression within 7 days under static conditions. However, interstitial flow greatly inhibited vessel regression and enhanced vascular perfusability and morphogenesis without the need for additional VEGF. These results revealed that the effect of interstitial flow might vary depending on the existence of VEGF and fibroblasts, and would provide some guidelines for constructing in vitro self-assembled vasculatures. The established transwell-based vascularized model provides a simple method to build perfusable vasculatures and could also be utilized for creating functional tissues in regenerative medicine.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
20
|
Matei AE, Kubánková M, Xu L, Györfi AH, Boxberger E, Soteriou D, Papava M, Prater J, Hong X, Bergmann C, Kräter M, Schett G, Guck J, Distler JHW. Identification of a Distinct Monocyte-Driven Signature in Systemic Sclerosis Using Biophysical Phenotyping of Circulating Immune Cells. Arthritis Rheumatol 2022; 75:768-781. [PMID: 36281753 DOI: 10.1002/art.42394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pathologically activated circulating immune cells, including monocytes, play major roles in systemic sclerosis (SSc). Their functional characterization can provide crucial information with direct clinical relevance. However, tools for the evaluation of pathologic immune cell activation and, in general, of clinical outcomes in SSc are scarce. Biophysical phenotyping (including characterization of cell mechanics and morphology) provides access to a novel, mostly unexplored layer of information regarding pathophysiologic immune cell activation. We hypothesized that the biophysical phenotyping of circulating immune cells, reflecting their pathologic activation, can be used as a clinical tool for the evaluation and risk stratification of patients with SSc. METHODS We performed biophysical phenotyping of circulating immune cells by real-time fluorescence and deformability cytometry (RT-FDC) in 63 SSc patients, 59 rheumatoid arthritis (RA) patients, 28 antineutrophil cytoplasmic antibody-associated vasculitis (AAV) patients, and 22 age- and sex-matched healthy donors. RESULTS We identified a specific signature of biophysical properties of circulating immune cells in SSc patients that was mainly driven by monocytes. Since it is absent in RA and AAV, this signature reflects an SSc-specific monocyte activation rather than general inflammation. The biophysical properties of monocytes indicate current disease activity, the extent of skin or lung fibrosis, and the severity of manifestations of microvascular damage, as well as the risk of disease progression in SSc patients. CONCLUSION Changes in the biophysical properties of circulating immune cells reflect their pathologic activation in SSc patients and are associated with clinical outcomes. As a high-throughput approach that requires minimal preparations, RT-FDC-based biophysical phenotyping of monocytes can serve as a tool for the evaluation and risk stratification of patients with SSc.
Collapse
Affiliation(s)
- Alexandru-Emil Matei
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Liyan Xu
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Evgenia Boxberger
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Despina Soteriou
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany
| | - Maria Papava
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Prater
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xuezhi Hong
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
21
|
Su Z, Chen Z, Ma K, Chen H, Ho JWK. Molecular determinants of intrinsic cellular stiffness in health and disease. Biophys Rev 2022; 14:1197-1209. [PMID: 36345276 PMCID: PMC9636357 DOI: 10.1007/s12551-022-00997-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/11/2022] [Indexed: 10/14/2022] Open
Abstract
In recent years, the role of intrinsic biophysical features, especially cellular stiffness, in diverse cellular and disease processes is being increasingly recognized. New high throughput techniques for the quantification of cellular stiffness facilitate the study of their roles in health and diseases. In this review, we summarized recent discovery about how cellular stiffness is involved in cell stemness, tumorigenesis, and blood diseases. In addition, we review the molecular mechanisms underlying the gene regulation of cellular stiffness in health and disease progression. Finally, we discussed the current understanding on how the cytoskeleton structure and the regulation of these genes contribute to cellular stiffness, highlighting where the field of cellular stiffness is headed.
Collapse
Affiliation(s)
- Zezhuo Su
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong, SAR China
| | - Zhenlin Chen
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR China
| | - Kun Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong, SAR China
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen, 518055 China
| | - Joshua W. K. Ho
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong, SAR China
| |
Collapse
|
22
|
Ozdemir S, Yalcin-Enis I, Yalcinkaya B, Yalcinkaya F. An Investigation of the Constructional Design Components Affecting the Mechanical Response and Cellular Activity of Electrospun Vascular Grafts. MEMBRANES 2022; 12:929. [PMID: 36295688 PMCID: PMC9607146 DOI: 10.3390/membranes12100929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Cardiovascular disease is anticipated to remain the leading cause of death globally. Due to the current problems connected with using autologous arteries for bypass surgery, researchers are developing tissue-engineered vascular grafts (TEVGs). The major goal of vascular tissue engineering is to construct prostheses that closely resemble native blood vessels in terms of morphological, mechanical, and biological features so that these scaffolds can satisfy the functional requirements of the native tissue. In this setting, morphology and cellular investigation are usually prioritized, while mechanical qualities are generally addressed superficially. However, producing grafts with good mechanical properties similar to native vessels is crucial for enhancing the clinical performance of vascular grafts, exposing physiological forces, and preventing graft failure caused by intimal hyperplasia, thrombosis, aneurysm, blood leakage, and occlusion. The scaffold's design and composition play a significant role in determining its mechanical characteristics, including suturability, compliance, tensile strength, burst pressure, and blood permeability. Electrospun prostheses offer various models that can be customized to resemble the extracellular matrix. This review aims to provide a comprehensive and comparative review of recent studies on the mechanical properties of fibrous vascular grafts, emphasizing the influence of structural parameters on mechanical behavior. Additionally, this review provides an overview of permeability and cell growth in electrospun membranes for vascular grafts. This work intends to shed light on the design parameters required to maintain the mechanical stability of vascular grafts placed in the body to produce a temporary backbone and to be biodegraded when necessary, allowing an autologous vessel to take its place.
Collapse
Affiliation(s)
- Suzan Ozdemir
- Textile Engineering Department, Textile Technologies and Design Faculty, Istanbul Technical University, Beyoglu, 34467 Istanbul, Turkey
| | - Ipek Yalcin-Enis
- Textile Engineering Department, Textile Technologies and Design Faculty, Istanbul Technical University, Beyoglu, 34467 Istanbul, Turkey
| | - Baturalp Yalcinkaya
- Department of Material Science, Faculty of Mechanical Engineering, Technical University of Liberec, 461 17 Liberec, Czech Republic
| | - Fatma Yalcinkaya
- Department of Environmental Technology, Institute for Nanomaterials, Advanced Technologies and Innovations, Technical University of Liberec, 461 17 Liberec, Czech Republic
| |
Collapse
|
23
|
Wang S, Liu Q, Cheng L, Wang L, Xu F, Yao C. Targeting biophysical cues to address platelet storage lesions. Acta Biomater 2022; 151:118-133. [PMID: 36028196 DOI: 10.1016/j.actbio.2022.08.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/06/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022]
Abstract
Platelets play vital roles in vascular repair, especially in primary hemostasis, and have been widely used in transfusion to prevent bleeding or manage active bleeding. Recently, platelets have been used in tissue repair (e.g., bone, skin, and dental alveolar tissue) and cell engineering as drug delivery carriers. However, the biomedical applications of platelets have been associated with platelet storage lesions (PSLs), resulting in poor clinical outcomes with reduced recovery, survival, and hemostatic function after transfusion. Accumulating evidence has shown that biophysical cues play important roles in platelet lesions, such as granule secretion caused by shear stress, adhesion affected by substrate stiffness, and apoptosis caused by low temperature. This review summarizes four major biophysical cues (i.e., shear stress, substrate stiffness, hydrostatic pressure, and thermal microenvironment) involved in the platelet preparation and storage processes, and discusses how they may synergistically induce PSLs such as platelet shape change, activation, apoptosis and clearance. We also review emerging methods for studying these biophysical cues in vitro and existing strategies targeting biophysical cues for mitigating PSLs. We conclude with a perspective on the future direction of biophysics-based strategies for inhibiting PSLs. STATEMENT OF SIGNIFICANCE: Platelet storage lesions (PSLs) involve a series of structural and functional changes. It has long been accepted that PSLs are initiated by biochemical cues. Our manuscript is the first to propose four major biophysical cues (shear stress, substrate stiffness, hydrostatic pressure, and thermal microenvironment) that platelets experience in each operation step during platelet preparation and storage processes in vitro, which may synergistically contribute to PSLs. We first clarify these biophysical cues and how they induce PSLs. Strategies targeting each biophysical cue to improve PSLs are also summarized. Our review is designed to draw the attention from a broad range of audience, including clinical doctors, biologists, physical scientists, engineers and materials scientists, and immunologist, who study on platelets physiology and pathology.
Collapse
Affiliation(s)
- Shichun Wang
- Department of Blood Transfusion, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Qi Liu
- Department of Blood Transfusion, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Lihan Cheng
- Department of Blood Transfusion, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Lu Wang
- Department of Blood Transfusion, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China.
| | - Chunyan Yao
- Department of Blood Transfusion, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
24
|
Pozhitkova AV, Kladko DV, Vinnik DA, Taskaev SV, Vinogradov VV. Reprogrammable Soft Swimmers for Minimally Invasive Thrombus Extraction. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23896-23908. [PMID: 35537068 DOI: 10.1021/acsami.2c04745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Thrombosis-related diseases are the primary cause of death in the world. Despite recent advances in thrombosis treatment methods, their invasive nature remains a crucial factor, which leads to considerable deadly consequences. Soft magnetic robots are attracting widespread interest due to their fast response, remote actuation, and shape reprogrammability and can potentially avoid the side effects of conventional approaches. This paper outlines a new approach to the thrombosis treatment via reprogrammable magnetic soft robots that penetrate, hook, and extract the plasma clots in a vein-mimicking system under applied rotating magnetic fields. We present shape-switching bioinspired soft swimmers, capable of locomotion by different mechanisms in vein-mimicking flow conditions and whose swimming efficiency is similar to animals. Further, we demonstrate the potential of a developed robot for minimally invasive thromboextraction with and without fibrinolytic usage, including hooking the plasma clot for 3.1 ± 1.1 min and extracting it from the vein-mimicking system under the applied magnetic fields. We consider an interesting solution for thrombosis treatment to avoid substantial drawbacks of the existing methods.
Collapse
Affiliation(s)
- Anna V Pozhitkova
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg 197101, Russia
| | - Daniil V Kladko
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg 197101, Russia
| | - Denis A Vinnik
- National Research South Ural State University, Chelyabinsk 454080, Russia
| | - Sergey V Taskaev
- National Research South Ural State University, Chelyabinsk 454080, Russia
- Chelyabinsk State University, Chelyabinsk 454001, Russia
| | - Vladimir V Vinogradov
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg 197101, Russia
| |
Collapse
|
25
|
Clinical Performance Study of a New Fully Automated Red Blood Cell Permeability Fragility Analyzer. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:5642907. [PMID: 35392140 PMCID: PMC8983219 DOI: 10.1155/2022/5642907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 11/21/2022]
Abstract
In order to verify the applicability of the erythrocyte fragility test (EFT) carried out by the new fully automatic erythrocyte permeability fragility analyzer RA-800 for thalassemia screening, a total of 100 cases of suspected thalassemia patients who underwent pregnancy examinations at Luohu District People's Hospital are included. The results of a new automatic erythrocyte permeability fragility analyzer RA-800 are compared with the results of the detection system composing of the KOFA erythrocyte fragility test kit currently used in clinical laboratories. The diagnosis confirmed by genetic testing is used as the gold standard to evaluate the applicability of RA-800. The sensitivity, specificity, and accuracy of the new automatic erythrocyte permeability fragility analyzer RA-800 screening for thalassemia were 66.67%, 92.86%, and 85.00%. The KOFA direct colorimetries are 76.67%, 81.43%, and 80.00%. The kappa value for the screening of thalassemia was 0.558, which concludes that the consistency was moderate. The ROC curve indicates that both two methods had diagnostic significance for the diagnostic value of thalassemia. The new automatic erythrocyte permeability fragility analyzer RA-800 is suitable for thalassemia screening, and the performance indexes meet the clinical requirements.
Collapse
|
26
|
Sachs L, Wesche J, Lenkeit L, Greinacher A, Bender M, Otto O, Palankar R. Ex vivo anticoagulants affect human blood platelet biomechanics with implications for high-throughput functional mechanophenotyping. Commun Biol 2022; 5:86. [PMID: 35064207 PMCID: PMC8782918 DOI: 10.1038/s42003-021-02982-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022] Open
Abstract
Inherited platelet disorders affecting the human platelet cytoskeleton result in increased bleeding risk. However, deciphering their impact on cytoskeleton-dependent intrinsic biomechanics of platelets remains challenging and represents an unmet need from a diagnostic and prognostic perspective. It is currently unclear whether ex vivo anticoagulants used during collection of peripheral blood impact the mechanophenotype of cellular components of blood. Using unbiased, high-throughput functional mechanophenotyping of single human platelets by real-time deformability cytometry, we found that ex vivo anticoagulants are a critical pre-analytical variable that differentially influences platelet deformation, their size, and functional response to agonists by altering the cytoskeleton. We applied our findings to characterize the functional mechanophenotype of platelets from a patient with Myosin Heavy Chain 9 (MYH9) related macrothrombocytopenia. Our data suggest that platelets from MYH9 p.E1841K mutation in humans affecting platelet non-muscle myosin heavy chain IIa (NMMHC-IIA) are biomechanically less deformable in comparison to platelets from healthy individuals. Sachs et al. examine the effects of different ex vivo anticoagulants on the biomechanical and functional properties of single platelets using high-throughput real-time fluorescence and deformability cytometry (RT-FDC). Their results demonstrate that the choice of ex vivo anticoagulant may strongly impact the outcomes of mechanophenotyping.
Collapse
|
27
|
Jiang S, Liu S, Lau S, Li J. Hemostatic biomaterials to halt non-compressible hemorrhage. J Mater Chem B 2022; 10:7239-7259. [DOI: 10.1039/d2tb00546h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Non-compressible hemorrhage is an unmet clinical challenge, which occurs in inaccessible sites in the body where compression cannot be applied to stop bleeding. Current treatments reliant on blood transfusion are...
Collapse
|
28
|
Rausch MK, Parekh SH, Dortdivanlioglu B, Rosales AM. Synthetic hydrogels as blood clot mimicking wound healing materials. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2021; 3:042006. [PMID: 35822083 PMCID: PMC9273113 DOI: 10.1088/2516-1091/ac23a4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Excessive bleeding-or hemorrhage-causes millions of civilian and non-civilian casualties every year. Additionally, wound sequelae, such as infections, are a significant source of chronic morbidity, even if the initial bleeding is successfully stopped. To treat acute and chronic wounds, numerous wound healing materials have been identified, tested, and adopted. Among them are topical dressings, such as gauzes, as well as natural and biomimetic materials. However, none of these materials successfully mimic the complex and dynamic properties of the body's own wound healing material: the blood clot. Specifically, blood clots exhibit complex mechanical and biochemical properties that vary across spatial and temporal scales to guide the wound healing response, which make them the ideal wound healing material. In this manuscript, we review blood clots' complex mechanical and biochemical properties, review current wound healing materials, and identify opportunities where new materials can provide additional functionality, with a specific focus on hydrogels. We highlight recent developments in synthetic hydrogels that make them capable of mimicking a larger subset of blood clot features: as plugs and as stimuli for tissue repair. We conclude that future hydrogel materials designed to mimic blood clot biochemistry, mechanics, and architecture can be combined with exciting platelet-like particles to serve as hemostats that also promote the biological wound healing response. Thus, we believe synthetic hydrogels are ideal candidates to address the clear need for better wound healing materials.
Collapse
Affiliation(s)
- Manuel K. Rausch
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, United States of America
- Department of Aerospace Engineering & Engineering Mechanics, University of Texas at Austin, Austin, TX 78712, United States of America
- Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712, United States of America
| | - Sapun H. Parekh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, United States of America
| | - Berkin Dortdivanlioglu
- Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712, United States of America
- Department of Civil, Architectural and Environmental Engineering, University of Texas at Austin, Austin, TX 78712, United States of America
| | - Adrianne M. Rosales
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, United States of America
| |
Collapse
|
29
|
Wang C, Zhao X, Sun Z, Yao N, Zhang A, Guo S. IMPROVEMENT OF CARDIOVASCULAR FUNCTION HEALTH LEVEL BY REGULAR SPORTS. REV BRAS MED ESPORTE 2021. [DOI: 10.1590/1517-8692202127032021_0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Introduction Regular physical activity helps improve cardiovascular and cerebrovascular skills. How to evaluate the nervous tension of the cardiovascular and cerebrovascular vessels through sports is a hot topic. Objective The paper discusses the influence of regular participation in sports on people’s cardiovascular function and blood-related indicators. Methods We select 30 healthy older adults who regularly participate in sports, record their ECG changes, blood pressure, heart rate and other related cardiovascular function indicators, and analyze the blood function of the elderly. Detection of blood cell count (RBC), red blood cell volume (MCV) and hemoglobin (Hb), serum creatinine (Cr), blood glucose (BGS), triglycerides (TG), cholesterol (TC), low-density lipoprotein (LDL) and high-Density lipoprotein (HDL) is measured. Results Older adults who persist in exercise for a long time have better indicators than those who do not exercise. Conclusions Appropriate aerobic exercise can reduce the stiffness of blood vessels in the elderly. Exercise can help the elderly increase heart rate variability and improve the heart’s autonomic nerve function’s blood indicators, and body mass. Level of evidence II; Therapeutic studies - investigation of treatment results.
Collapse
Affiliation(s)
| | | | | | - Nan Yao
- Hebei North University, China
| | | | | |
Collapse
|
30
|
Arora N, Hazra JP, Rakshit S. Anisotropy in mechanical unfolding of protein upon partner-assisted pulling and handle-assisted pulling. Commun Biol 2021; 4:925. [PMID: 34326473 PMCID: PMC8322310 DOI: 10.1038/s42003-021-02445-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Proteins as force-sensors respond to mechanical cues and regulate signaling in physiology. Proteins commonly connect the source and response points of mechanical cues in two conformations, independent proteins in end-to-end geometry and protein complexes in handshake geometry. The force-responsive property of independent proteins in end-to-end geometry is studied extensively using single-molecule force spectroscopy (SMFS). The physiological significance of the complex conformations in force-sensing is often disregarded as mere surge protectors. However, with the potential of force-steering, protein complexes possess a distinct mechano-responsive property over individual force-sensors. To decipher, we choose a force-sensing protein, cadherin-23, from tip-link complex and perform SMFS using end-to-end geometry and handshake complex geometry. We measure higher force-resilience of cadherin-23 with preferential shorter extensions in handshake mode of pulling over the direct mode. The handshake geometry drives the force-response of cadherin-23 through different potential-energy landscapes than direct pulling. Analysis of the dynamic network structure of cadherin-23 under tension indicates narrow force-distributions among residues in cadherin-23 in direct pulling, resulting in low force-dissipation paths and low resilience to force. Overall, the distinct and superior mechanical responses of cadherin-23 in handshake geometry than single protein geometry highlight a probable evolutionary drive of protein-protein complexes as force-conveyors over independent ones.
Collapse
Affiliation(s)
- Nisha Arora
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Jagadish Prasad Hazra
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| | - Sabyasachi Rakshit
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| |
Collapse
|
31
|
Kubánková M, Hohberger B, Hoffmanns J, Fürst J, Herrmann M, Guck J, Kräter M. Physical phenotype of blood cells is altered in COVID-19. Biophys J 2021; 120:2838-2847. [PMID: 34087216 PMCID: PMC8169220 DOI: 10.1016/j.bpj.2021.05.025] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
Clinical syndrome coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 is characterized by rapid spreading and high mortality worldwide. Although the pathology is not yet fully understood, hyperinflammatory response and coagulation disorders leading to congestions of microvessels are considered to be key drivers of the still-increasing death toll. Until now, physical changes of blood cells have not been considered to play a role in COVID-19 related vascular occlusion and organ damage. Here, we report an evaluation of multiple physical parameters including the mechanical features of five frequent blood cell types, namely erythrocytes, lymphocytes, monocytes, neutrophils, and eosinophils. More than four million blood cells of 17 COVID-19 patients at different levels of severity, 24 volunteers free from infectious or inflammatory diseases, and 14 recovered COVID-19 patients were analyzed. We found significant changes in lymphocyte stiffness, monocyte size, neutrophil size and deformability, and heterogeneity of erythrocyte deformation and size. Although some of these changes recovered to normal values after hospitalization, others persisted for months after hospital discharge, evidencing the long-term imprint of COVID-19 on the body.
Collapse
Affiliation(s)
- Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Jakob Hoffmanns
- Department of Ophthalmology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Fürst
- Department of Internal Medicine 1, University Medical Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, University Medical Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| |
Collapse
|
32
|
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
|
33
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
Isiksacan Z, Serhatlioglu M, Elbuken C. In vitro analysis of multiple blood flow determinants using red blood cell dynamics under oscillatory flow. Analyst 2021; 145:5996-6005. [PMID: 32720945 DOI: 10.1039/d0an00604a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The flow behavior of blood is determined mainly by red blood cell (RBC) deformation and aggregation as well as blood viscoelasticity. These intricately interdependent parameters should be monitored by healthcare providers to understand all aspects of circulatory flow dynamics under numerous cases including cardiovascular and infectious diseases. Current medical instruments and microfluidic systems lack the ability to quantify these parameters all at once and in physiologically relevant flow conditions. This work presents a handheld platform and a measurement method for quantitative analysis of multiple of these parameters from 50 μl undiluted blood inside a miniaturized channel. The assay is based on an optical transmission analysis of collective RBC deformation and aggregation under near-infrared illumination during a 1 s damped oscillatory flow and at stasis, respectively. Measurements with blood of different hemo-rheological properties demonstrate that the presented approach holds a potential for initiating simultaneous and routine on-chip blood flow analysis even in resource-poor settings.
Collapse
Affiliation(s)
- Ziya Isiksacan
- UNAM - National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey.
| | | | | |
Collapse
|
35
|
Xia J, Cai LH, Wu H, MacKintosh FC, Weitz DA. Anomalous mechanics of Zn 2+-modified fibrin networks. Proc Natl Acad Sci U S A 2021; 118:e2020541118. [PMID: 33649231 PMCID: PMC7958264 DOI: 10.1073/pnas.2020541118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fibrin is the main component of blood clots. The mechanical properties of fibrin are therefore of critical importance in successful hemostasis. One of the divalent cations released by platelets during hemostasis is Zn2+; however, its effect on the network structure of fibrin gels and on the resultant mechanical properties remains poorly understood. Here, by combining mechanical measurements with three-dimensional confocal microscopy imaging, we show that Zn2+ can tune the fibrin network structure and alter its mechanical properties. In the presence of Zn2+, fibrin protofibrils form large bundles that cause a coarsening of the fibrin network due to an increase in fiber diameter and reduction of the total fiber length. We further show that the protofibrils in these bundles are loosely coupled to one another, which results in a decrease of the elastic modulus with increasing Zn2+ concentrations. We explore the elastic properties of these networks at both low and high stress: At low stress, the elasticity originates from pulling the thermal slack out of the network, and this is consistent with the thermal bending of the fibers. By contrast, at high stress, the elasticity exhibits a common master curve consistent with the stretching of individual protofibrils. These results show that the mechanics of a fibrin network are closely correlated with its microscopic structure and inform our understanding of the structure and physical mechanisms leading to defective or excessive clot stiffness.
Collapse
Affiliation(s)
- Jing Xia
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Li-Heng Cai
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Department of Materials Science and Engineering, University of Virginia, Charlottesville, VA 22904
| | - Huayin Wu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Frederick C MacKintosh
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX 77005
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77030
- Department of Chemistry and Physics, Rice University, Houston, TX 77005
- Department of Astronomy, Rice University, Houston, TX 77005
| | - David A Weitz
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138;
- Department of Physics, Harvard University, Cambridge, MA 02138
| |
Collapse
|
36
|
Wang Q, Chan KF, Schweizer K, Du X, Jin D, Yu SCH, Nelson BJ, Zhang L. Ultrasound Doppler-guided real-time navigation of a magnetic microswarm for active endovascular delivery. SCIENCE ADVANCES 2021; 7:7/9/eabe5914. [PMID: 33637532 PMCID: PMC7909881 DOI: 10.1126/sciadv.abe5914] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/12/2021] [Indexed: 05/18/2023]
Abstract
Swarming micro/nanorobots offer great promise in performing targeted delivery inside diverse hard-to-reach environments. However, swarm navigation in dynamic environments challenges delivery capability and real-time swarm localization. Here, we report a strategy to navigate a nanoparticle microswarm in real time under ultrasound Doppler imaging guidance for active endovascular delivery. A magnetic microswarm was formed and navigated near the boundary of vessels, where the reduced drag of blood flow and strong interactions between nanoparticles enable upstream and downstream navigation in flowing blood (mean velocity up to 40.8 mm/s). The microswarm-induced three-dimensional blood flow enables Doppler imaging from multiple viewing configurations and real-time tracking in different environments (i.e., stagnant, flowing blood, and pulsatile flow). We also demonstrate the ultrasound Doppler-guided swarm formation and navigation in the porcine coronary artery ex vivo. Our strategy presents a promising connection between swarm control and real-time imaging of microrobotic swarms for localized delivery in dynamic environments.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong (CUHK), Shatin, NT, Hong Kong, China
| | - Kai Fung Chan
- Chow Yuk Ho Technology Centre for Innovative Medicine, CUHK, Shatin, NT, Hong Kong, China
- Department of Biomedical Engineering, CUHK, Shatin, NT, Hong Kong, China
| | - Kathrin Schweizer
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong (CUHK), Shatin, NT, Hong Kong, China
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
| | - Xingzhou Du
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong (CUHK), Shatin, NT, Hong Kong, China
- Department of Biomedical Engineering, CUHK, Shatin, NT, Hong Kong, China
| | - Dongdong Jin
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong (CUHK), Shatin, NT, Hong Kong, China
- Department of Biomedical Engineering, CUHK, Shatin, NT, Hong Kong, China
| | - Simon Chun Ho Yu
- Department of Imaging and Interventional Radiology, CUHK, Shatin, NT, Hong Kong, China
| | - Bradley J Nelson
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
| | - Li Zhang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong (CUHK), Shatin, NT, Hong Kong, China.
- Chow Yuk Ho Technology Centre for Innovative Medicine, CUHK, Shatin, NT, Hong Kong, China
- CUHK T Stone Robotics Institute, CUHK, Shatin, NT, Hong Kong, China
| |
Collapse
|
37
|
Esteves M, Monteiro MP, Duarte JA. The effects of vascularization on tumor development: A systematic review and meta-analysis of pre-clinical studies. Crit Rev Oncol Hematol 2021; 159:103245. [PMID: 33508446 DOI: 10.1016/j.critrevonc.2021.103245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE This review aimed to systematize and quantify the existing evidence about the effect of tumor vascularization on its growth, in preclinical studies. METHODOLOGY A computerized research on databases PubMed, Scopus and EBSCO was performed to identify studies that evaluate both the vascularization parameters and the development of the tumors in animal models and the mean differences were calculated through a random effects model. RESULTS Thirteen studies met the inclusion criteria and were included in the systematic review, of which, 6 studies were included in the meta-analysis. Besides tumor vascular density that all studies evaluated, 3 studies analysed the tumor perfusion, 2 studies the tumor hypoxia and 3 studies assessed the grade of vessel maturation. Most of the studies (11) related decreased tumor vascularization and a concomitant inhibition of tumor growth or metastasis development. Quantitatively, the decrease in tumor vascularization contributed to a significant decrease in the tumor growing rate of 5.23 (-9.20, -1.26). CONCLUSION A reduced level of tumor vascularization seems to be able to inhibit tumor growth and progression.
Collapse
Affiliation(s)
- Mário Esteves
- Department of Physical Medicine and Rehabilitation, Hospital-Escola, Fernando Pessoa University, Avenida Fernando Pessoa 150, 4420-096 Gondomar, Portugal; Laboratory of Biochemistry and Experimental Morphology, CIAFEL, R. Dr. Plácido Costa 91, 4200-450 Porto, Portugal.
| | - Mariana P Monteiro
- Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - José Alberto Duarte
- CIAFEL, Faculty of Sports, University of Porto, R. Dr. Plácido Costa 91, 4200-450 Porto, Portugal; Instituto Universitário de Ciências da Saúde, R. Central da Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
38
|
Xu J, Zhang Y, Nie G. Intelligent antithrombotic nanomedicines: Progress, opportunities, and challenges. VIEW 2021. [DOI: 10.1002/viw.20200145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
- GBA Research Innovation Institute for Nanotechnology Guangdong China
- Henan Institute of Advanced Technology Zhengzhou University Zhengzhou China
| |
Collapse
|
39
|
3D printing of tissue engineering scaffolds: a focus on vascular regeneration. Biodes Manuf 2021; 4:344-378. [PMID: 33425460 PMCID: PMC7779248 DOI: 10.1007/s42242-020-00109-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/24/2020] [Indexed: 01/31/2023]
Abstract
Tissue engineering is an emerging means for resolving the problems of tissue repair and organ replacement in regenerative medicine. Insufficient supply of nutrients and oxygen to cells in large-scale tissues has led to the demand to prepare blood vessels. Scaffold-based tissue engineering approaches are effective methods to form new blood vessel tissues. The demand for blood vessels prompts systematic research on fabrication strategies of vascular scaffolds for tissue engineering. Recent advances in 3D printing have facilitated fabrication of vascular scaffolds, contributing to broad prospects for tissue vascularization. This review presents state of the art on modeling methods, print materials and preparation processes for fabrication of vascular scaffolds, and discusses the advantages and application fields of each method. Specially, significance and importance of scaffold-based tissue engineering for vascular regeneration are emphasized. Print materials and preparation processes are discussed in detail. And a focus is placed on preparation processes based on 3D printing technologies and traditional manufacturing technologies including casting, electrospinning, and Lego-like construction. And related studies are exemplified. Transformation of vascular scaffolds to clinical application is discussed. Also, four trends of 3D printing of tissue engineering vascular scaffolds are presented, including machine learning, near-infrared photopolymerization, 4D printing, and combination of self-assembly and 3D printing-based methods.
Collapse
|
40
|
Esteves M, Monteiro MP, Duarte JA. Role of Regular Physical Exercise in Tumor Vasculature: Favorable Modulator of Tumor Milieu. Int J Sports Med 2020; 42:389-406. [PMID: 33307553 DOI: 10.1055/a-1308-3476] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The tumor vessel network has been investigated as a precursor of an inhospitable tumor microenvironment, including its repercussions in tumor perfusion, oxygenation, interstitial fluid pressure, pH, and immune response. Dysfunctional tumor vasculature leads to the extravasation of blood to the interstitial space, hindering proper perfusion and causing interstitial hypertension. Consequently, the inadequate delivery of oxygen and clearance of by-products of metabolism promote the development of intratumoral hypoxia and acidification, hampering the action of immune cells and resulting in more aggressive tumors. Thus, pharmacological strategies targeting tumor vasculature were developed, but the overall outcome was not satisfactory due to its transient nature and the higher risk of hypoxia and metastasis. Therefore, physical exercise emerged as a potential favorable modulator of tumor vasculature, improving intratumoral vascularization and perfusion. Indeed, it seems that regular exercise practice is associated with lasting tumor vascular maturity, reduced vascular resistance, and increased vascular conductance. Higher vascular conductance reduces intratumoral hypoxia and increases the accessibility of circulating immune cells to the tumor milieu, inhibiting tumor development and improving cancer treatment. The present paper describes the implications of abnormal vasculature on the tumor microenvironment and the underlying mechanisms promoted by regular physical exercise for the re-establishment of more physiological tumor vasculature.
Collapse
Affiliation(s)
- Mário Esteves
- Laboratory of Biochemistry and Experimental Morphology, CIAFEL, Porto, Portugal.,Department of Physical Medicine and Rehabilitation, Hospital-Escola, Fernando Pessoa University, Gondomar, Portugal
| | - Mariana P Monteiro
- Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Jose Alberto Duarte
- CIAFEL - Faculty of Sport, University of Porto, Porto, Portugal.,Instituto Universitário de Ciências da Saúde, Gandra, Portugal
| |
Collapse
|
41
|
Zhao Z, Pan DC, Qi QM, Kim J, Kapate N, Sun T, Shields CW, Wang LLW, Wu D, Kwon CJ, He W, Guo J, Mitragotri S. Engineering of Living Cells with Polyphenol-Functionalized Biologically Active Nanocomplexes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2003492. [PMID: 33150643 DOI: 10.1002/adma.202003492] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/15/2020] [Indexed: 06/11/2023]
Abstract
Approaches to safely and effectively augment cellular functions without compromising the inherent biological properties of the cells, especially through the integration of biologically labile domains, remain of great interest. Here, a versatile strategy to assemble biologically active nanocomplexes, including proteins, DNA, mRNA, and even viral carriers, on cellular surfaces to generate a cell-based hybrid system referred to as "Cellnex" is established. This strategy can be used to engineer a wide range of cell types used in adoptive cell transfers, including erythrocytes, macrophages, NK cells, T cells, etc. Erythrocytenex can enhance the delivery of cargo proteins to the lungs in vivo by 11-fold as compared to the free cargo counterpart. Biomimetic microfluidic experiments and modeling provided detailed insights into the targeting mechanism. In addition, Macrophagenex is capable of enhancing the therapeutic efficiency of anti-PD-L1 checkpoint inhibitors in vivo. This simple and adaptable approach may offer a platform for the rapid generation of complex cellular systems.
Collapse
Affiliation(s)
- Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Daniel C Pan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Qin M Qi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Christopher J Kwon
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Junling Guo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
42
|
Mayuri PV, Bhatt A, Parameswaran R. Investigation of the potency of leukodepletion filter membranes immobilized with bovine serum albumin via polydopamine spacer. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-03515-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
43
|
Liu X, Gao Q, Zhang Y, Li Y, Li B. In Vivo Optofluidic Switch for Controlling Blood Microflow. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001414. [PMID: 32714772 PMCID: PMC7375249 DOI: 10.1002/advs.202001414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/12/2020] [Indexed: 05/13/2023]
Abstract
Control of blood microflow is crucial for the prevention and therapy of blood disorders, such as cardiovascular diseases and their complications. Conventional control strategies generally implant exogenous synthetic materials into blood vessels as labeling markers or actuating sources, which are invasive and incompatible with biological systems. Here, a label-free, noninvasive, and biocompatible device constructed from natural red blood cells (RBCs) for controlling blood microflow in vivo is reported. The RBCs, optically manipulated, arranged, and rotated using scanning optical tweezers, can function as an optofluidic switch for targeted switching, directional enrichment, dynamic redirecting, and rotary actuation of blood microflow inside zebrafish. The regulation precision of the switch is determined to be at the single-cell level, and the response time is measured as ≈200 ms using a streamline tracking method. This in vivo optofluidic switch may provide a biofriendly device for exploring blood microenvironments in a noncontact and noninvasive manner.
Collapse
Affiliation(s)
- Xiaoshuai Liu
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Qing Gao
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Yao Zhang
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Yuchao Li
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Baojun Li
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| |
Collapse
|
44
|
van Campen C(LM, Rowe PC, Visser FC. Cerebral Blood Flow Is Reduced in Severe Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients During Mild Orthostatic Stress Testing: An Exploratory Study at 20 Degrees of Head-Up Tilt Testing. Healthcare (Basel) 2020; 8:healthcare8020169. [PMID: 32545797 PMCID: PMC7349207 DOI: 10.3390/healthcare8020169] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction: In a study of 429 adults with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), we demonstrated that 86% had symptoms of orthostatic intolerance in daily life. Using extracranial Doppler measurements of the internal carotid and vertebral arteries during a 30-min head-up tilt to 70 degrees, 90% had an abnormal reduction in cerebral blood flow (CBF). A standard head-up tilt test of this duration might not be tolerated by the most severely affected bed-ridden ME/CFS patients. This study examined whether a shorter 15-min test at a lower 20 degree tilt angle would be sufficient to provoke reductions in cerebral blood flow in severe ME/CFS patients. Methods and results: Nineteen severe ME/CFS patients with orthostatic intolerance complaints in daily life were studied: 18 females. The mean (SD) age was 35(14) years, body surface area (BSA) was 1.8(0.2) m2 and BMI was 24.0(5.4) kg/m2. The median disease duration was 14 (IQR 5–18) years. Heart rate increased, and stroke volume index and end-tidal CO2 decreased significantly during the test (p ranging from <0.001 to <0.0001). The cardiac index decreased by 26(7)%: p < 0.0001. CBF decreased from 617(72) to 452(63) mL/min, a 27(5)% decline. All 19 severely affected ME/CFS patients met the criteria for an abnormal CBF reduction. Conclusions: Using a less demanding 20 degree tilt test for 15 min in severe ME/CFS patients resulted in a mean CBF decline of 27%. This is comparable to the mean 26% decline previously noted in less severely affected patients studied during a 30-min 70 degree head-up tilt. These observations have implications for the evaluation and treatment of severely affected individuals with ME/CFS.
Collapse
Affiliation(s)
| | - Peter C. Rowe
- Department of Paediatrics, John Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | | |
Collapse
|
45
|
Kenny M, Schoen I. A handshake between platelets and neutrophils might fuel deep vein thrombosis. Platelets 2020; 31:624-626. [DOI: 10.1080/09537104.2020.1769053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Martin Kenny
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
46
|
Zamani Kouhpanji MR, Stadler BJH. A Guideline for Effectively Synthesizing and Characterizing Magnetic Nanoparticles for Advancing Nanobiotechnology: A Review. SENSORS (BASEL, SWITZERLAND) 2020; 20:E2554. [PMID: 32365832 PMCID: PMC7248791 DOI: 10.3390/s20092554] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
The remarkable multimodal functionalities of magnetic nanoparticles, conferred by their size and morphology, are very important in resolving challenges slowing the progression of nanobiotechnology. The rapid and revolutionary expansion of magnetic nanoparticles in nanobiotechnology, especially in nanomedicine and therapeutics, demands an overview of the current state of the art for synthesizing and characterizing magnetic nanoparticles. In this review, we explain the synthesis routes for tailoring the size, morphology, composition, and magnetic properties of the magnetic nanoparticles. The pros and cons of the most popularly used characterization techniques for determining the aforementioned parameters, with particular focus on nanomedicine and biosensing applications, are discussed. Moreover, we provide numerous biomedical applications and highlight their challenges and requirements that must be met using the magnetic nanoparticles to achieve the most effective outcomes. Finally, we conclude this review by providing an insight towards resolving the persisting challenges and the future directions. This review should be an excellent source of information for beginners in this field who are looking for a groundbreaking start but they have been overwhelmed by the volume of literature.
Collapse
Affiliation(s)
- Mohammad Reza Zamani Kouhpanji
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bethanie J. H. Stadler
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Zamani Kouhpanji MR, Stadler BJH. Beyond the qualitative description of complex magnetic nanoparticle arrays using FORC measurement. NANO EXPRESS 2020. [DOI: 10.1088/2632-959x/ab844d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
48
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
49
|
Kim MG, Brown DK, Brand O. Nanofabrication for all-soft and high-density electronic devices based on liquid metal. Nat Commun 2020; 11:1002. [PMID: 32081910 PMCID: PMC7035367 DOI: 10.1038/s41467-020-14814-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/27/2020] [Indexed: 12/02/2022] Open
Abstract
Innovations in soft material synthesis and fabrication technologies have led to the development of integrated soft electronic devices. Such soft devices offer opportunities to interact with biological cells, mimicking their soft environment. However, existing fabrication technologies cannot create the submicron-scale, soft transducers needed for healthcare and medical applications involving single cells. This work presents a nanofabrication strategy to create submicron-scale, all-soft electronic devices based on eutectic gallium-indium alloy (EGaIn) using a hybrid method utilizing electron-beam lithography and soft lithography. The hybrid lithography process is applied to a biphasic structure, comprising a metallic adhesion layer coated with EGaIn, to create soft nano/microstructures embedded in elastomeric materials. Submicron-scale EGaIn thin-film patterning with feature sizes as small as 180 nm and 1 μm line spacing was achieved, resulting in the highest resolution EGaIn patterning technique to date. The resulting soft and stretchable EGaIn patterns offer a currently unrivaled combination of resolution, electrical conductivity, and electronic/wiring density.
Collapse
Affiliation(s)
- Min-Gu Kim
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Devin K Brown
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Oliver Brand
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
50
|
Brás MM, Radmacher M, Sousa SR, Granja PL. Melanoma in the Eyes of Mechanobiology. Front Cell Dev Biol 2020; 8:54. [PMID: 32117980 PMCID: PMC7027391 DOI: 10.3389/fcell.2020.00054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
Skin is the largest organ of the human body with several important functions that can be impaired by injury, genetic or chronic diseases. Among all skin diseases, melanoma is one of the most severe, which can lead to death, due to metastization. Mechanotransduction has a crucial role for motility, invasion, adhesion and metastization processes, since it deals with the response of cells to physical forces. Signaling pathways are important to understand how physical cues produced or mediated by the Extracellular Matrix (ECM), affect healthy and tumor cells. During these processes, several molecules in the nucleus and cytoplasm are activated. Melanocytes, keratinocytes, fibroblasts and the ECM, play a crucial role in melanoma formation. This manuscript will address the synergy among melanocytes, keratinocytes, fibroblasts cells and the ECM considering their mechanical contribution and relevance in this disease. Mechanical properties of melanoma cells can also be influenced by pigmentation, which can be associated with changes in stiffness. Mechanical changes can be related with the adhesion, migration, or invasiveness potential of melanoma cells promoting a high metastization capacity of this cancer. Mechanosensing, mechanotransduction, and mechanoresponse will be highlighted with respect to the motility, invasion, adhesion and metastization in melanoma cancer.
Collapse
Affiliation(s)
- M. Manuela Brás
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | | | - Susana R. Sousa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Pedro L. Granja
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| |
Collapse
|