1
|
Sammartino C, Pinchasik BE. Liquid Zener diodes. MATERIALS HORIZONS 2024; 11:4925-4931. [PMID: 39324522 DOI: 10.1039/d4mh00688g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The Zener diode (Z-diode) is often used as a voltage regulator, and is designed to operate in reverse breakdown. Namely, it can conduct current in the reverse direction when a certain voltage, known as the Zener voltage, is applied. Analogous to electric diodes, liquid diodes are microscale surface structures that promote spontaneous unidirectional liquid flow. In nature, they are used to increase water collection and uptake, for reproduction, and feeding. While such structures are usually deterministic, and used to guide liquids in specific directions while preventing backflow, the ability to render them responsive to external stimuli holds great potential for actuating and manipulating pump-free liquid flow in capillary networks. Inspired by electric Z-diodes, we demonstrate a local break in the diodic nature of flexible liquid diodes, in response to global compression or bending. Under specific conditions, flexural deformations trigger the formation of a capillary bridge in the reverse flow direction. This localized actuated flow is confined to where the liquid front is halted, preserving the integrity of liquid diodes elsewhere. This innovative concept showcases sequence-dependent liquid propagation within capillary networks, opening avenues for implementing network memory, reaction-dependent actuation, and the design of dynamic capillary networks.
Collapse
Affiliation(s)
- Camilla Sammartino
- Tel-Aviv University School of Mechanical Engineering Faculty of Engineering, 6997801 Tel-Aviv, Israel
| | - Bat-El Pinchasik
- Tel-Aviv University School of Mechanical Engineering Faculty of Engineering, 6997801 Tel-Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
2
|
Monroy-Romero AX, Nieto-Rivera B, Xiao W, Hautefeuille M. Microvascular Engineering for the Development of a Nonembedded Liver Sinusoid with a Lumen: When Endothelial Cells Do Not Lose Their Edge. ACS Biomater Sci Eng 2024. [PMID: 39390649 DOI: 10.1021/acsbiomaterials.4c00939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Microvascular engineering seeks to exploit known cell-cell and cell-matrix interactions in the context of vasculogenesis to restore homeostasis or disease development of reliable capillary models in vitro. However, current systems generally focus on recapitulating microvessels embedded in thick gels of extracellular matrix, overlooking the significance of discontinuous capillaries, which play a vital role in tissue-blood exchanges particularly in organs like the liver. In this work, we introduce a novel method to stimulate the spontaneous organization of endothelial cells into nonembedded microvessels. By creating an anisotropic micropattern at the edge of a development-like matrix dome using Marangoni flow, we achieved a long, nonrandom orientation of endothelial cells, laying a premise for stable lumenized microvessels. Our findings revealed a distinctive morphogenetic process leading to mature lumenized capillaries, demonstrated with both murine and human immortalized liver sinusoidal endothelial cell lines (LSECs). The progression of cell migration, proliferation, and polarization was clearly guided by the pattern, initiating the formation of a multicellular cord that caused a deformation spanning extensive regions and generated a wave-like folding of the gel, hinged at a laminin-depleted zone, enveloping the cord with gel proteins. This event marked the onset of lumenogenesis, regulated by the gradual apico-basal polarization of the wrapped cells, leading to the maturation of vessel tight junctions, matrix remodeling, and ultimately the formation of a lumen─recapitulating the development of vessels in vivo. Furthermore, we demonstrate that the process strongly relies on the initial gel edge topography, while the geometry of the vessels can be tuned from a curved to a straight structure. We believe that our facile engineering method, guiding an autonomous self-organization of vessels without the need for supporting cells or complex prefabricated scaffolds, holds promise for future integration into microphysiological systems featuring discontinuous, fenestrated capillaries.
Collapse
Affiliation(s)
- Ana Ximena Monroy-Romero
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, 03100 Mexico, México
- Laboratoire de Biologie du Développement (UMR 7622), Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France
| | - Brenda Nieto-Rivera
- Laboratoire de Biologie du Développement (UMR 7622), Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France
| | - Wenjin Xiao
- Laboratoire de Biologie du Développement (UMR 7622), Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
3
|
Su R, Ai Y, Wang J, Wu L, Sun H, Ding M, Xie R, Liang Q. Engineered Microfibers for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5823-5840. [PMID: 39145987 DOI: 10.1021/acsabm.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Hydrogel microfibers are hydrogel materials engineered into fiber structures. Techniques such as wet spinning, microfluidic spinning, and 3D bioprinting are often used to prepare microfibers due to their ability to precisely control the size, morphology, and structure of the microfibers. Microfibers with different structural morphologies have different functions; they provide a flow-through culture environment for cells to improve viability, and can also be used to induce the differentiation of cells such as skeletal muscle and cardiac muscle cells to eventually form functional organs in vitro through special morphologies. This Review introduces recent advances in microfluidics, 3D bioprinting, and wet spinning in the preparation of microfibers, focusing on the materials and fabrication methods. The applications of microfibers in tissue engineering are highlighted by summarizing their contributions in engineering biomimetic blood vessels, vascularized tissues, bone, heart, pancreas, kidney, liver, and fat. Furthermore, applications of engineered fibers in tissue repair and drug screening are also discussed.
Collapse
Affiliation(s)
- Riguga Su
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Jingyu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ruoxiao Xie
- Department of Materials, Design and Manufacturing Engineering, School of Engineering, University of Liverpool, Liverpool L69 3BX, U.K
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
4
|
Jiang W, Xu H, Gao Z, Wu Z, Zhao Z, Wang J, Wu Y, Ke H, Mao C, Wan M, Zhou M. Artificial Neutrophils Against Vascular Graft Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402768. [PMID: 38874399 PMCID: PMC11321623 DOI: 10.1002/advs.202402768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/12/2024] [Indexed: 06/15/2024]
Abstract
Efficient neutrophil migration to infection sites plays a vital role in the body's defense against bacterial infections and natural immune responses. Neutrophils have a short lifespan and cannot be mass-cultured in vitro. Therefore, developing more stable artificial neutrophils (AN) in a controllable manner has become a research focus. However, existing AN lack chemotaxis, which is the ability to migrate toward high-signal-concentration positions in a dynamic blood- flow environment. Supplying AN with chemotaxis is key to designing AN that are more similar to natural neutrophils in terms of morphology and function. In this study, micrometer-sized, spherical, biocompatible AN are developed. These AN consist of zeolitic imidazolate framework-8 nanoparticles encapsulating two enzymes, coacervate droplet frameworks, and outer phospholipid bilayers carrying enzymes. The AN exhibit responsiveness to elevated hydrogen peroxide levels at inflammation sites, actively chemotaxing toward these sites along concentration gradients. They also demonstrate effective combat against Staphylococcus aureus infections. The capabilities of the AN are further validated through in vitro experiments and in vivo evaluations using vascular graft infection models. This study replicates natural neutrophils in terms of chemical composition, functionality, and physiological impact. It introduces new ideas for advancing the development of advanced artificial cells.
Collapse
Affiliation(s)
- Wentao Jiang
- Department of Vascular SurgeryCardiovascular centerNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Huizi Xu
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
| | - Zheng Gao
- Department of Vascular SurgeryCardiovascular centerNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Ziyu Wu
- Department of Vascular SurgeryCardiovascular centerNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008China
- Institute for Life and HealthNanjing Drum Tower HospitalNanjing Normal UniversityNanjing210023China
| | - Zichun Zhao
- Department of Vascular SurgeryCardiovascular centerNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Jun Wang
- Department of Vascular SurgeryNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210008China
| | - Yawen Wu
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
| | - Haifeng Ke
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
- Institute for Life and HealthNanjing Drum Tower HospitalNanjing Normal UniversityNanjing210023China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
- Institute for Life and HealthNanjing Drum Tower HospitalNanjing Normal UniversityNanjing210023China
| | - Min Zhou
- Department of Vascular SurgeryCardiovascular centerNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008China
- Institute for Life and HealthNanjing Drum Tower HospitalNanjing Normal UniversityNanjing210023China
- Department of Vascular SurgeryNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210008China
| |
Collapse
|
5
|
De Ornelas B, Sucato V, Vadalà G, Buono A, Galassi AR. Myocardial Bridge and Atherosclerosis, an Intimal Relationship. Curr Atheroscler Rep 2024; 26:353-366. [PMID: 38822987 DOI: 10.1007/s11883-024-01219-1] [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] [Accepted: 05/18/2024] [Indexed: 06/03/2024]
Abstract
PURPOSE OF REVIEW This review investigates the relationship between myocardial bridges (MBs), intimal thickening in coronary arteries, and Atherosclerotic cardiovascular disease. It focuses on the role of mechanical forces, such as circumferential strain, in arterial wall remodeling and aims to clarify how MBs affect coronary artery pathology. REVIEW FINDINGS MBs have been identified as influential in modulating coronary artery intimal thickness, demonstrating a protective effect against thickening within the MB segment and an increase in thickness proximal to the MB. This is attributed to changes in mechanical stress and hemodynamics. Research involving arterial hypertension models and vein graft disease has underscored the importance of circumferential strain in vascular remodeling and intimal hyperplasia. Understanding the complex dynamics between MBs, mechanical strain, and vascular remodeling is crucial for advancing our knowledge of coronary artery disease mechanisms. This could lead to improved management strategies for cardiovascular diseases, highlighting the need for further research into MB-related vascular changes.
Collapse
Affiliation(s)
- Benjamin De Ornelas
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Vincenzo Sucato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Vadalà
- Division of Cardiology, University Hospital "P. Giaccone", Palermo, Italy
| | - Andrea Buono
- Interventional Cardiology Unit, Cardiovascular Department, Fondazione Poliambulanza Institute, Brescia, Italy
| | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
6
|
An J, Sugita N, Shinshi T. Microbubble detection on ultrasound imaging by utilizing phase patterned waves. Phys Med Biol 2024; 69:135003. [PMID: 38843808 DOI: 10.1088/1361-6560/ad5511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Objective.Super-resolution ultrasonography offers the advantage of visualization of intricate microvasculature, which is crucial for disease diagnosis. Mapping of microvessels is possible by localizing microbubbles (MBs) that act as contrast agents and tracking their location. However, there are limitations such as the low detectability of MBs and the utilization of a diluted concentration of MBs, leading to the extension of the acquisition time. We aim to enhance the detectability of MBs to reduce the acquisition time of acoustic data necessary for mapping the microvessels.Approach.We propose utilizing phase patterned waves (PPWs) characterized by spatially patterned phase distributions in the incident beam to achieve this. In contrast to conventional ultrasound irradiation methods, this irradiation method alters bubble interactions, enhancing the oscillation response of MBs and generating more significant scattered waves from specific MBs. This enhances the detectability of MBs, thereby enabling the detection of MBs that were undetectable by the conventional method. The objective is to maximize the overall detection of bubbles by utilizing ultrasound imaging with additional PPWs, including the conventional method. In this paper, we apply PPWs to ultrasound imaging simulations considering bubble-bubble interactions to elucidate the characteristics of PPWs and demonstrate their efficacy by employing PPWs on MBs fixed in a phantom by the experiment.Main results.By utilizing two types of PPWs in addition to the conventional ultrasound irradiation method, we confirmed the detection of up to 93.3% more MBs compared to those detected using the conventional method alone.Significance.Ultrasound imaging using additional PPWs made it possible to increase the number of detected MBs, which is expected to improve the efficiency of bubble detection.
Collapse
Affiliation(s)
- Junseok An
- Department of Mechanical Engineering, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Naohiro Sugita
- Laboratory for Future Interdisciplinary Research of Science and Technology (FIRST), Institute of Innovative Research (IIR), Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Tadahiko Shinshi
- Laboratory for Future Interdisciplinary Research of Science and Technology (FIRST), Institute of Innovative Research (IIR), Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
7
|
Ochieng BO, Zhao L, Ye Z. Three-Dimensional Bioprinting in Vascular Tissue Engineering and Tissue Vascularization of Cardiovascular Diseases. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:340-358. [PMID: 37885200 DOI: 10.1089/ten.teb.2023.0175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
In the 21st century, significant progress has been made in repairing damaged materials through material engineering. However, the creation of large-scale artificial materials still faces a major challenge in achieving proper vascularization. To address this issue, researchers have turned to biomaterials and three-dimensional (3D) bioprinting techniques, which allow for the combination of multiple biomaterials with improved mechanical and biological properties that mimic natural materials. Hydrogels, known for their ability to support living cells and biological components, have played a crucial role in this research. Among the recent developments, 3D bioprinting has emerged as a promising tool for constructing hybrid scaffolds. However, there are several challenges in the field of bioprinting, including the need for nanoscale biomimicry, the formulation of hydrogel blends, and the ongoing complexity of vascularizing biomaterials, which requires further research. On a positive note, 3D bioprinting offers a solution to the vascularization problem due to its precise spatial control, scalability, and reproducibility compared with traditional fabrication methods. This paper aims at examining the recent advancements in 3D bioprinting technology for creating blood vessels, vasculature, and vascularized materials. It provides a comprehensive overview of the progress made and discusses the limitations and challenges faced in current 3D bioprinting of vascularized tissues. In addition, the paper highlights the future research directions focusing on the development of 3D bioprinting techniques and bioinks for creating functional materials.
Collapse
Affiliation(s)
- Ben Omondi Ochieng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Leqian Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
- Department of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australia
| | - Zhiyi Ye
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
8
|
Zhang Y, Hu P, Li L, Cao R, Khadria A, Maslov K, Tong X, Zeng Y, Jiang L, Zhou Q, Wang LV. Ultrafast longitudinal imaging of haemodynamics via single-shot volumetric photoacoustic tomography with a single-element detector. Nat Biomed Eng 2024; 8:712-725. [PMID: 38036618 PMCID: PMC11136871 DOI: 10.1038/s41551-023-01149-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
Techniques for imaging haemodynamics use ionizing radiation or contrast agents or are limited by imaging depth (within approximately 1 mm), complex and expensive data-acquisition systems, or low imaging speeds, system complexity or cost. Here we show that ultrafast volumetric photoacoustic imaging of haemodynamics in the human body at up to 1 kHz can be achieved using a single laser pulse and a single element functioning as 6,400 virtual detectors. The technique, which does not require recalibration for different objects or during long-term operation, enables the longitudinal volumetric imaging of haemodynamics in vasculature a few millimetres below the skin's surface. We demonstrate this technique in vessels in the feet of healthy human volunteers by capturing haemodynamic changes in response to vascular occlusion. Single-shot volumetric photoacoustic imaging using a single-element detector may facilitate the early detection and monitoring of peripheral vascular diseases and may be advantageous for use in biometrics and point-of-care testing.
Collapse
Affiliation(s)
- Yide Zhang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Peng Hu
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Lei Li
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rui Cao
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Anjul Khadria
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Konstantin Maslov
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xin Tong
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yushun Zeng
- USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Laiming Jiang
- USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Qifa Zhou
- USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
9
|
Guo X, Yang Y, Tang J, Xiang J. Ephs in cancer progression: complexity and context-dependent nature in signaling, angiogenesis and immunity. Cell Commun Signal 2024; 22:299. [PMID: 38811954 PMCID: PMC11137953 DOI: 10.1186/s12964-024-01580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/23/2024] [Indexed: 05/31/2024] Open
Abstract
Eph receptors constitute the largest family of receptor tyrosine kinases, comprising 14 distinct members classified into two subgroups: EphAs and EphBs.. Despite their essential functions in normal physiological processes, accumulating evidence suggests that the involvement of the Eph family in cancer is characterized by a dual and often contradictory nature. Research indicates that Eph/ephrin bidirectional signaling influences cell-cell communication, subsequently regulating cell migration, adhesion, differentiation and proliferation. The contradictory functionalities may arise from the diversity of Eph signaling pathways and the heterogeneity of different cancer microenvironment. In this review, we aim to discuss the dual role of the Eph receptors in tumor development, attempting to elucidate the paradoxical functionality through an exploration of Eph receptor signaling pathways, angiogenesis, immune responses, and more. Our objective is to provide a comprehensive understanding of the molecular mechanisms underlying tumor development. Additionally, we will explore the evolving landscape of utilizing Eph receptors as potential targets for tumor therapy and diagnostic tools.
Collapse
Affiliation(s)
- Xiaoting Guo
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, the Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanyi Yang
- Health Management Center, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, the Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
- Department of Thoracic Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, the Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Hall E, Mendiola K, Lightsey NK, Hanjaya-Putra D. Mimicking blood and lymphatic vasculatures using microfluidic systems. BIOMICROFLUIDICS 2024; 18:031502. [PMID: 38726373 PMCID: PMC11081709 DOI: 10.1063/5.0175154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
The role of the circulatory system, containing the blood and lymphatic vasculatures, within the body, has become increasingly focused on by researchers as dysfunction of either of the systems has been linked to serious complications and disease. Currently, in vivo models are unable to provide the sufficient monitoring and level of manipulation needed to characterize the fluidic dynamics of the microcirculation in blood and lymphatic vessels; thus in vitro models have been pursued as an alternative model. Microfluidic devices have the required properties to provide a physiologically relevant circulatory system model for research as well as the experimental tools to conduct more advanced research analyses of microcirculation flow. In this review paper, the physiological behavior of fluid flow and electrical communication within the endothelial cells of the systems are detailed and discussed to highlight their complexities. Cell co-culturing methods and other relevant organ-on-a-chip devices will be evaluated to demonstrate the feasibility and relevance of the in vitro microfluidic model. Microfluidic systems will be determined as a noteworthy model that can display physiologically relevant flow of the cardiovascular and lymphatic systems, which will enable researchers to investigate the systems' prevalence in diseases and identify potential therapeutics.
Collapse
Affiliation(s)
- Eva Hall
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | - N. Keilany Lightsey
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | |
Collapse
|
11
|
Hu P, Du Y, Xu Y, Ye P, Xia J. The role of transcription factors in the pathogenesis and therapeutic targeting of vascular diseases. Front Cardiovasc Med 2024; 11:1384294. [PMID: 38745757 PMCID: PMC11091331 DOI: 10.3389/fcvm.2024.1384294] [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: 02/12/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Transcription factors (TFs) constitute an essential component of epigenetic regulation. They contribute to the progression of vascular diseases by regulating epigenetic gene expression in several vascular diseases. Recently, numerous regulatory mechanisms related to vascular pathology, ranging from general TFs that are continuously activated to histiocyte-specific TFs that are activated under specific circumstances, have been studied. TFs participate in the progression of vascular-related diseases by epigenetically regulating vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). The Krüppel-like family (KLF) TF family is widely recognized as the foremost regulator of vascular diseases. KLF11 prevents aneurysm progression by inhibiting the apoptosis of VSMCs and enhancing their contractile function. The presence of KLF4, another crucial member, suppresses the progression of atherosclerosis (AS) and pulmonary hypertension by attenuating the formation of VSMCs-derived foam cells, ameliorating endothelial dysfunction, and inducing vasodilatory effects. However, the mechanism underlying the regulation of the progression of vascular-related diseases by TFs has remained elusive. The present study categorized the TFs involved in vascular diseases and their regulatory mechanisms to shed light on the potential pathogenesis of vascular diseases, and provide novel insights into their diagnosis and treatment.
Collapse
Affiliation(s)
- Poyi Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Xu
- Institute of Reproduction Health Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Wille A, Weske S, von Wnuck Lipinski K, Wollnitzke P, Schröder NH, Thomas N, Nowak MK, Deister-Jonas J, Behr B, Keul P, Levkau B. Sphingosine-1-phosphate promotes osteogenesis by stimulating osteoblast growth and neovascularization in a vascular endothelial growth factor-dependent manner. J Bone Miner Res 2024; 39:357-372. [PMID: 38477738 PMCID: PMC11240155 DOI: 10.1093/jbmr/zjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 03/14/2024]
Abstract
Sphingosine-1-phosphate (S1P) plays multiple roles in bone metabolism and regeneration. Here, we have identified a novel S1P-regulated osteoanabolic mechanism functionally connecting osteoblasts (OBs) to the highly specialized bone vasculature. We demonstrate that S1P/S1PR3 signaling in OBs stimulates vascular endothelial growth factor a (VEGFa) expression and secretion to promote bone growth in an autocrine and boost osteogenic H-type differentiation of bone marrow endothelial cells in a paracrine manner. VEGFa-neutralizing antibodies and VEGF receptor inhibition by axitinib abrogated OB growth in vitro and bone formation in male C57BL/6J in vivo following S1P stimulation and S1P lyase inhibition, respectively. Pharmacological S1PR3 inhibition and genetic S1PR3 deficiency suppressed VEGFa production, OB growth in vitro, and inhibited H-type angiogenesis and bone growth in male mice in vivo. Together with previous work on the osteoanabolic functions of S1PR2 and S1PR3, our data suggest that S1P-dependent bone regeneration employs several nonredundant positive feedback loops between OBs and the bone vasculature. The identification of this yet unappreciated aspect of osteoanabolic S1P signaling may have implications for regular bone homeostasis as well as diseases where the bone microvasculature is affected such as age-related osteopenia and posttraumatic bone regeneration.
Collapse
Affiliation(s)
- Annalena Wille
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nadine Thomas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K Nowak
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jennifer Deister-Jonas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Behr
- Department of Plastic Surgery, University Hospital BG Bergmannsheil, 44789 Bochum, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
13
|
Makode S, Maurya S, Niknam SA, Mollocana-Lara E, Jaberi K, Faramarzi N, Tamayol A, Mortazavi M. Three dimensional (bio)printing of blood vessels: from vascularized tissues to functional arteries. Biofabrication 2024; 16:022005. [PMID: 38277671 DOI: 10.1088/1758-5090/ad22ed] [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: 06/28/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
Tissue engineering has emerged as a strategy for producing functional tissues and organs to treat diseases and injuries. Many chronic conditions directly or indirectly affect normal blood vessel functioning, necessary for material exchange and transport through the body and within tissue-engineered constructs. The interest in vascular tissue engineering is due to two reasons: (1) functional grafts can be used to replace diseased blood vessels, and (2) engineering effective vasculature within other engineered tissues enables connection with the host's circulatory system, supporting their survival. Among various practices, (bio)printing has emerged as a powerful tool to engineer biomimetic constructs. This has been made possible with precise control of cell deposition and matrix environment along with the advancements in biomaterials. (Bio)printing has been used for both engineering stand-alone vascular grafts as well as vasculature within engineered tissues for regenerative applications. In this review article, we discuss various conditions associated with blood vessels, the need for artificial blood vessels, the anatomy and physiology of different blood vessels, available 3D (bio)printing techniques to fabricate tissue-engineered vascular grafts and vasculature in scaffolds, and the comparison among the different techniques. We conclude our review with a brief discussion about future opportunities in the area of blood vessel tissue engineering.
Collapse
Affiliation(s)
- Shubham Makode
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Satyajit Maurya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Seyed A Niknam
- Department of Industrial Engineering, Western New England University, Springfield, MA, United States of America
| | - Evelyn Mollocana-Lara
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Kiana Jaberi
- Department of Nutritional Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Faramarzi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Mehdi Mortazavi
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, United States of America
| |
Collapse
|
14
|
Gaudreau LI, Stewart EJ. Vasculature-on-a-chip technologies as platforms for advanced studies of bacterial infections. BIOMICROFLUIDICS 2024; 18:021503. [PMID: 38560344 PMCID: PMC10977040 DOI: 10.1063/5.0179281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
Bacterial infections frequently occur within or near the vascular network as the vascular network connects organ systems and is essential in delivering and removing blood, essential nutrients, and waste products to and from organs. In turn, the vasculature plays a key role in the host immune response to bacterial infections. Technological advancements in microfluidic device design and development have yielded increasingly sophisticated and physiologically relevant models of the vasculature including vasculature-on-a-chip and organ-on-a-chip models. This review aims to highlight advancements in microfluidic device development that have enabled studies of the vascular response to bacteria and bacterial-derived molecules at or near the vascular interface. In the first section of this review, we discuss the use of parallel plate flow chambers and flow cells in studies of bacterial adhesion to the vasculature. We then highlight microfluidic models of the vasculature that have been utilized to study bacteria and bacterial-derived molecules at or near the vascular interface. Next, we review organ-on-a-chip models inclusive of the vasculature and pathogenic bacteria or bacterial-derived molecules that stimulate an inflammatory response within the model system. Finally, we provide recommendations for future research in advancing the understanding of host-bacteria interactions and responses during infections as well as in developing innovative antimicrobials for preventing and treating bacterial infections that capitalize on technological advancements in microfluidic device design and development.
Collapse
Affiliation(s)
- Lily Isabelle Gaudreau
- Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | | |
Collapse
|
15
|
McIntosh MC, Anglin DA, Robinson AT, Beck DT, Roberts MD. Making the case for resistance training in improving vascular function and skeletal muscle capillarization. Front Physiol 2024; 15:1338507. [PMID: 38405119 PMCID: PMC10884331 DOI: 10.3389/fphys.2024.1338507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Through decades of empirical data, it has become evident that resistance training (RT) can improve strength/power and skeletal muscle hypertrophy. Yet, until recently, vascular outcomes have historically been underemphasized in RT studies, which is underscored by several exercise-related reviews supporting the benefits of endurance training on vascular measures. Several lines of evidence suggest large artery diameter and blood flow velocity increase after a single bout of resistance exercise, and these events are mediated by vasoactive substances released from endothelial cells and myofibers (e.g., nitric oxide). Weeks to months of RT can also improve basal limb blood flow and arterial diameter while lowering blood pressure. Although several older investigations suggested RT reduces skeletal muscle capillary density, this is likely due to most of these studies being cross-sectional in nature. Critically, newer evidence from longitudinal studies contradicts these findings, and a growing body of mechanistic rodent and human data suggest skeletal muscle capillarity is related to mechanical overload-induced skeletal muscle hypertrophy. In this review, we will discuss methods used by our laboratories and others to assess large artery size/function and skeletal muscle capillary characteristics. Next, we will discuss data by our groups and others examining large artery and capillary responses to a single bout of resistance exercise and chronic RT paradigms. Finally, we will discuss RT-induced mechanisms associated with acute and chronic vascular outcomes.
Collapse
Affiliation(s)
| | - Derick A. Anglin
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | | | - Darren T. Beck
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Edward Via College of Osteopathic Medicine–Auburn Campus, Auburn, AL, United States
| | - Michael D. Roberts
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Edward Via College of Osteopathic Medicine–Auburn Campus, Auburn, AL, United States
| |
Collapse
|
16
|
Li R, Jiang M, Liu B, Jiang S, Chen C, Liang M, Qu L, Wang C, Zhao G, Hu Y, Wu D, Chu J, Li J. High-performance magnetic metal microrobot prepared by a two-photon polymerization and sintering method. LAB ON A CHIP 2024. [PMID: 38235769 DOI: 10.1039/d3lc01084h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Magnetically-actuated microrobots (MARs) exhibit great potential in biomedicine owing to their precise navigation, wireless actuation and remote operation in confined space. However, most previously explored MARs unfold the drawback of hypodynamic magnetic torque due to low magnetic content, leading to their limited applications in controlled locomotion in fast-flowing fluid and massive cargo carrying to the target position. Here, we report a high-performance pure-nickel magnetically-actuated microrobot (Ni-MAR), prepared by a femtosecond laser polymerization followed by sintering method. Our Ni-MAR possesses a high magnetic content (∼90 wt%), thus resulting in enhanced magnetic torque under low-strength rotating magnetic fields, which enables the microrobot to exhibit high-speed swimming and superior cargo carrying. The maximum velocity of our Ni-MAR, 12.5 body lengths per second, outperforms the velocity of traditional helical MARs. The high-speed Ni-MAR is capable of maintaining controlled locomotion in fast-flowing fluid. Moreover, the Ni-MAR with massive cargo carrying capability can push a 200-times heavier microcube with translation and rotation motion. A single cell and multiple cells can be transported facilely by a single Ni-MAR to the target position. This work provides a scheme for fabricating high-performance magnetic microrobots, which holds great promise for targeted therapy and drug delivery in vivo.
Collapse
Affiliation(s)
- Rui Li
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Modong Jiang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Bingrui Liu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Shaojun Jiang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Chao Chen
- Department of Materials Physics and New Energy Device, School of Materials Science and Engineering, Hefei University of Technology, Hefei 230009, China
| | - Mengxue Liang
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Lijie Qu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Chaowei Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Gang Zhao
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Yanlei Hu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Dong Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Jiaru Chu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| | - Jiawen Li
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
17
|
Hua R, Gao H, He C, Xin S, Wang B, Zhang S, Gao L, Tao Q, Wu W, Sun F, Xu J. An emerging view on vascular fibrosis molecular mediators and relevant disorders: from bench to bed. Front Cardiovasc Med 2023; 10:1273502. [PMID: 38179503 PMCID: PMC10764515 DOI: 10.3389/fcvm.2023.1273502] [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: 08/06/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Vascular fibrosis is a widespread pathologic condition that arises during vascular remodeling in cardiovascular dysfunctions. According to previous studies, vascular fibrosis is characterized by endothelial matrix deposition and vascular wall thickening. The RAAS and TGF-β/Smad signaling pathways have been frequently highlighted. It is, however, far from explicit in terms of understanding the cause and progression of vascular fibrosis. In this review, we collected and categorized a large number of molecules which influence the fibrosing process, in order to acquire a better understanding of vascular fibrosis, particularly of pathologic dysfunction. Furthermore, several mediators that prevent vascular fibrosis are discussed in depth in this review, with the aim that this will contribute to the future prevention and treatment of related conditions.
Collapse
Affiliation(s)
- Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Peking University, Beijing, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Qiang Tao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Wenqi Wu
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuan-Wu Hospital of Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Tscheuschner L, Tzafriri AR. Cardiovascular Tissue Engineering Models for Atherosclerosis Treatment Development. Bioengineering (Basel) 2023; 10:1373. [PMID: 38135964 PMCID: PMC10740643 DOI: 10.3390/bioengineering10121373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
In the early years of tissue engineering, scientists focused on the generation of healthy-like tissues and organs to replace diseased tissue areas with the aim of filling the gap between organ demands and actual organ donations. Over time, the realization has set in that there is an additional large unmet need for suitable disease models to study their progression and to test and refine different treatment approaches. Increasingly, researchers have turned to tissue engineering to address this need for controllable translational disease models. We review existing and potential uses of tissue-engineered disease models in cardiovascular research and suggest guidelines for generating adequate disease models, aimed both at studying disease progression mechanisms and supporting the development of dedicated drug-delivery therapies. This involves the discussion of different requirements for disease models to test drugs, nanoparticles, and drug-eluting devices. In addition to realistic cellular composition, the different mechanical and structural properties that are needed to simulate pathological reality are addressed.
Collapse
Affiliation(s)
- Linnea Tscheuschner
- Department of Vascular Surgery, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Abraham R. Tzafriri
- Department of Research and Innovation, CBSET Inc., Lexington, MA 02421, USA;
| |
Collapse
|
19
|
Wu Y, Zhao Y, Islam K, Zhou Y, Omidi S, Berdichevsky Y, Liu Y. Acoustofluidic Engineering of Functional Vessel-on-a-Chip. ACS Biomater Sci Eng 2023; 9:6273-6281. [PMID: 37787770 PMCID: PMC10646832 DOI: 10.1021/acsbiomaterials.3c00925] [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: 07/09/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
Construction of in vitro vascular models is of great significance to various biomedical research, such as pharmacokinetics and hemodynamics, and thus is an important direction in the tissue engineering field. In this work, a standing surface acoustic wave field was constructed to spatially arrange suspended endothelial cells into a designated acoustofluidic pattern. The cell patterning was maintained after the acoustic field was withdrawn within the solidified hydrogel. Then, interstitial flow was provided to activate vessel tube formation. In this way, a functional vessel network with specific vessel geometry was engineered on-chip. Vascular function, including perfusability and vascular barrier function, was characterized by microbead loading and dextran diffusion, respectively. A computational atomistic simulation model was proposed to illustrate how solutes cross the vascular membrane lipid bilayer. The reported acoustofluidic methodology is capable of facile and reproducible fabrication of the functional vessel network with specific geometry and high resolution. It is promising to facilitate the development of both fundamental research and regenerative therapy.
Collapse
Affiliation(s)
- Yue Wu
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yuwen Zhao
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Khayrul Islam
- Department
of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yuyuan Zhou
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Saeed Omidi
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yevgeny Berdichevsky
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yaling Liu
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
20
|
Murphy AR, Allenby MC. In vitro microvascular engineering approaches and strategies for interstitial tissue integration. Acta Biomater 2023; 171:114-130. [PMID: 37717711 DOI: 10.1016/j.actbio.2023.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
The increasing gap between clinical demand for tissue or organ transplants and the availability of donated tissue highlights the emerging opportunities for lab-grown or synthetically engineered tissue. While the field of tissue engineering has existed for nearly half a century, its clinical translation remains unrealised, in part, due to a limited ability to engineer sufficient vascular supply into fabricated tissue, which is necessary to enable nutrient and waste exchange, prevent cellular necrosis, and support tissue proliferation. Techniques to develop anatomically relevant, functional vascular networks in vitro have made significant progress in the last decade, however, the challenge now remains as to how best incorporate these throughout dense parenchymal tissue-like structures to address diffusion-limited development and allow for the fabrication of large-scale vascularised tissue. This review explores advances made in the laboratory engineering of vasculature structures and summarises recent attempts to integrate vascular networks together with sophisticated in vitro avascular tissue and organ-like structures. STATEMENT OF SIGNIFICANCE: The ability to grow full scale, functional tissue and organs in vitro is primarily limited by an inability to adequately diffuse oxygen and nutrients throughout developing cellularised structures, which generally results from the absence of perfusable vessel networks. Techniques to engineering both perfusable vascular networks and avascular miniaturised organ-like structures have recently increased in complexity, sophistication, and physiological relevance. However, integrating these two essential elements into a single functioning vascularised tissue structure represents a significant spatial and temporal engineering challenge which is yet to be surmounted. Here, we explore a range of vessel morphogenic phenomena essential for tissue-vascular co-development, as well as evaluate a range of recent noteworthy approaches for generating vascularised tissue products in vitro.
Collapse
Affiliation(s)
- A R Murphy
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia
| | - M C Allenby
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia; Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
21
|
Wei G, Kudo Y, Matsuda T, Wang ZJ, Mu QF, King DR, Nakajima T, Gong JP. Sustainable mechanochemical growth of double-network hydrogels supported by vascular-like perfusion. MATERIALS HORIZONS 2023; 10:4882-4891. [PMID: 37602807 DOI: 10.1039/d3mh01038d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Double-network (DN) gels are unique mechanochemical materials owing to their structures that can be dynamically remodelled during use. The mechanical energy applied to DN gels is efficiently transferred to the chemical bonds of the brittle network, generating mechanoradicals that initiate the polymerisation of pre-loaded monomers, thereby remodelling the materials. To attain continuous remodelling or growth in response to repetitive mechanical stimuli, a sustainable supply of chemical reagents to such dynamic materials is essential. In this study, inspired by the vascular perfusion transporting nutrients to cells, we constructed a circulatory system for a continuous supply of chemicals to channel-containing DN hydrogels (c-DN gels). The perfusion of monomer solutions through the channel and permeability of the c-DN gels not only replenishes the monomers consumed by the polymerisation but also replenishes the water loss caused by the surface evaporation of hydrogel, thereby freeing the mechanochemical process of DN gels from the constraints of the underwater environment. The facile chemical supply enabled us to modulate the mechanical enhancement of the c-DN gel and attain muscle-like strengthening under repeated mechanical training in deoxygenated air. We also studied the kinetics of polymer growth and strengthening and deciphered unique features of mechanochemical reaction in DN gels including the extremely long-living radicals and delayed mechanical strengthening.
Collapse
Affiliation(s)
- Gumi Wei
- Graduate School of Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Yumeko Kudo
- Graduate School of Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Takahiro Matsuda
- Graduate School of Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Zhi Jian Wang
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan.
| | - Qi Feng Mu
- Graduate School of Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Daniel R King
- Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Tasuku Nakajima
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan.
- Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Jian Ping Gong
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan.
- Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| |
Collapse
|
22
|
Carretero VJ, Ramos E, Segura-Chama P, Hernández A, Baraibar AM, Álvarez-Merz I, Muñoz FL, Egea J, Solís JM, Romero A, Hernández-Guijo JM. Non-Excitatory Amino Acids, Melatonin, and Free Radicals: Examining the Role in Stroke and Aging. Antioxidants (Basel) 2023; 12:1844. [PMID: 37891922 PMCID: PMC10603966 DOI: 10.3390/antiox12101844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The aim of this review is to explore the relationship between melatonin, free radicals, and non-excitatory amino acids, and their role in stroke and aging. Melatonin has garnered significant attention in recent years due to its diverse physiological functions and potential therapeutic benefits by reducing oxidative stress, inflammation, and apoptosis. Melatonin has been found to mitigate ischemic brain damage caused by stroke. By scavenging free radicals and reducing oxidative damage, melatonin may help slow down the aging process and protect against age-related cognitive decline. Additionally, non-excitatory amino acids have been shown to possess neuroprotective properties, including antioxidant and anti-inflammatory in stroke and aging-related conditions. They can attenuate oxidative stress, modulate calcium homeostasis, and inhibit apoptosis, thereby safeguarding neurons against damage induced by stroke and aging processes. The intracellular accumulation of certain non-excitatory amino acids could promote harmful effects during hypoxia-ischemia episodes and thus, the blockade of the amino acid transporters involved in the process could be an alternative therapeutic strategy to reduce ischemic damage. On the other hand, the accumulation of free radicals, specifically mitochondrial reactive oxygen and nitrogen species, accelerates cellular senescence and contributes to age-related decline. Recent research suggests a complex interplay between melatonin, free radicals, and non-excitatory amino acids in stroke and aging. The neuroprotective actions of melatonin and non-excitatory amino acids converge on multiple pathways, including the regulation of calcium homeostasis, modulation of apoptosis, and reduction of inflammation. These mechanisms collectively contribute to the preservation of neuronal integrity and functions, making them promising targets for therapeutic interventions in stroke and age-related disorders.
Collapse
Affiliation(s)
- Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Pedro Segura-Chama
- Investigador por México-CONAHCYT, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Huipulco, Tlalpan, Mexico City 14370, Mexico
| | - Adan Hernández
- Institute of Neurobiology, Universidad Nacional Autónoma of México, Juriquilla, Santiago de Querétaro 76230, Querétaro, Mexico
| | - Andrés M Baraibar
- Department of Neurosciences, Universidad del País Vasco UPV/EHU, Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, 48940 Leioa, Spain
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Francisco López Muñoz
- Faculty of Health Sciences, University Camilo José Cela, C/Castillo de Alarcón 49, Villanueva de la Cañada, 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute (i + 12), Avda. Córdoba, s/n, 28041 Madrid, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Health Research Institute, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - José M Solís
- Neurobiology-Research Service, Hospital Ramón y Cajal, Carretera de Colmenar Viejo, Km. 9, 28029 Madrid, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jesús M Hernández-Guijo
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Hospital Ramón y Cajal, Carretera de Colmenar Viejo, Km. 9, 28029 Madrid, Spain
| |
Collapse
|
23
|
Cohen R, Baruch ES, Cabilly I, Shapira A, Dvir T. Modified ECM-Based Bioink for 3D Printing of Multi-Scale Vascular Networks. Gels 2023; 9:792. [PMID: 37888365 PMCID: PMC10606913 DOI: 10.3390/gels9100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
The survival and function of tissues depend on appropriate vascularization. Blood vessels of the tissues supply oxygen, and nutrients and remove waste and byproducts. Incorporating blood vessels into engineered tissues is essential for overcoming diffusion limitations, improving tissue function, and thus facilitating the fabrication of thick tissues. Here, we present a modified ECM bioink, with enhanced mechanical properties and endothelial cell-specific adhesion motifs, to serve as a building material for 3D printing of a multiscale blood vessel network. The bioink is composed of natural ECM and alginate conjugated with a laminin adhesion molecule motif (YIGSR). The hybrid hydrogel was characterized for its mechanical properties, biochemical content, and ability to interact with endothelial cells. The pristine and modified hydrogels were mixed with induced pluripotent stem cells derived endothelial cells (iPSCs-ECs) and used to print large blood vessels with capillary beds in between.
Collapse
Affiliation(s)
- Roni Cohen
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ester-Sapir Baruch
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Department of Materials Science and Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Itai Cabilly
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
| | - Assaf Shapira
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
| | - Tal Dvir
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
24
|
Pineda-Castillo SA, Acar H, Detamore MS, Holzapfel GA, Lee CH. Modulation of Smooth Muscle Cell Phenotype for Translation of Tissue-Engineered Vascular Grafts. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:574-588. [PMID: 37166394 PMCID: PMC10618830 DOI: 10.1089/ten.teb.2023.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Translation of small-diameter tissue-engineered vascular grafts (TEVGs) for the treatment of coronary artery disease (CAD) remains an unfulfilled promise. This is largely due to the limited integration of TEVGs into the native vascular wall-a process hampered by the insufficient smooth muscle cell (SMC) infiltration and extracellular matrix deposition, and low vasoactivity. These processes can be promoted through the judicious modulation of the SMC toward a synthetic phenotype to promote remodeling and vascular integration; however, the expression of synthetic markers is often accompanied by a decrease in the expression of contractile proteins. Therefore, techniques that can precisely modulate the SMC phenotypical behavior could have the potential to advance the translation of TEVGs. In this review, we describe the phenotypic diversity of SMCs and the different environmental cues that allow the modulation of SMC gene expression. Furthermore, we describe the emerging biomaterial approaches to modulate the SMC phenotype in TEVG design and discuss the limitations of current techniques. In addition, we found that current studies in tissue engineering limit the analysis of the SMC phenotype to a few markers, which are often the characteristic of early differentiation only. This limited scope has reduced the potential of tissue engineering to modulate the SMC toward specific behaviors and applications. Therefore, we recommend using the techniques presented in this review, in addition to modern single-cell proteomics analysis techniques to comprehensively characterize the phenotypic modulation of SMCs. Expanding the holistic potential of SMC modulation presents a great opportunity to advance the translation of living conduits for CAD therapeutics.
Collapse
Affiliation(s)
- Sergio A. Pineda-Castillo
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Chung-Hao Lee
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
25
|
Bi CX, Jin KQ, Yan J, Qin Y, Hong F, Huang WH, Liu YL. Nanofiber-based Stretchable Electrodes for Oriented Culture and Mechanotransduction Monitoring of Smooth Muscle Cells. ACS Sens 2023; 8:3248-3256. [PMID: 37581426 DOI: 10.1021/acssensors.3c01135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Vascular smooth muscle cells (SMCs) are circumferentially oriented perpendicular to the blood vessel and maintain the contractile phenotype in physiological conditions. They can sense the mechanical forces of blood vessels expanding and contracting and convert them into biochemical signals to regulate vascular homeostasis. However, the real-time monitoring of mechanically evoked biochemical response while maintaining SMC oriented growth remains an important challenge. Herein, we developed a stretchable electrochemical sensor by electrospinning aligned and elastic polyurethane (PU) nanofibers on the surface of PDMS film and further modification of conductive polymer PEDOT:PSS-LiTFSI-CoPc (PPLC) on the nanofibers (denoted as PPLC/PU/PDMS). The aligned nanofibers on the electrode surface could guide the oriented growth of SMCs and maintain the contractile phenotype, and the modification of PPLC endowed the electrode with good electrochemical sensing performance and stability under mechanical deformation. By culturing cells on the electrode surface, the oriented growth of SMCs and real-time monitoring of stretch-induced H2O2 release were achieved. On this basis, the changes of H2O2 level released by SMCs under the pathology (hypertension) and intervention of natural product resveratrol were quantitatively monitored, which will be helpful to further understand the occurrence and development of vascular-related diseases and the mechanisms of pharmaceutical intervention.
Collapse
Affiliation(s)
- Chen-Xi Bi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Kai-Qi Jin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Yan
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Qin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Feng Hong
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yan-Ling Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
26
|
Kim SJ, Kim MG, Kim J, Jeon JS, Park J, Yi HG. Bioprinting Methods for Fabricating In Vitro Tubular Blood Vessel Models. CYBORG AND BIONIC SYSTEMS 2023; 4:0043. [PMID: 37533545 PMCID: PMC10393580 DOI: 10.34133/cbsystems.0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Dysfunctional blood vessels are implicated in various diseases, including cardiovascular diseases, neurodegenerative diseases, and cancer. Several studies have attempted to prevent and treat vascular diseases and understand interactions between these diseases and blood vessels across different organs and tissues. Initial studies were conducted using 2-dimensional (2D) in vitro and animal models. However, these models have difficulties in mimicking the 3D microenvironment in human, simulating kinetics related to cell activities, and replicating human pathophysiology; in addition, 3D models involve remarkably high costs. Thus, in vitro bioengineered models (BMs) have recently gained attention. BMs created through biofabrication based on tissue engineering and regenerative medicine are breakthrough models that can overcome limitations of 2D and animal models. They can also simulate the natural microenvironment in a patient- and target-specific manner. In this review, we will introduce 3D bioprinting methods for fabricating bioengineered blood vessel models, which can serve as the basis for treating and preventing various vascular diseases. Additionally, we will describe possible advancements from tubular to vascular models. Last, we will discuss specific applications, limitations, and future perspectives of fabricated BMs.
Collapse
Affiliation(s)
- Seon-Jin Kim
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Min-Gyun Kim
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
27
|
Alamilla-Sanchez ME, Alcala-Salgado MA, Cerezo Samperio B, Prado Lozano P, Diaz Garcia JD, Gonzalez Fuentes C, Yama Estrella MB, Morales Lopez EF. Advances in the Physiology of Transvascular Exchange and A New Look At Rational Fluid Prescription. Int J Gen Med 2023; 16:2753-2770. [PMID: 37408844 PMCID: PMC10319290 DOI: 10.2147/ijgm.s405926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/28/2023] [Indexed: 07/07/2023] Open
Abstract
The Starling principle is a model that explains the transvascular distribution of fluids essentially governed by hydrostatic and oncotic forces, which dynamically allow vascular refilling according to the characteristics of the blood vessel. However, careful analysis of fluid physiology has shown that the principle, while correct, is not complete. The revised Starling principle (Michel-Weinbaum model) provides relevant information on fluid kinetics. Special emphasis has been placed on the endothelial glycocalyx, whose subendothelial area allows a restricted oncotic pressure that limits the reabsorption of fluid from the interstitial space, so that transvascular refilling occurs mainly from the lymphatic vessels. The close correlation between pathological states of the endothelium (eg: sepsis, acute inflammation, or chronic kidney disease) and the prescription of fluids forces the physician to understand the dynamics of fluids in the organism; this will allow rational fluid prescriptions. A theory that integrates the physiology of exchange and transvascular refilling is the "microconstant model", whose variables include dynamic mechanisms that can explain edematous states, management of acute resuscitation, and type of fluids for common clinical conditions. The clinical-physiological integration of the concepts will be the hinges that allow a rational and dynamic prescription of fluids.
Collapse
Affiliation(s)
| | | | | | - Pamela Prado Lozano
- Department of Nephrology, Centro Medico Nacional “20 de Noviembre”, Mexico City, Mexico
| | | | | | | | | |
Collapse
|
28
|
Weigel C, Bellaci J, Spiegel S. Sphingosine-1-phosphate and its receptors in vascular endothelial and lymphatic barrier function. J Biol Chem 2023; 299:104775. [PMID: 37142226 PMCID: PMC10220486 DOI: 10.1016/j.jbc.2023.104775] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jacqueline Bellaci
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
29
|
Fritzen R, Davies A, Veenhuizen M, Campbell M, Pitt SJ, Ajjan RA, Stewart AJ. Magnesium Deficiency and Cardiometabolic Disease. Nutrients 2023; 15:nu15102355. [PMID: 37242238 DOI: 10.3390/nu15102355] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Magnesium (Mg2+) has many physiological functions within the body. These include important roles in maintaining cardiovascular functioning, where it contributes to the regulation of cardiac excitation-contraction coupling, endothelial functioning and haemostasis. The haemostatic roles of Mg2+ impact upon both the protein and cellular arms of coagulation. In this review, we examine how Mg2+ homeostasis is maintained within the body and highlight the various molecular roles attributed to Mg2+ in the cardiovascular system. In addition, we describe how nutritional and/or disease-associated magnesium deficiency, seen in some metabolic conditions, has the potential to influence cardiac and vascular outcomes. Finally, we also examine the potential for magnesium supplements to be employed in the prevention and treatment of cardiovascular disorders and in the management of cardiometabolic health.
Collapse
Affiliation(s)
- Remi Fritzen
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Amy Davies
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Miriam Veenhuizen
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Matthew Campbell
- School of Nursing and Health Sciences, University of Sunderland, Sunderland SR1 3DS, UK
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Samantha J Pitt
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Ramzi A Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Alan J Stewart
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| |
Collapse
|
30
|
Nguyen HT, Peirsman A, Tirpakova Z, Mandal K, Vanlauwe F, Maity S, Kawakita S, Khorsandi D, Herculano R, Umemura C, Yilgor C, Bell R, Hanson A, Li S, Nanda HS, Zhu Y, Najafabadi AH, Jucaud V, Barros N, Dokmeci MR, Khademhosseini A. Engineered Vasculature for Cancer Research and Regenerative Medicine. MICROMACHINES 2023; 14:978. [PMID: 37241602 PMCID: PMC10221678 DOI: 10.3390/mi14050978] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Engineered human tissues created by three-dimensional cell culture of human cells in a hydrogel are becoming emerging model systems for cancer drug discovery and regenerative medicine. Complex functional engineered tissues can also assist in the regeneration, repair, or replacement of human tissues. However, one of the main hurdles for tissue engineering, three-dimensional cell culture, and regenerative medicine is the capability of delivering nutrients and oxygen to cells through the vasculatures. Several studies have investigated different strategies to create a functional vascular system in engineered tissues and organ-on-a-chips. Engineered vasculatures have been used for the studies of angiogenesis, vasculogenesis, as well as drug and cell transports across the endothelium. Moreover, vascular engineering allows the creation of large functional vascular conduits for regenerative medicine purposes. However, there are still many challenges in the creation of vascularized tissue constructs and their biological applications. This review will summarize the latest efforts to create vasculatures and vascularized tissues for cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Zuzana Tirpakova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Florian Vanlauwe
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Rondinelli Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Bioengineering & Biomaterials Group, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil
| | - Christian Umemura
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Adrian Hanson
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Biomedical Engineering and Technology Laboratory, PDPM—Indian Institute of Information Technology Design Manufacturing, Jabalpur 482005, Madhya Pradesh, India
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Natan Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
31
|
Biophysical determinants of cancer organotropism. Trends Cancer 2023; 9:188-197. [PMID: 36494310 DOI: 10.1016/j.trecan.2022.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Metastasis remains the leading cause of cancer lethality. The 'seed/soil' hypothesis provides the framework to explain this cancer phenomenon where the concept of organotropism has been in part mechanistically explained by the properties of the tumor cells and their compatibility with the stromal environment of the distal site. The 'mechanical' hypothesis counters that non-random seeding is driven solely by the circulation patterns and vascular networks of organ systems. We incorporate concepts of mechanobiology and revisit the two hypotheses to provide additional insights into the mechanisms that regulate organ selection during metastatic outgrowth. We focus on the latter stages of the metastatic cascade and examine the role of the endothelium in regulating organ selectivity.
Collapse
|
32
|
Sturgess V, Azubuike UF, Tanner K. Vascular regulation of disseminated tumor cells during metastatic spread. BIOPHYSICS REVIEWS 2023; 4:011310. [PMID: 38510161 PMCID: PMC10903479 DOI: 10.1063/5.0106675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/17/2023] [Indexed: 03/22/2024]
Abstract
Cancer cells can travel to other organs via interconnected vascular systems to form new lesions in a process known as metastatic spread. Unfortunately, metastasis remains the leading cause of patient lethality. In recent years, it has been demonstrated that physical cues are just as important as chemical and genetic perturbations in driving changes in gene expression, cell motility, and survival. In this concise review, we focus on the physical cues that cancer cells experience as they migrate through the lymphatic and blood vascular networks. We also present an overview of steps that may facilitate organ specific metastasis.
Collapse
Affiliation(s)
- Victoria Sturgess
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Udochi F. Azubuike
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| |
Collapse
|
33
|
Watanabe T, Sassi S, Ulziibayar A, Hama R, Kitsuka T, Shinoka T. The Application of Porous Scaffolds for Cardiovascular Tissues. Bioengineering (Basel) 2023; 10:236. [PMID: 36829730 PMCID: PMC9952004 DOI: 10.3390/bioengineering10020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
As the number of arteriosclerotic diseases continues to increase, much improvement is still needed with treatments for cardiovascular diseases. This is mainly due to the limitations of currently existing treatment options, including the limited number of donor organs available or the long-term durability of the artificial organs. Therefore, tissue engineering has attracted significant attention as a tissue regeneration therapy in this area. Porous scaffolds are one of the effective methods for tissue engineering. However, it could be better, and its effectiveness varies depending on the tissue application. This paper will address the challenges presented by various materials and their combinations. We will also describe some of the latest methods for tissue engineering.
Collapse
Affiliation(s)
- Tatsuya Watanabe
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Salha Sassi
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Anudari Ulziibayar
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Rikako Hama
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Takahiro Kitsuka
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Surgery, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43205, USA
- Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
34
|
Bovolini A, Costa-Brito AR, Martins F, Furtado GE, Mendonça GV, Vila-Chã C. Impact of Exercise on Vascular Function in Middle-Aged and Older Adults: A Scoping Review. Sports (Basel) 2022; 10:sports10120208. [PMID: 36548505 PMCID: PMC9786288 DOI: 10.3390/sports10120208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
There is a substantial literature gap related to the vascular response to different types of exercise training in middle-aged and older populations. Thus, this scoping review aimed to examine the outcomes of controlled trials testing the long-term effects of exercise interventions on vascular function-related outcomes in middle-aged and older populations. The literature search was conducted following PRISMA guidelines. Data sources: five databases were used (EBSCO, MEDLINE, Web of Science, Science Direct, and Google Scholar). Eligibility criteria: controlled trials, published in the last 10 years, in English, containing well-described exercise interventions, reporting vascular quantitative effects of exercise in middle-aged and older people. A total of 62 publications were included. The studies included distinct types and intensities of exercise and were heterogeneous in volume and frequency. The assessed vascular outcomes also presented considerable variability. Overall, most studies reported positive effects of exercise on vascular function outcomes, regardless of exercise characteristics. Different exercise interventions can be applied to improve vascular function in middle-aged and older adults. Studies on combined and stretching exercises reported encouraging results in improving vascular function. Stretching exercises rise as an effective alternative in promoting vascular function among older adults, while combined exercise delivered promising vascular benefits in both populations.
Collapse
Affiliation(s)
- Antonio Bovolini
- Laboratory for the Evaluation of Sports Performance, Physical Exercise, and Health (LABMOV), Polytechnic of Guarda, 6300-559 Guarda, Portugal
- Research Center in Sports Sciences, Health Sciences, and Human Development (CIDESD), 5001-801 Vila Real, Portugal
- Correspondence: (A.B.); (C.V.-C.)
| | - Ana Raquel Costa-Brito
- Laboratory for the Evaluation of Sports Performance, Physical Exercise, and Health (LABMOV), Polytechnic of Guarda, 6300-559 Guarda, Portugal
| | - Faber Martins
- Laboratory for the Evaluation of Sports Performance, Physical Exercise, and Health (LABMOV), Polytechnic of Guarda, 6300-559 Guarda, Portugal
| | - Guilherme Eustáquio Furtado
- Laboratory for the Evaluation of Sports Performance, Physical Exercise, and Health (LABMOV), Polytechnic of Guarda, 6300-559 Guarda, Portugal
| | - Gonçalo V. Mendonça
- Neuromuscular Research Laboratory, Faculty of Human Motricity, University of Lisbon, Cruz Quebrada-Dafundo, 1495-751 Lisbon, Portugal
- Interdisciplinary Centre for the Study of Human Performance (CIPER), Faculty of Human Motricity, University of Lisbon, Cruz Quebrada-Dafundo, 1495-751 Lisbon, Portugal
| | - Carolina Vila-Chã
- Laboratory for the Evaluation of Sports Performance, Physical Exercise, and Health (LABMOV), Polytechnic of Guarda, 6300-559 Guarda, Portugal
- Research Center in Sports Sciences, Health Sciences, and Human Development (CIDESD), 5001-801 Vila Real, Portugal
- Correspondence: (A.B.); (C.V.-C.)
| |
Collapse
|
35
|
Yang GH, Kang D, An S, Ryu JY, Lee K, Kim JS, Song MY, Kim YS, Kwon SM, Jung WK, Jeong W, Jeon H. Advances in the development of tubular structures using extrusion-based 3D cell-printing technology for vascular tissue regenerative applications. Biomater Res 2022; 26:73. [PMID: 36471437 PMCID: PMC9720982 DOI: 10.1186/s40824-022-00321-2] [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: 07/31/2022] [Accepted: 11/13/2022] [Indexed: 12/11/2022] Open
Abstract
Until recent, there are no ideal small diameter vascular grafts available on the market. Most of the commercialized vascular grafts are used for medium to large-sized blood vessels. As a solution, vascular tissue engineering has been introduced and shown promising outcomes. Despite these optimistic results, there are limitations to commercialization. This review will cover the need for extrusion-based 3D cell-printing technique capable of mimicking the natural structure of the blood vessel. First, we will highlight the physiological structure of the blood vessel as well as the requirements for an ideal vascular graft. Then, the essential factors of 3D cell-printing including bioink, and cell-printing system will be discussed. Afterwards, we will mention their applications in the fabrication of tissue engineered vascular grafts. Finally, conclusions and future perspectives will be discussed.
Collapse
Affiliation(s)
- Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - SangHyun An
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jeong Yeop Ryu
- grid.258803.40000 0001 0661 1556Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, 130 Dongdeok‑ro, Jung‑gu, Daegu, 41944 South Korea
| | - KyoungHo Lee
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jun Sik Kim
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Moon-Yong Song
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Young-Sik Kim
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Sang-Mo Kwon
- grid.262229.f0000 0001 0719 8572Department of Physiology, School of Medicine, Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870 South Korea
| | - Won-Kyo Jung
- grid.412576.30000 0001 0719 8994Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Daeyeon-dong, Nam-gu, Busan, 48513 South Korea
| | - Woonhyeok Jeong
- grid.412091.f0000 0001 0669 3109Department of Plastic and Reconstructive Surgery, Dongsan Medical Center, Keimyung University College of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu, 42601 South Korea
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| |
Collapse
|
36
|
Abstract
This Review examines the state-of-the-art in the delivery of nucleic acid therapies that are directed to the vascular endothelium. First, we review the most important homeostatic functions and properties of the vascular endothelium and summarize the nucleic acid tools that are currently available for gene therapy and nucleic acid delivery. Second, we consider the opportunities available with the endothelium as a therapeutic target and the experimental models that exist to evaluate the potential of those opportunities. Finally, we review the progress to date from investigations that are directly targeting the vascular endothelium: for vascular disease, for peri-transplant therapy, for angiogenic therapies, for pulmonary endothelial disease, and for the blood-brain barrier, ending with a summary of the future outlook in this field.
Collapse
Affiliation(s)
| | | | | | - W. Mark Saltzman
- Department of Biomedical Engineering
- Department of Chemical & Environmental Engineering
- Department of Cellular & Molecular Physiology
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|
37
|
Huang S, Gao Y, Lv Y, Wang Y, Cao Y, Zhao W, Zuo D, Mu H, Hua Y. Applications of Nano/Micromotors for Treatment and Diagnosis in Biological Lumens. MICROMACHINES 2022; 13:mi13101780. [PMID: 36296133 PMCID: PMC9610721 DOI: 10.3390/mi13101780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 06/01/2023]
Abstract
Natural biological lumens in the human body, such as blood vessels and the gastrointestinal tract, are important to the delivery of materials. Depending on the anatomic features of these biological lumens, the invention of nano/micromotors could automatically locomote targeted sites for disease treatment and diagnosis. These nano/micromotors are designed to utilize chemical, physical, or even hybrid power in self-propulsion or propulsion by external forces. In this review, the research progress of nano/micromotors is summarized with regard to treatment and diagnosis in different biological lumens. Challenges to the development of nano/micromotors more suitable for specific biological lumens are discussed, and the overlooked biological lumens are indicated for further studies.
Collapse
Affiliation(s)
- Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yinghua Gao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yinghao Cao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Weisong Zhao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Dongqing Zuo
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Shanghai Bone Tumor Institution, Shanghai 201620, China
| |
Collapse
|
38
|
Urbanczyk M, Zbinden A, Schenke-Layland K. Organ-specific endothelial cell heterogenicity and its impact on regenerative medicine and biomedical engineering applications. Adv Drug Deliv Rev 2022; 186:114323. [PMID: 35568103 DOI: 10.1016/j.addr.2022.114323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) are a key cellular component of the vascular system as they form the inner lining of the blood vessels. Recent findings highlight that ECs express extensive phenotypic heterogenicity when following the vascular tree from the major vasculature down to the organ capillaries. However, in vitro models, used for drug development and testing, or to study the role of ECs in health and disease, rarely acknowledge this EC heterogenicity. In this review, we highlight the main differences between different EC types, briefly summarize their different characteristics and focus on the use of ECs in in vitro models. We introduce different approaches on how ECs can be utilized in co-culture test systems in the field of brain, pancreas, and liver research to study the role of the endothelium in health and disease. Finally, we discuss potential improvements to current state-of-the-art in vitro models and future directions.
Collapse
|
39
|
Wu YY, Shan SK, Lin X, Xu F, Zhong JY, Wu F, Duan JY, Guo B, Li FXZ, Wang Y, Zheng MH, Xu QS, Lei LM, Ou-Yang WL, Tang KX, Li CC, Ullah MHE, Yuan LQ. Cellular Crosstalk in the Vascular Wall Microenvironment: The Role of Exosomes in Vascular Calcification. Front Cardiovasc Med 2022; 9:912358. [PMID: 35677687 PMCID: PMC9168031 DOI: 10.3389/fcvm.2022.912358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/02/2022] [Indexed: 07/20/2023] Open
Abstract
Vascular calcification is prevalent in aging, diabetes, chronic kidney disease, cardiovascular disease, and certain genetic disorders. However, the pathogenesis of vascular calcification is not well-understood. It has been progressively recognized that vascular calcification depends on the bidirectional interactions between vascular cells and their microenvironment. Exosomes are an essential bridge to mediate crosstalk between cells and organisms, and thus they have attracted increased research attention in recent years. Accumulating evidence has indicated that exosomes play an important role in cardiovascular disease, especially in vascular calcification. In this review, we introduce vascular biology and focus on the crosstalk between the different vessel layers and how their interplay controls the process of vascular calcification.
Collapse
Affiliation(s)
- Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yu Zhong
- Department of Nuclear Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ou-Yang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
40
|
Locatelli L, Castiglioni S, Maier JAM. From Cultured Vascular Cells to Vessels: The Cellular and Molecular Basis of Vascular Dysfunction in Space. Front Bioeng Biotechnol 2022; 10:862059. [PMID: 35480977 PMCID: PMC9036997 DOI: 10.3389/fbioe.2022.862059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Life evolved on this planet under the pull of gravity, shielded from radiation by the magnetosphere and shaped by circadian rhythms due to Earth’s rotation on its axis. Once living beings leave such a protective environment, adaptive responses are activated to grant survival. In view of long manned mission out of Earth’s orbit, it is relevant to understand how humans adapt to space and if the responses activated might reveal detrimental in the long run. Here we review present knowledge about the effects on the vessels of various extraterrestrial factors on humans as well as in vivo and in vitro experimental models. It emerges that the vasculature activates complex adaptive responses finalized to supply oxygen and nutrients to all the tissues and to remove metabolic waste and carbon dioxide. Most studies point to oxidative stress and mitochondrial dysfunction as mediators of vascular alterations in space. Unraveling the cellular and molecular mechanisms involved in these adaptive processes might offer hints to design proper and personalized countermeasures to predict a safe future in space.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Jeanette A M Maier
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy.,Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa), Università di Milano, Milan, Italy
| |
Collapse
|
41
|
Salg GA, Poisel E, Neulinger-Munoz M, Gerhardus J, Cebulla D, Bludszuweit-Philipp C, Vieira V, Nickel F, Herr I, Blaeser A, Giese NA, Hackert T, Kenngott HG. Toward 3D-bioprinting of an endocrine pancreas: A building-block concept for bioartificial insulin-secreting tissue. J Tissue Eng 2022; 13:20417314221091033. [PMID: 35462988 PMCID: PMC9024162 DOI: 10.1177/20417314221091033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional bioprinting of an endocrine pancreas is a promising future
curative treatment for patients with insulin secretion deficiency. In this
study, we present an end-to-end concept from the molecular to the macroscopic
level. Building-blocks for a hybrid scaffold device of hydrogel and
functionalized polycaprolactone were manufactured by 3D-(bio)printing.
Pseudoislet formation from INS-1 cells after bioprinting resulted in a viable
and proliferative experimental model. Transcriptomics showed an upregulation of
proliferative and ß-cell-specific signaling cascades, downregulation of
apoptotic pathways, overexpression of extracellular matrix proteins, and VEGF
induced by pseudoislet formation and 3D-culture. Co-culture with endothelial
cells created a natural cellular niche with enhanced insulin secretion after
glucose stimulation. Survival and function of pseudoislets after explantation
and extensive scaffold vascularization of both hydrogel and heparinized
polycaprolactone were demonstrated in vivo. Computer
simulations of oxygen, glucose and insulin flows were used to evaluate scaffold
architectures and Langerhans islets at a future perivascular transplantation
site.
Collapse
Affiliation(s)
- Gabriel A Salg
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Eric Poisel
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Neulinger-Munoz
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Department of Dermatology and Allergy, University Hospital LMU Munich, Munich, Germany
| | - Jamina Gerhardus
- Technical University of Darmstadt, Institute for BioMedical Printing Technology, Darmstadt, Germany
| | - Daniel Cebulla
- ASD Advanced Simulation and Design GmbH, Rostock, Germany
| | | | | | - Felix Nickel
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Blaeser
- Technical University of Darmstadt, Institute for BioMedical Printing Technology, Darmstadt, Germany
| | - Nathalia A Giese
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Hannes G Kenngott
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
Guarracino F, Bertini P. Perioperative hypotension: causes and remedies. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2022; 2:17. [PMCID: PMC10245539 DOI: 10.1186/s44158-022-00045-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 06/24/2023]
Abstract
Background Arterial hypotension is common in patients during surgery and those admitted to the intensive care unit (ICU) postoperatively. Perioperative arterial hypotension reportedly significantly affects surgical patients’ outcomes. Blood pressure level is the most crucial factor that influences organ perfusion. Hypoperfusion and organ dysfunction are correlated based on their severity associated with hypotension. As several factors can cause intraoperative hypotension, anesthetists must promptly identify the etiology for appropriate treatment and revert the patient’s hemodynamic profile. Objectives This review discusses the concept of perioperative hypotension, identifies its effects in clinical situations, and provides remedies and techniques to predict and avoid its significant consequences. Conclusions The primary determinant of organ perfusion is blood pressure. On the other hand, profound hypotension is common in surgical patients and is connected to hypoperfusion and organ failure. Currently, hypotension is addressed once low blood pressure levels are recorded. Early detection of oncoming hypotension or its clinical prediction is of paramount importance in allowing the clinician to treat hypotension and reduce the incidence and length of hypotensive episodes promptly and aggressively.
Collapse
Affiliation(s)
- Fabio Guarracino
- Department of Anaesthesia and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, Via Paradisa 2, 56123 Pisa, Italy
| | - Pietro Bertini
- Department of Anaesthesia and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, Via Paradisa 2, 56123 Pisa, Italy
| |
Collapse
|
43
|
Nuñez-Borque E, Fernandez-Bravo S, Yuste-Montalvo A, Esteban V. Pathophysiological, Cellular, and Molecular Events of the Vascular System in Anaphylaxis. Front Immunol 2022; 13:836222. [PMID: 35371072 PMCID: PMC8965328 DOI: 10.3389/fimmu.2022.836222] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Anaphylaxis is a systemic hypersensitivity reaction that can be life threatening. Mechanistically, it results from the immune activation and release of a variety of mediators that give rise to the signs and symptoms of this pathological event. For years, most of the research in anaphylaxis has focused on the contribution of the immune component. However, approaches that shed light on the participation of other cellular and molecular agents are necessary. Among them, the vascular niche receives the various signals (e.g., histamine) that elicit the range of anaphylactic events. Cardiovascular manifestations such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and cardiac alterations are crucial in the pathophysiology of anaphylaxis and are highly involved to the development of the most severe cases. Specifically, the endothelium, vascular smooth muscle cells, and their molecular signaling outcomes play an essential role downstream of the immune reaction. Therefore, in this review, we synthesized the vascular changes observed during anaphylaxis as well as its cellular and molecular components. As the risk of anaphylaxis exists both in clinical procedures and in routine life, increasing our knowledge of the vascular physiology and their molecular mechanism will enable us to improve the clinical management and how to treat or prevent anaphylaxis. Key Message Anaphylaxis, the most severe allergic reaction, involves a variety of immune and non-immune molecular signals that give rise to its pathophysiological manifestations. Importantly, the vascular system is engaged in processes relevant to anaphylactic events such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and decreased cardiac output. The novelty of this review focuses on the fact that new studies will greatly improve the understanding of anaphylaxis when viewed from a vascular molecular angle and specifically from the endothelium. This knowledge will improve therapeutic options to treat or prevent anaphylaxis.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sergio Fernandez-Bravo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alma Yuste-Montalvo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain
| |
Collapse
|
44
|
Jin J, Liu J, Luo Y, He H, Zheng X, Zheng C, Huang Y, Chen Y. High fructose induces dysfunctional vasodilatation via PP2A-mediated eNOS Ser1177 dephosphorylation. Nutr Metab (Lond) 2022; 19:24. [PMID: 35331293 PMCID: PMC8944156 DOI: 10.1186/s12986-022-00659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background Processed foods are popular and contain large amounts of industrial fructose, which changes people’s diet and exacerbates the negative health effects of high fructose. Several studies have shown that excessive intake of fructose has a major impact on vascular disease. However, the mechanism of the effect of high fructose on blood vessels is currently unclear. Methods The effect of fructose on the vasodilatation of isolated thoracic aortic rings was observed by using wire myography in wild-type (WT) mice. Cell viability and nitric oxide (NO) production were assessed by the corresponding kits in mouse vascular endothelial cells. The effect of fructose on endothelial nitric oxide synthase (eNOS) and protein phosphatase 2A (PP2A) and their changes in phosphorylation were detected by using Western blots. Moreover, a PP2A inhibitor (okadaic acid, OA) was used to evaluate the relationship between fructose and PP2A. Furthermore, PP2ACα endothelial-specific knockout (PP2A cKO) mice were used to detect the vasodilatation of in vitro fructose-incubated thoracic aortic rings by using wire myography. Results High fructose induced endothelium-dependent dysfunctional vasodilatation. High fructose reduced acetylcholine (Ach)-induced vasodilation but did not affect sodium nitroprusside (SNP)-induced vasodilation. Accordingly, NO production and the phosphorylation level of eNOS at serine (Ser) 1177 (P-eNOS) in vascular endothelial cells were remarkably reduced without changes in cell viability. The expression of protein phosphatase 2A catalytic subunit (PP2AC) was increased and the expression of phosphorylated PP2AC (P-PP2A, tyrosine [Tyr] 307) was significantly decreased. Nevertheless, these effects were reversed by OA. Moreover, knockout of the PP2A gene could recover the response of vessels to Ach under high fructose stimulation. Conclusions Our observations demonstrate an underlying mechanism of fructose-induced dysfunctional vasodilatation. Fructose could activate PP2A, which leads to decrease in the phosphorylation of eNOS at Ser1177 and the reduction of NO release, thus leading to the occurrence of endothelium-dependent dysfunctional vasodilatation.
Collapse
Affiliation(s)
- Jiaqi Jin
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Jingya Liu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Yong Luo
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Hong He
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Xinyue Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chaoyang Zheng
- Department of Cardiology, The Second Clinical Medical College and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yi Huang
- Department of Stomatology, The First Affiliated Hospital, The School of Dental Medicine, Jinan University, No. 613W. Huangpu Avenue, Guangzhou, 510630, China.
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China.
| |
Collapse
|
45
|
Novak R, Hrkac S, Salai G, Bilandzic J, Mitar L, Grgurevic L. The Role of ADAMTS-4 in Atherosclerosis and Vessel Wall Abnormalities. J Vasc Res 2022; 59:69-77. [PMID: 35051931 DOI: 10.1159/000521498] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/13/2021] [Indexed: 11/19/2022] Open
Abstract
Extracellular matrix proteins are regulated by metzincin proteases, like the disintegrin metalloproteinases with thrombospondin motifs (ADAMTS) family members. This review focuses on the emerging role which ADAMTS-4 might play in vascular pathology, which has implications for atherosclerosis and vessel wall abnormalities, as well as for the resulting diseases, such as cardiovascular and cerebrovascular disease, aortic aneurysms, and dissections. Major substrates of ADAMTS-4 are proteoglycans expressed physiologically in smooth muscle cells of blood vessels. Good examples are versican and aggrecan, principal vessel wall proteoglycans that are targeted by ADAMTS-4, driving blood vessel atrophy, which is why this metzincin protease was implicated in the pathophysiology of vascular diseases with an atherosclerotic background. Despite emerging evidence, it is important not to exaggerate the role of ADAMTS-4 as it is likely only a small piece of the complex atherosclerosis puzzle and one that could be functionally redundant due to its high structural similarity to other ADAMTS family members. The therapeutic potential of inhibiting ADAMTS-4 to halt the progression of vascular disease after initialization of treatment is unlikely. However, it is not excluded that it might find a purpose as a biomarker of vascular disease, possibly as an indicator in a larger cytokine panel.
Collapse
Affiliation(s)
- Rudjer Novak
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Stela Hrkac
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Grgur Salai
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Teaching Institute of Emergency Medicine of the City of Zagreb, Zagreb, Croatia
| | - Josko Bilandzic
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Luka Mitar
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Lovorka Grgurevic
- Department of Proteomics, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Department of Anatomy, "Drago Perovic," School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
46
|
Benjamins JW, Yeung MW, van de Vegte YJ, Said MA, van der Linden T, Ties D, Juarez-Orozco LE, Verweij N, van der Harst P. Genomic insights in ascending aortic size and distensibility. EBioMedicine 2022; 75:103783. [PMID: 34968759 PMCID: PMC8718733 DOI: 10.1016/j.ebiom.2021.103783] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alterations in the anatomic and biomechanical properties of the ascending aorta (AAo) can give rise to various vascular pathologies. The aim of the current study is to gain additional insights in the biology of the AAo size and function. METHODS We developed an AI based analysis pipeline for the segmentation of the AAo, and the extraction of AAO parameters. We then performed genome-wide association studies of AAo maximum area, AAo minimum area and AAo distensibility in up to 37,910 individuals from the UK Biobank. Variants that were significantly associated with AAo phenotypes were used as instrumental variables in Mendelian randomization analyses to investigate potential causal relationships with coronary artery disease, myocardial infarction, stroke and aneurysms. FINDINGS Genome-wide association studies revealed a total of 107 SNPs in 78 loci. We annotated 101 candidate genes involved in various biological processes, including connective tissue development (THSD4 and COL6A3). Mendelian randomization analyses showed a causal association with aneurysm development, but not with other vascular diseases. INTERPRETATION We identified 78 loci that provide insights into mechanisms underlying AAo size and function in the general population and provide genetic evidence for their role in aortic aneurysm development.
Collapse
Affiliation(s)
- Jan Walter Benjamins
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| | - Ming Wai Yeung
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands; Department of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherland
| | - Yordi J van de Vegte
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - M Abdullah Said
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Thijs van der Linden
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Daan Ties
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Luis E Juarez-Orozco
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands; Department of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherland
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands; Department of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherland
| |
Collapse
|
47
|
Wang Y, Huang Y, Du R, Ge S, Li Y, Wang G, Wang Y, Yin T. The crosstalk between arterial components and the bioresorbable, 3-D printed poly-l-lactic acid scaffolds. Biomater Sci 2022; 10:5121-5133. [DOI: 10.1039/d2bm00732k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bioresorbable scaffolds (BRS) are designed to provide a temporary support that subsequently leaves behind native vessels after their complete degradation. The accumulation of mechanical changes influences the vascular histological characteristics...
Collapse
|
48
|
Mikołajczyk K, Spyt D, Zielińska W, Żuryń A, Faisal I, Qamar M, Świniarski P, Grzanka A, Gagat M. The Important Role of Endothelium and Extracellular Vesicles in the Cellular Mechanism of Aortic Aneurysm Formation. Int J Mol Sci 2021; 22:ijms222313157. [PMID: 34884962 PMCID: PMC8658239 DOI: 10.3390/ijms222313157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Homeostasis is a fundamental property of biological systems consisting of the ability to maintain a dynamic balance of the environment of biochemical processes. The action of endogenous and exogenous factors can lead to internal balance disorder, which results in the activation of the immune system and the development of inflammatory response. Inflammation determines the disturbances in the structure of the vessel wall, connected with the change in their diameter. These disorders consist of accumulation in the space between the endothelium and the muscle cells of low-density lipoproteins (LDL), resulting in the formation of fatty streaks narrowing the lumen and restricting the blood flow in the area behind the structure. The effect of inflammation may also be pathological dilatation of the vessel wall associated with the development of aneurysms. Described disease entities strongly correlate with the increased migration of immune cells. Recent scientific research indicates the secretion of specific vesicular structures during migration activated by the inflammation. The review focuses on the link between endothelial dysfunction and the inflammatory response and the impact of these processes on the development of disease entities potentially related to the secretion of extracellular vesicles (EVs).
Collapse
Affiliation(s)
- Klaudia Mikołajczyk
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Dominika Spyt
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Wioletta Zielińska
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Agnieszka Żuryń
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Inaz Faisal
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Murtaz Qamar
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Piotr Świniarski
- Department of Urology and Andrology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland;
| | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
- Correspondence:
| |
Collapse
|
49
|
Schwickert A, Henrich W, Vogel M, Melchior K, Ehrlich L, Ochs M, Braun T. Placenta Percreta Presents with Neoangiogenesis of Arteries with Von Willebrand Factor-Negative Endothelium. Reprod Sci 2021; 29:1136-1144. [PMID: 34766259 PMCID: PMC8907099 DOI: 10.1007/s43032-021-00763-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 10/02/2021] [Indexed: 11/28/2022]
Abstract
In placenta percreta cases, large vessels are present on the precrete surface area. As these vessels are not found in normal placentation, we examined their histological structure for features that might explain the pathogenesis of neoangiogenesis induced by placenta accreta spectrum disorders (PAS). In two patients with placenta percreta (FIGO grade 3a) of the anterior uterine wall, one strikingly large vessel of 2 cm length was excised. The samples were formalin fixed and paraffin-embedded. Gomori trichrome staining was used to evaluate the muscular layers and Weigert-Van Gieson staining for elastic fibers. Immunohistochemical staining of the vessel endothelium was performed for Von Willebrand factor (VWF), platelet endothelial cell adhesion molecule (CD31), Ephrin B2, and EPH receptor B4. The structure of the vessel walls appeared artery-like. The vessel of patient one further exhibited an unorderly muscular layer and a lack of elastic laminae, whereas these features appeared normal in the vessel of the other patient. The endothelium of both vessels stained VWF-negative and CD31-positive. In conclusion, this study showed VWF-negative vessel endothelia of epiplacental arteries in placenta accreta spectrum. VWF is known to regulate artery formation, as the absence of VWF has been shown to cause enhanced vascularization. Therefore, we suppose that PAS provokes increased vascularization through suppression of VWF. This process might be associated with the immature vessel architecture as found in one of the vessels and Ephrin B2 and EPH receptor B4 negativity of both artery-like vessels. The underlying pathomechanism needs to be evaluated in a greater set of patients.
Collapse
Affiliation(s)
- Alexander Schwickert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Obstetrics, Berlin, Germany.
| | - Wolfgang Henrich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Berlin, Germany
| | - Martin Vogel
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pathology, Pediatric Pathology and Placentology, Berlin, Germany
| | - Kerstin Melchior
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Obstetrics, Berlin, Germany
| | - Loreen Ehrlich
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Obstetrics, Berlin, Germany
| | - Matthias Ochs
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Functional Anatomy, Berlin, Germany
| | - Thorsten Braun
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Obstetrics, Berlin, Germany
| |
Collapse
|
50
|
Hauser PV, Chang HM, Nishikawa M, Kimura H, Yanagawa N, Hamon M. Bioprinting Scaffolds for Vascular Tissues and Tissue Vascularization. Bioengineering (Basel) 2021; 8:178. [PMID: 34821744 PMCID: PMC8615027 DOI: 10.3390/bioengineering8110178] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
In recent years, tissue engineering has achieved significant advancements towards the repair of damaged tissues. Until this day, the vascularization of engineered tissues remains a challenge to the development of large-scale artificial tissue. Recent breakthroughs in biomaterials and three-dimensional (3D) printing have made it possible to manipulate two or more biomaterials with complementary mechanical and/or biological properties to create hybrid scaffolds that imitate natural tissues. Hydrogels have become essential biomaterials due to their tissue-like physical properties and their ability to include living cells and/or biological molecules. Furthermore, 3D printing, such as dispensing-based bioprinting, has progressed to the point where it can now be utilized to construct hybrid scaffolds with intricate structures. Current bioprinting approaches are still challenged by the need for the necessary biomimetic nano-resolution in combination with bioactive spatiotemporal signals. Moreover, the intricacies of multi-material bioprinting and hydrogel synthesis also pose a challenge to the construction of hybrid scaffolds. This manuscript presents a brief review of scaffold bioprinting to create vascularized tissues, covering the key features of vascular systems, scaffold-based bioprinting methods, and the materials and cell sources used. We will also present examples and discuss current limitations and potential future directions of the technology.
Collapse
Affiliation(s)
- Peter Viktor Hauser
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (P.V.H.); (H.-M.C.); (N.Y.)
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91343, USA
| | - Hsiao-Min Chang
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (P.V.H.); (H.-M.C.); (N.Y.)
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91343, USA
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo 113-8654, Japan;
| | - Hiroshi Kimura
- Department of Mechanical Engineering, School of Engineering, Tokai University, Isehara 259-1207, Japan;
| | - Norimoto Yanagawa
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (P.V.H.); (H.-M.C.); (N.Y.)
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91343, USA
| | - Morgan Hamon
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (P.V.H.); (H.-M.C.); (N.Y.)
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91343, USA
| |
Collapse
|