1
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
2
|
Ebrahimi A, Ghorbanpoor H, Apaydın E, Demir Cevizlidere B, Özel C, Tüfekçioğlu E, Koç Y, Topal AE, Tomsuk Ö, Güleç K, Abdullayeva N, Kaya M, Ghorbani A, Şengel T, Benzait Z, Uysal O, Eker Sarıboyacı A, Doğan Güzel F, Singh H, Hassan S, Ankara H, Pat S, Atalay E, Avci H. Convenient rapid prototyping microphysiological niche for mimicking liver native basement membrane: Liver sinusoid on a chip. Colloids Surf B Biointerfaces 2024; 245:114292. [PMID: 39383580 DOI: 10.1016/j.colsurfb.2024.114292] [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: 07/08/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Liver is responsible for the metabolization processes of up to 90 % of compounds and toxins in the body. Therefore liver-on-a-chip systems, as an in vitro promising cell culture platform, have great importance for fundamental science and drug development. In most of the liver-on-a-chip studies, seeding cells on both sides of a porous membrane, which represents the basement membrane, fail to resemble the native characteristics of biochemical, biophysical, and mechanical properties. In this study, polycarbonate (PC) and polyethylene terephthalate (PET) membranes were coated with gelatin to address this issue by accurately mimicking the native basement membrane present in the space of Disse. Various coating methods were used, including doctor blade, gel micro-injection, electrospinning, and spin coating. Spin coating was demonstrated to be the most effective technique owing to the ability to produce thin gel thickness with desirable surface roughness for cell interactions on both sides of the membrane. HepG2 and EA.HY926 cells were seeded on the upper and bottom sides of the gelatin-coated PET membrane and cultured on-chip for 7 days. Cell viability increased from 90 % to 95 %, while apoptotic index decreased. Albumin secretion notably rose between days 1-7 and 4-7, while GST-α secretion decreased from day 1 to day 7. In conclusion, the optimized spin coating process reported here can effectively modify the membranes to better mimic the native basement membrane niche characteristics.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Hamed Ghorbanpoor
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biomedical Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Elif Apaydın
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Bahar Demir Cevizlidere
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ceren Özel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Emre Tüfekçioğlu
- Department of Industrial Design/Department of Industrial Design, Faculty of Architecture and Design, Eskisehir Technical University, Eskisehir, Türkiye
| | - Yücel Koç
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ahmet Emin Topal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Türkiye
| | - Özlem Tomsuk
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Mechanical Engineering, Middle East Technical University, Ankara 06800, Türkiye
| | - Kadri Güleç
- Department of Analytical Chemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Nuran Abdullayeva
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Murat Kaya
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Aynaz Ghorbani
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Tayfun Şengel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Central Research Laboratory Research and Application Center (ARUM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Zineb Benzait
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ayla Eker Sarıboyacı
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Fatma Doğan Güzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, Ankara, Türkiye
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Hüseyin Ankara
- Mining Engineering Department, Engineering-Architecture Faculty, Eskisehir Osmangazi University, Meşelik Campus, Eskisehir 26480, Türkiye
| | - Suat Pat
- Eskisehir Osmangazi University, Faculty of Science, Department of Physics, Eskisehir TR-26040, Türkiye
| | - Eray Atalay
- Department of Ophthalmology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye; Translational Medicine Research and Clinical Center (TATUM), Eskisehir Osmangazi University, Eskisehir, Türkiye.
| |
Collapse
|
3
|
Modi PS, Singh A, Chaturvedi A, Agarwal S, Dutta R, Nayak R, Singh AK. Tissue chips as headway model and incitement technology. Synth Syst Biotechnol 2024; 10:86-101. [PMID: 39286054 PMCID: PMC11403008 DOI: 10.1016/j.synbio.2024.08.007] [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: 01/12/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Tissue on a chip or organ-on-chip (OOC) is a technology that's dignified to form a transformation in drug discovery through the use of advanced platforms. These are 3D in-vitro cell culture models that mimic micro-environment of human organs or tissues on artificial microstructures built on a portable microfluidic chip without involving sacrificial humans or animals. This review article aims to offer readers a thorough and insightful understanding of technology. It begins with an in-depth understanding of chip design and instrumentation, underlining its pivotal role and the imperative need for its development in the modern scientific landscape. The review article explores into the myriad applications of OOC technology, showcasing its transformative impact on fields such as radiobiology, drug discovery and screening, and its pioneering use in space research. In addition to highlighting these diverse applications, the article provides a critical analysis of the current challenges that OOC technology faces. It examines both the biological and technical limitations that hinder its progress and efficacy and discusses the potential advancements and innovations that could drive the OOC technology forward. Through this comprehensive review, readers will gain a deep appreciation of the significance, capabilities, and evolving landscape of OOC technology.
Collapse
Affiliation(s)
- Prerna Suchitan Modi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Abhishek Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Awyang Chaturvedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shailly Agarwal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Raghav Dutta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Alok Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
4
|
Florido MHC, Ziats NP. Endothelial dysfunction and cardiovascular diseases: The role of human induced pluripotent stem cells and tissue engineering. J Biomed Mater Res A 2024; 112:1286-1304. [PMID: 38230548 DOI: 10.1002/jbm.a.37669] [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: 08/28/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Cardiovascular disease (CVD) remains to be the leading cause of death globally today and therefore the need for the development of novel therapies has become increasingly important in the cardiovascular field. The mechanism(s) behind the pathophysiology of CVD have been laboriously investigated in both stem cell and bioengineering laboratories. Scientific breakthroughs have paved the way to better mimic cell types of interest in recent years, with the ability to generate any cell type from reprogrammed human pluripotent stem cells. Mimicking the native extracellular matrix using both organic and inorganic biomaterials has allowed full organs to be recapitulated in vitro. In this paper, we will review techniques from both stem cell biology and bioengineering which have been fruitfully combined and have fueled advances in the cardiovascular disease field. We will provide a brief introduction to CVD, reviewing some of the recent studies as related to the role of endothelial cells and endothelial cell dysfunction. Recent advances and the techniques widely used in both bioengineering and stem cell biology will be discussed, providing a broad overview of the collaboration between these two fields and their overall impact on tissue engineering in the cardiovascular devices and implications for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mary H C Florido
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Nicholas P Ziats
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Biomedical Engineering and Anatomy, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Martier A, Chen Z, Schaps H, Mondrinos MJ, Fang JS. Capturing physiological hemodynamic flow and mechanosensitive cell signaling in vessel-on-a-chip platforms. Front Physiol 2024; 15:1425618. [PMID: 39135710 PMCID: PMC11317428 DOI: 10.3389/fphys.2024.1425618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Recent advances in organ chip (or, "organ-on-a-chip") technologies and microphysiological systems (MPS) have enabled in vitro investigation of endothelial cell function in biomimetic three-dimensional environments under controlled fluid flow conditions. Many current organ chip models include a vascular compartment; however, the design and implementation of these vessel-on-a-chip components varies, with consequently varied impact on their ability to capture and reproduce hemodynamic flow and associated mechanosensitive signaling that regulates key characteristics of healthy, intact vasculature. In this review, we introduce organ chip and vessel-on-a-chip technology in the context of existing in vitro and in vivo vascular models. We then briefly discuss the importance of mechanosensitive signaling for vascular development and function, with focus on the major mechanosensitive signaling pathways involved. Next, we summarize recent advances in MPS and organ chips with an integrated vascular component, with an emphasis on comparing both the biomimicry and adaptability of the diverse approaches used for supporting and integrating intravascular flow. We review current data showing how intravascular flow and fluid shear stress impacts vessel development and function in MPS platforms and relate this to existing work in cell culture and animal models. Lastly, we highlight new insights obtained from MPS and organ chip models of mechanosensitive signaling in endothelial cells, and how this contributes to a deeper understanding of vessel growth and function in vivo. We expect this review will be of broad interest to vascular biologists, physiologists, and cardiovascular physicians as an introduction to organ chip platforms that can serve as viable model systems for investigating mechanosensitive signaling and other aspects of vascular physiology.
Collapse
Affiliation(s)
- A. Martier
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Z. Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - H. Schaps
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - M. J. Mondrinos
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - J. S. Fang
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
6
|
Yang S, Luo J, Zou W, Zhu Q, Cen J, Gao Q. Research trends in vascular chips from 2012 to 2022: a bibliometrix and visualized analysis. Front Bioeng Biotechnol 2024; 12:1409467. [PMID: 39055344 PMCID: PMC11269249 DOI: 10.3389/fbioe.2024.1409467] [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: 03/30/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024] Open
Abstract
Objective The vascular chip has emerged as a significant research tool, garnering increasing interest and exploration. We utilize bibliometric techniques to analyze literature from the Web of Science (WOS) database, focusing on core journal publications. The aim is to provide a systematic review and prospective outlook on research trends within the vascular chip field, delving into current dynamics and highlighting areas for further investigation. Methods We retrieved articles, proceedings papers, and early-access publications related to vascular chips published between January 2012 and December 2022 reported by Web of Science Core Collection (WoSCC) in 2023. Scientific bibliometric analysis was performed using R-bibliometrix, CiteSpace, VOSviewer, and Microsoft Excel software tools. Results A total of 456 publications were obtained, including 444 articles, 11 proceedings papers, and one early-access article. These originated from 167 academic journals and 751 research institutions across 44 countries/regions. The United States contributed the majority of publications (41%), with Harvard University leading in contributions (6.6%). Lab on a Chip was the top journal in terms of publications. Notably, authors Jeon NL and Huh D wielded significant influence, with the former being the most prolific author and the latter garnering the most citations. Recent research has predominantly focused on angiogenesis in relation to endothelial cells. Conclusion This scientometric investigation comprehensively surveys literature on vascular chips over past decade, providing valuable insights for scholars in the field. Our study reveals global increases in publications, with endothelial cells and angiogenesis being primary research focuses. This trend will persist, drawing continued attention from researchers.
Collapse
Affiliation(s)
- Song Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jing Luo
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Wanwan Zou
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
- School of Medicine South China University of Technology, Guangzhou, China
| | - Qikun Zhu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jianzheng Cen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Laboratory of Artificial Intelligence and 3D Technologies for Cardiovascular Diseases, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiang Gao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Laboratory of Artificial Intelligence and 3D Technologies for Cardiovascular Diseases, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
7
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
8
|
Li X, Zhu H, Gu B, Yao C, Gu Y, Xu W, Zhang J, He J, Liu X, Li D. Advancing Intelligent Organ-on-a-Chip Systems with Comprehensive In Situ Bioanalysis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305268. [PMID: 37688520 DOI: 10.1002/adma.202305268] [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: 06/02/2023] [Revised: 08/03/2023] [Indexed: 09/11/2023]
Abstract
In vitro models are essential to a broad range of biomedical research, such as pathological studies, drug development, and personalized medicine. As a potentially transformative paradigm for 3D in vitro models, organ-on-a-chip (OOC) technology has been extensively developed to recapitulate sophisticated architectures and dynamic microenvironments of human organs by applying the principles of life sciences and leveraging micro- and nanoscale engineering capabilities. A pivotal function of OOC devices is to support multifaceted and timely characterization of cultured cells and their microenvironments. However, in-depth analysis of OOC models typically requires biomedical assay procedures that are labor-intensive and interruptive. Herein, the latest advances toward intelligent OOC (iOOC) systems, where sensors integrated with OOC devices continuously report cellular and microenvironmental information for comprehensive in situ bioanalysis, are examined. It is proposed that the multimodal data in iOOC systems can support closed-loop control of the in vitro models and offer holistic biomedical insights for diverse applications. Essential techniques for establishing iOOC systems are surveyed, encompassing in situ sensing, data processing, and dynamic modulation. Eventually, the future development of iOOC systems featuring cross-disciplinary strategies is discussed.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hui Zhu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bingsong Gu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Cong Yao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuyang Gu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wangkai Xu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jia Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyu Liu
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, M5S 3G8, Canada
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
9
|
Marder M, Remmert C, Perschel JA, Otgonbayar M, von Toerne C, Hauck S, Bushe J, Feuchtinger A, Sheikh B, Moussus M, Meier M. Stem cell-derived vessels-on-chip for cardiovascular disease modeling. Cell Rep 2024; 43:114008. [PMID: 38536819 DOI: 10.1016/j.celrep.2024.114008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
The metabolic syndrome is accompanied by vascular complications. Human in vitro disease models are hence required to better understand vascular dysfunctions and guide clinical therapies. Here, we engineered an open microfluidic vessel-on-chip platform that integrates human pluripotent stem cell-derived endothelial cells (SC-ECs). The open microfluidic design enables seamless integration with state-of-the-art analytical technologies, including single-cell RNA sequencing, proteomics by mass spectrometry, and high-resolution imaging. Beyond previous systems, we report SC-EC maturation by means of barrier formation, arterial toning, and high nitric oxide synthesis levels under gravity-driven flow. Functionally, we corroborate the hallmarks of early-onset atherosclerosis with low sample volumes and cell numbers under flow conditions by determining proteome and secretome changes in SC-ECs stimulated with oxidized low-density lipoprotein and free fatty acids. More broadly, our organ-on-chip platform enables the modeling of patient-specific human endothelial tissue and has the potential to become a general tool for animal-free vascular research.
Collapse
Affiliation(s)
- Maren Marder
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Caroline Remmert
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Julius A Perschel
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | | | | | - Stefanie Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Judith Bushe
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Bilal Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany; Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Michel Moussus
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany; Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
10
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [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: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
11
|
Zhou Q, Liu Q, Wang Y, Chen J, Schmid O, Rehberg M, Yang L. Bridging Smart Nanosystems with Clinically Relevant Models and Advanced Imaging for Precision Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308659. [PMID: 38282076 PMCID: PMC11005737 DOI: 10.1002/advs.202308659] [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: 11/12/2023] [Indexed: 01/30/2024]
Abstract
Intracellular delivery of nano-drug-carriers (NDC) to specific cells, diseased regions, or solid tumors has entered the era of precision medicine that requires systematic knowledge of nano-biological interactions from multidisciplinary perspectives. To this end, this review first provides an overview of membrane-disruption methods such as electroporation, sonoporation, photoporation, microfluidic delivery, and microinjection with the merits of high-throughput and enhanced efficiency for in vitro NDC delivery. The impact of NDC characteristics including particle size, shape, charge, hydrophobicity, and elasticity on cellular uptake are elaborated and several types of NDC systems aiming for hierarchical targeting and delivery in vivo are reviewed. Emerging in vitro or ex vivo human/animal-derived pathophysiological models are further explored and highly recommended for use in NDC studies since they might mimic in vivo delivery features and fill the translational gaps from animals to humans. The exploration of modern microscopy techniques for precise nanoparticle (NP) tracking at the cellular, organ, and organismal levels informs the tailored development of NDCs for in vivo application and clinical translation. Overall, the review integrates the latest insights into smart nanosystem engineering, physiological models, imaging-based validation tools, all directed towards enhancing the precise and efficient intracellular delivery of NDCs.
Collapse
Affiliation(s)
- Qiaoxia Zhou
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Forensic PathologyWest China School of Preclinical and Forensic MedicineSichuan UniversityNo. 17 Third Renmin Road NorthChengdu610041China
- Burning Rock BiotechBuilding 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou510300China
| | - Qiongliang Liu
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Thoracic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Yan Wang
- Qingdao Central HospitalUniversity of Health and Rehabilitation Sciences (Qingdao Central Medical Group)Qingdao266042China
| | - Jie Chen
- Department of Respiratory MedicineNational Key Clinical SpecialtyBranch of National Clinical Research Center for Respiratory DiseaseXiangya HospitalCentral South UniversityChangshaHunan410008China
- Center of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center for Respiratory Diseases in Hunan ProvinceChangshaHunan410008China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory DiseaseChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaHunan410008P. R. China
| | - Otmar Schmid
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| | - Markus Rehberg
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| | - Lin Yang
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| |
Collapse
|
12
|
Svensson E, von Mentzer U, Stubelius A. Achieving Precision Healthcare through Nanomedicine and Enhanced Model Systems. ACS MATERIALS AU 2024; 4:162-173. [PMID: 38496040 PMCID: PMC10941278 DOI: 10.1021/acsmaterialsau.3c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 03/19/2024]
Abstract
The ability to customize medical choices according to an individual's genetic makeup and biomarker patterns marks a significant advancement toward overall improved healthcare for both individuals and society at large. By transitioning from the conventional one-size-fits-all approach to tailored treatments that can account for predispositions of different patient populations, nanomedicines can be customized to target the specific molecular underpinnings of a patient's disease, thus mitigating the risk of collateral damage. However, for these systems to reach their full potential, our understanding of how nano-based therapeutics behave within the intricate human body is necessary. Effective drug administration to the targeted organ or pathological niche is dictated by properties such as nanocarrier (NC) size, shape, and targeting abilities, where understanding how NCs change their properties when they encounter biomolecules and phenomena such as shear stress in flow remains a major challenge. This Review specifically focuses on vessel-on-a-chip technology that can provide increased understanding of NC behavior in blood and summarizes the specialized environment of the joint to showcase advanced tissue models as approaches to address translational challenges. Compared to conventional cell studies or animal models, these advanced models can integrate patient material for full customization. Combining such models with nanomedicine can contribute to making personalized medicine achievable.
Collapse
Affiliation(s)
| | | | - Alexandra Stubelius
- Division of Chemical Biology,
Department of Life Sciences, Chalmers University
of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
13
|
Ranjbar J, Yang Y, Harper AGS. Developing human tissue engineered arterial constructs to simulate human in vivo thrombus formation. Platelets 2023; 34:2153823. [PMID: 36550074 DOI: 10.1080/09537104.2022.2153823] [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: 12/24/2022]
Abstract
Thrombus formation is highly dependent upon the physico-chemical environment in which it is triggered. Our ability to understand how thrombus formation is initiated, regulated, and resolved in the human body is dependent upon our ability to replicate the mechanical and biological properties of the arterial wall. Current in vitro thrombosis models principally use reductionist approaches to model the complex biochemical and cellular milieu present in the arterial wall, and so researcher have favored the use of in vivo models. The field of vascular tissue engineering has developed a range of techniques for culturing artificial human arteries for use as vascular grafts. These techniques therefore provide a basis for developing more sophisticated 3D replicas of the arterial wall that can be used in in vitro thrombosis models. In this review, we consider how tissue engineering approaches can be used to generate 3D models of the arterial wall that improve upon current in vivo and in vitro approaches. We consider the current benefits and limitations of reported 3D tissue engineered models and consider what additional evidence is required to validate them as alternatives to current in vivo models.
Collapse
Affiliation(s)
| | - Ying Yang
- School of Pharmacy & Bioengineering, Keele University, Keele, UK
| | | |
Collapse
|
14
|
Thompson CL, Hopkins T, Bevan C, Screen HRC, Wright KT, Knight MM. Human vascularised synovium-on-a-chip: a mechanically stimulated, microfluidic model to investigate synovial inflammation and monocyte recruitment. Biomed Mater 2023; 18:065013. [PMID: 37703884 DOI: 10.1088/1748-605x/acf976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/13/2023] [Indexed: 09/15/2023]
Abstract
Healthy synovium is critical for joint homeostasis. Synovial inflammation (synovitis) is implicated in the onset, progression and symptomatic presentation of arthritic joint diseases such as rheumatoid arthritis and osteoarthritis. Thus, the synovium is a promising target for the development of novel, disease-modifying therapeutics. However, target exploration is hampered by a lack of good pre-clinical models that accurately replicate human physiology and that are developed in a way that allows for widespread uptake. The current study presents a multi-channel, microfluidic, organ-on-a-chip (OOAC) model, comprising a 3D configuration of the human synovium and its associated vasculature, with biomechanical and inflammatory stimulation, built upon a commercially available OOAC platform. Healthy human fibroblast-like synoviocytes (hFLS) were co-cultured with human umbilical vein endothelial cells (HUVECs) with appropriate matrix proteins, separated by a flexible, porous membrane. The model was developed within the Emulate organ-chip platform enabling the application of physiological biomechanical stimulation in the form of fluid shear and cyclic tensile strain. The hFLS exhibited characteristic morphology, cytoskeletal architecture and matrix protein deposition. Synovial inflammation was initiated through the addition of interleukin-1β(IL-1β) into the synovium channel resulting in the increased secretion of inflammatory and catabolic mediators, interleukin-6 (IL-6), prostaglandin E2 (PGE2), matrix metalloproteinase 1 (MMP-1), as well as the synovial fluid constituent protein, hyaluronan. Enhanced expression of the inflammatory marker, intercellular adhesion molecule-1 (ICAM-1), was observed in HUVECs in the vascular channel, accompanied by increased attachment of circulating monocytes. This vascularised human synovium-on-a-chip model recapitulates a number of the functional characteristics of both healthy and inflamed human synovium. Thus, this model offers the first human synovium organ-chip suitable for widespread adoption to understand synovial joint disease mechanisms, permit the identification of novel therapeutic targets and support pre-clinical testing of therapies.
Collapse
Affiliation(s)
- Clare L Thompson
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Timothy Hopkins
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, United Kingdom
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire, United Kingdom
| | - Catrin Bevan
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Hazel R C Screen
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Karina T Wright
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, United Kingdom
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire, United Kingdom
| | - Martin M Knight
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
15
|
Kawakita S, Li S, Nguyen HT, Maity S, Haghniaz R, Bahari J, Yu N, Mandal K, Bandaru P, Mou L, Ermis M, Khalil E, Khosravi S, Peirsman A, Nasiri R, Adachi A, Nakayama A, Bell R, Zhu Y, Jucaud V, Dokmeci MR, Khademhosseini A. Rapid integration of screen-printed electrodes into thermoplastic organ-on-a-chip devices for real-time monitoring of trans-endothelial electrical resistance. Biomed Microdevices 2023; 25:37. [PMID: 37740819 DOI: 10.1007/s10544-023-00669-9] [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: 07/24/2023] [Indexed: 09/25/2023]
Abstract
Trans-endothelial electrical resistance (TEER) is one of the most widely used indicators to quantify the barrier integrity of endothelial layers. Over the last decade, the integration of TEER sensors into organ-on-a-chip (OOC) platforms has gained increasing interest for its efficient and effective measurement of TEER in OOCs. To date, microfabricated electrodes or direct insertion of wires has been used to integrate TEER sensors into OOCs, with each method having advantages and disadvantages. In this study, we developed a TEER-SPE chip consisting of carbon-based screen-printed electrodes (SPEs) embedded in a poly(methyl methacrylate) (PMMA)-based multi-layered microfluidic device with a porous poly(ethylene terephthalate) membrane in-between. As proof of concept, we demonstrated the successful cultures of hCMEC/D3 cells and the formation of confluent monolayers in the TEER-SPE chip and obtained TEER measurements for 4 days. Additionally, the TEER-SPE chip could detect changes in the barrier integrity due to shear stress or an inflammatory cytokine (i.e., tumor necrosis factor-α). The novel approach enables a low-cost and facile fabrication of carbon-based SPEs on PMMA substrates and the subsequent assembly of PMMA layers for rapid prototyping. Being cost-effective and cleanroom-free, our method lowers the existing logistical and technical barriers presenting itself as another step forward to the broader adoption of OOCs with TEER measurement capability.
Collapse
Affiliation(s)
- Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Jamal Bahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Ning Yu
- Department of Chemical and Environmental Engineering, University of California-Riverside, Riverside, California, 92521, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Praveen Bandaru
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Lei Mou
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Enam Khalil
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Plastic, Aesthetic & Reconstructive Surgery and Laboratory of Experimental Cancer Research, Ghent University, 9000, Ghent, Belgium
| | - Rohollah Nasiri
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165, Solna, Sweden
| | - Annie Adachi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, 94158, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, 91125, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, 29208, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| |
Collapse
|
16
|
Velichkova G, Dobreva G. Human pluripotent stem cell-based models of heart development and disease. Cells Dev 2023; 175:203857. [PMID: 37257755 DOI: 10.1016/j.cdev.2023.203857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/16/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
The heart is a complex organ composed of distinct cell types, such as cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, neuronal cells and immune cells. All these cell types contribute to the structural, electrical and mechanical properties of the heart. Genetic manipulation and lineage tracing studies in mice have been instrumental in gaining critical insights into the networks regulating cardiac cell lineage specification, cell fate and plasticity. Such knowledge has been of fundamental importance for the development of efficient protocols for the directed differentiation of pluripotent stem cells (PSCs) in highly specialized cardiac cell types. In this review, we summarize the evolution and current advances in protocols for cardiac subtype specification, maturation, and assembly in cardiac microtissues and organoids.
Collapse
Affiliation(s)
- Gabriel Velichkova
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (DZHK), Germany.
| |
Collapse
|
17
|
House A, Cornick J, Butt Q, Guvendiren M. Elastomeric platform with surface wrinkling patterns to control cardiac cell alignment. J Biomed Mater Res A 2023; 111:1228-1242. [PMID: 36762538 DOI: 10.1002/jbm.a.37511] [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: 08/05/2022] [Revised: 12/07/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
There is a growing interest in creating 2D cardiac tissue models that display native extracellular matrix (ECM) cues of the heart tissue. Cellular alignment alone is known to be a crucial cue for cardiac tissue development by regulating cell-cell and cell-ECM interactions. In this study, we report a simple and robust approach to create lamellar surface wrinkling patterns enabling spatial control of pattern dimensions with a wide range of pattern amplitude (A ≈ 2-55 μm) and wavelength (λ ≈ 35-100 μm). For human cardiomyocytes (hCMs) and human cardiac fibroblasts (hCFs), our results indicate that the degree of cellular alignment and pattern recognition are correlated with pattern A and λ. We also demonstrate fabrication of devices composed of micro-well arrays with user-defined lamellar patterns on the bottom surface of each well for high-throughput screening studies. Results from a screening study indicate that cellular alignment is strongly diminished with increasing seeding density. In another study, we show our ability to vary hCM/hCF seeding ratio for each well to create co-culture systems where seeding ratio is independent of cellular alignment.
Collapse
Affiliation(s)
- Andrew House
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Jason Cornick
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Quratulain Butt
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
18
|
Boisnic S, Branchet MC, Quioc-Salomon B, Doan J, Delva C, Gendron C. Anti-Inflammatory and Antioxidant Effects of Diosmetin-3- O-β-d-Glucuronide, the Main Metabolite of Diosmin: Evidence from Ex Vivo Human Skin Models. Molecules 2023; 28:5591. [PMID: 37513462 PMCID: PMC10383842 DOI: 10.3390/molecules28145591] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Diosmin is used to relieve chronic venous disease (CVD) symptoms. This study aimed to investigate the anti-inflammatory and antioxidant effects of diosmetin-3-O-β-d-glucuronide, the major metabolite of diosmin, using human skin explants. The explants were exposed to substance P (inflammation model) or UVB irradiation (oxidative model) and to five diosmetin-3-O-β-d-glucuronide concentrations. Inflammation was evaluated through interleukin-8 (IL-8) secretion measurements and capillary dilation observation, and oxidation was evaluated by measuring the hydrogen peroxide levels and observing cyclobutane pyrimidine dimers (CPDs). In substance-P-exposed explants, diosmetin-3-O-β-d-glucuronide induced a significant decrease in IL-8 secretions, with a maximal effect at 2700 pg/mL (-49.6%), and it reduced the proportion of dilated capillaries and the mean luminal cross-sectional area (p < 0.0001 at all tested concentrations), indicating a vasoconstrictive effect. In UVB-irradiated fragments, diosmetin-3-O-β-d-glucuronide induced a significant decrease in hydrogen peroxide production and in the number of CPD-positive cells, reaching a maximal effect at the concentration of 2700 pg/mL (-48.6% and -52.0%, respectively). Diosmetin-3-O-β-d-glucuronide induced anti-inflammatory and antioxidant responses, with the maximal effect being reached at 2700 pg/mL and corresponding to the peak plasma concentration estimated after the oral intake of 600 mg of diosmin, the daily dose usually recommended for the treatment of CVD. These ex vivo findings suggest a protective role of diosmetin-3-O-β-d-glucuronide against inflammatory and oxidative stress affecting the vascular system in CVD pathophysiology.
Collapse
Affiliation(s)
- Sylvie Boisnic
- GREDECO (Group of Research and Evaluation in Dermatology and Cosmetology), 69 Rue de la Tour, 75016 Paris, France
| | - Marie-Christine Branchet
- GREDECO (Group of Research and Evaluation in Dermatology and Cosmetology), 69 Rue de la Tour, 75016 Paris, France
| | - Barbara Quioc-Salomon
- Laboratoire Innotech International, 22 Avenue Aristide Briand, 94110 Arcueil, France
| | - Julie Doan
- Laboratoire Innotech International, 22 Avenue Aristide Briand, 94110 Arcueil, France
| | | | - Célia Gendron
- Laboratoire Innotech International, 22 Avenue Aristide Briand, 94110 Arcueil, France
| |
Collapse
|
19
|
Scalia A, Doumani L, Kindt N, Journé F, Trelcat A, Carlier S. The Interplay between Atherosclerosis and Cancer: Breast Cancer Cells Increase the Expression of Endothelial Cell Adhesion Markers. BIOLOGY 2023; 12:896. [PMID: 37508329 PMCID: PMC10376633 DOI: 10.3390/biology12070896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Cardiovascular diseases are the leading causes of death worldwide, closely followed by cancer. To investigate the impact of breast cancer cell lines (SKBR3, MCF-7, and MDA-MB-231) on endothelial cell adhesion, a blended medium containing 30% breast-cancer-conditioned medium was prepared. This medium was then exposed to human umbilical vein endothelial cells (HUVECs) and monocytes (THP-1) for 48 h. Homemade oxidized low-density lipoproteins (oxLDL) were optionally added to the blended medium. Immunofluorescence was performed to assess the expression of E-selectin, connexin-43, and ICAM-1 on HUVECs, as well as LOX-1, CD36, and CD162 on THP-1. Additionally, unoxidized LDL was exposed to the three breast cancer cell lines for 48 h, and the formation of oxLDL was quantified. Our results revealed an upregulation of all six adhesion markers involved in the initiation of atherosclerosis when HUVECs and THP-1 were exposed to the breast-cancer-conditioned medium. Furthermore, this expression was further increased by exposure to oxLDL. We also observed a significant elevation in oxLDL levels when LDL was exposed to breast cancer cells. In conclusion, our findings successfully demonstrate an increased LDL oxidation in the presence of breast cancer cells, accompanied by an augmented expression of receptors involved in atherosclerosis initiation. These findings shed new light on the clinically observed interplay between atherosclerosis and cancer.
Collapse
Affiliation(s)
- Alessandro Scalia
- Department of Cardiology, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
| | - Lesly Doumani
- Department of Cardiology, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
| | - Nadège Kindt
- Department of Cardiology, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Department of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Fabrice Journé
- Department of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Anne Trelcat
- Department of Cardiology, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
| | - Stéphane Carlier
- Department of Cardiology, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
| |
Collapse
|
20
|
Stavrou M, Phung N, Grimm J, Andreou C. Organ-on-chip systems as a model for nanomedicine. NANOSCALE 2023; 15:9927-9940. [PMID: 37254663 PMCID: PMC10619891 DOI: 10.1039/d3nr01661g] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Nanomedicine is giving rise to increasing numbers of successful drugs, including cancer treatments, molecular imaging agents, and novel vaccine formulations. However, traditionally available model systems offer limited clinical translation and, compared to the number of preclinical studies, the approval rate of nanoparticles (NPs) for clinical use remains disappointingly low. A new paradigm of modeling biological systems on microfluidic chips has emerged in the last decade and is being gradually adopted by the nanomedicine community. These systems mimic tissues, organs, and diseases like cancer, on devices with small physical footprints and complex geometries. In this review, we report studies that used organ-on-chip approaches to study the interactions of NPs with biological systems. We present examples of NP toxicity studies, studies using biological NPs such as viruses, as well as modeling biological barriers and cancer on chip. Organ-on-chip systems present an exciting opportunity and can provide a renewed direction for the nanomedicine community.
Collapse
Affiliation(s)
- Marios Stavrou
- University of Cyprus, Department of Electrical and Computer Engineering, Nicosia, Cyprus.
| | - Ngan Phung
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA
- Weill Cornell Medical College, Department of Pharmacology, New York, NY, USA
| | - Jan Grimm
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA
- Weill Cornell Medical College, Department of Pharmacology, New York, NY, USA
| | - Chrysafis Andreou
- University of Cyprus, Department of Electrical and Computer Engineering, Nicosia, Cyprus.
| |
Collapse
|
21
|
Shevchuk O, Palii S, Pak A, Chantada N, Seoane N, Korda M, Campos-Toimil M, Álvarez E. Vessel-on-a-Chip: A Powerful Tool for Investigating Endothelial COVID-19 Fingerprints. Cells 2023; 12:cells12091297. [PMID: 37174696 PMCID: PMC10177552 DOI: 10.3390/cells12091297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Coronavirus disease (COVID-19) causes various vascular and blood-related reactions, including exacerbated responses. The role of endothelial cells in this acute response is remarkable and may remain important beyond the acute phase. As we move into a post-COVID-19 era (where most people have been or will be infected by the SARS-CoV-2 virus), it is crucial to define the vascular consequences of COVID-19, including the long-term effects on the cardiovascular system. Research is needed to determine whether chronic endothelial dysfunction following COVID-19 could lead to an increased risk of cardiovascular and thrombotic events. Endothelial dysfunction could also serve as a diagnostic and therapeutic target for post-COVID-19. This review covers these topics and examines the potential of emerging vessel-on-a-chip technology to address these needs. Vessel-on-a-chip would allow for the study of COVID-19 pathophysiology in endothelial cells, including the analysis of SARS-CoV-2 interactions with endothelial function, leukocyte recruitment, and platelet activation. "Personalization" could be implemented in the models through induced pluripotent stem cells, patient-specific characteristics, or genetic modified cells. Adaptation for massive testing under standardized protocols is now possible, so the chips could be incorporated for the personalized follow-up of the disease or its sequalae (long COVID) and for the research of new drugs against COVID-19.
Collapse
Affiliation(s)
- Oksana Shevchuk
- Department of Pharmacology and Clinical Pharmacology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Svitlana Palii
- Department of Pharmacology and Clinical Pharmacology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Anastasiia Pak
- Department of Medical Biochemistry, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Nuria Chantada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Nuria Seoane
- Physiology and Pharmacology of Chronic Diseases (FIFAEC) Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mykhaylo Korda
- Department of Medical Biochemistry, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Manuel Campos-Toimil
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Physiology and Pharmacology of Chronic Diseases (FIFAEC) Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ezequiel Álvarez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, 28220 Madrid, Spain
| |
Collapse
|
22
|
Nazari H, Shrestha J, Naei VY, Bazaz SR, Sabbagh M, Thiery JP, Warkiani ME. Advances in TEER measurements of biological barriers in microphysiological systems. Biosens Bioelectron 2023; 234:115355. [PMID: 37159988 DOI: 10.1016/j.bios.2023.115355] [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: 08/11/2022] [Revised: 03/10/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023]
Abstract
Biological barriers are multicellular structures that precisely regulate the transport of ions, biomolecules, drugs, cells, and other organisms. Transendothelial/epithelial electrical resistance (TEER) is a label-free method for predicting the properties of biological barriers. Understanding the mechanisms that control TEER significantly enhances our knowledge of the physiopathology of different diseases and aids in the development of new drugs. Measuring TEER values within microphysiological systems called organ-on-a-chip devices that simulate the microenvironment, architecture, and physiology of biological barriers in the body provides valuable insight into the behavior of barriers in response to different drugs and pathogens. These integrated systems should increase the accuracy, reproducibility, sensitivity, resolution, high throughput, speed, cost-effectiveness, and reliable predictability of TEER measurements. Implementing advanced micro and nanoscale manufacturing techniques, surface modification methods, biomaterials, biosensors, electronics, and stem cell biology is necessary for integrating TEER measuring systems with organ-on-chip technology. This review focuses on the applications, advantages, and future perspectives of integrating organ-on-a-chip technology with TEER measurement methods for studying biological barriers. After briefly reviewing the role of TEER in the physiology and pathology of barriers, standard techniques for measuring TEER, including Ohm's law and impedance spectroscopy, and commercially available devices are described. Furthermore, advances in TEER measurement are discussed in multiple barrier-on-a-chip system models representing different organs. Finally, we outline future trends in implementing advanced technologies to design and fabricate nanostructured electrodes, complicated microfluidic chips, and membranes for more advanced and accurate TEER measurements.
Collapse
Affiliation(s)
- Hojjatollah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Vahid Yaghoubi Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Milad Sabbagh
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | | | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia; Institute of Molecular Medicine, Sechenov University, 119991, Moscow, Russia.
| |
Collapse
|
23
|
Ferrari D, Sengupta A, Heo L, Pethö L, Michler J, Geiser T, de Jesus Perez VA, Kuebler WM, Zeinali S, Guenat OT. Effects of biomechanical and biochemical stimuli on angio- and vasculogenesis in a complex microvasculature-on-chip. iScience 2023; 26:106198. [PMID: 36879808 PMCID: PMC9985038 DOI: 10.1016/j.isci.2023.106198] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/31/2022] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The endothelium of blood vessels is a vital organ that reacts differently to subtle changes in stiffness and mechanical forces exerted on its environment (extracellular matrix (ECM)). Upon alteration of these biomechanical cues, endothelial cells initiate signaling pathways that govern vascular remodeling. The emerging organs-on-chip technologies allow the mimicking of complex microvasculature networks, identifying the combined or singular effects of these biomechanical or biochemical stimuli. Here, we present a microvasculature-on-chip model to investigate the singular effect of ECM stiffness and mechanical cyclic stretch on vascular development. Following two different approaches for vascular growth, the effect of ECM stiffness on sprouting angiogenesis and the effect of cyclic stretch on endothelial vasculogenesis are studied. Our results indicate that ECM hydrogel stiffness controls the size of the patterned vasculature and the density of sprouting angiogenesis. RNA sequencing shows that the cellular response to stretching is characterized by the upregulation of certain genes such as ANGPTL4+5, PDE1A, and PLEC.
Collapse
Affiliation(s)
- Dario Ferrari
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Lyong Heo
- Stanford Center for Genomics and Personalized Medicine, Palo Alto, CA, USA
| | - Laszlo Pethö
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Johann Michler
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Almeida GHDR, Iglesia RP, Rinaldi JDC, Murai MK, Calomeno CVAQ, da Silva Junior LN, Horvath-Pereira BDO, Pinho LBM, Miglino MA, Carreira ACO. Current Trends on Bioengineering Approaches for Ovarian Microenvironment Reconstruction. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36355603 DOI: 10.1089/ten.teb.2022.0171] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian tissue has a unique microarchitecture and a complex cellular and molecular dynamics that are essential for follicular survival and development. Due to this great complexity, several factors may lead to ovarian insufficiency, and therefore to systemic metabolic disorders and female infertility. Techniques currently used in the reproductive clinic such as oocyte cryopreservation or even ovarian tissue transplant, although effective, have several limitations, which impair their wide application. In this scenario, mimetic ovarian tissue reconstruction comes as an innovative alternative to develop new methodologies for germ cells preservation and ovarian functions restoration. The ovarian extracellular matrix (ECM) is crucial for oocyte viability maintenance, once it acts actively in folliculogenesis. One of the key components of ovarian bioengineering is biomaterials application that mimics ECM and provides conditions for cell anchorage, proliferation, and differentiation. Therefore, this review aims at describing ovarian tissue engineering approaches and listing the main limitations of current methods for preservation and reestablishment of ovarian fertility. In addition, we describe the main elements that structure this study field, highlighting the main advances and the challenges to overcome to develop innovative methodologies to be applied in reproductive medicine. Impact Statement This review presents the main advances in the application of tissue bioengineering in the ovarian tissue reconstruction to develop innovative solutions for ovarian fertility reestablishment.
Collapse
Affiliation(s)
| | - Rebeca Piatniczka Iglesia
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mikaelly Kiemy Murai
- Department of Morphological Sciences, State University of Maringa, Maringá, Brazil
| | | | | | | | - Letícia Beatriz Mazo Pinho
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Center of Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
| |
Collapse
|
25
|
Wu L, Ai Y, Xie R, Xiong J, Wang Y, Liang Q. Organoids/organs-on-a-chip: new frontiers of intestinal pathophysiological models. LAB ON A CHIP 2023; 23:1192-1212. [PMID: 36644984 DOI: 10.1039/d2lc00804a] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Organoids/organs-on-a-chip open up new frontiers for basic and clinical research of intestinal diseases. Species-specific differences hinder research on animal models, while organoids are emerging as powerful tools due to self-organization from stem cells and the reproduction of the functional properties in vivo. Organs-on-a-chip is also accelerating the process of faithfully mimicking the intestinal microenvironment. And by combining organoids and organ-on-a-chip technologies, they further are expected to serve as innovative preclinical tools and could outperform traditional cell culture models or animal models in the future. Above all, organoids/organs-on-a-chip with other strategies like genome editing, 3D printing, and organoid biobanks contribute to modeling intestinal homeostasis and disease. Here, the current challenges and future trends in intestinal pathophysiological models will be summarized.
Collapse
Affiliation(s)
- 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, 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, Beijing 100084, P.R. China.
| | - Ruoxiao Xie
- 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, Beijing 100084, P.R. China.
| | - Jialiang Xiong
- 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, Beijing 100084, P.R. China.
| | - Yu 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, Beijing 100084, P.R. China.
| | - 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, Beijing 100084, P.R. China.
| |
Collapse
|
26
|
Sisodia Y, Shah K, Ali Sayyed A, Jain M, Ali SA, Gondaliya P, Kalia K, Tekade RK. Lung-on-chip microdevices to foster pulmonary drug discovery. Biomater Sci 2023; 11:777-790. [PMID: 36537540 DOI: 10.1039/d2bm00951j] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Respiratory diseases account for unprecedented mortality owing to a lack of personalized or insufficient therapeutic interventions. Fostering pulmonary research into managing pulmonary threat requires a potential alternative approach that can mimick the in vivo complexities of the human body. The in vitro miniaturized bionic simulation of the lung holds great potential in the quest for a successful therapeutic intervention. This review discusses the emerging roles of lung-on-chip microfluidic simulator devices in fostering translational pulmonary drug discovery and personalized medicine. This review also explicates how the lung-on-chip model emulates the breathing patterns, elasticity, and vascularization of lungs in creating a 3D pulmonary microenvironment.
Collapse
Affiliation(s)
- Yashi Sisodia
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Komal Shah
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Adil Ali Sayyed
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Transplantation, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Meenakshi Jain
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Syed Ansar Ali
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Piyush Gondaliya
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Transplantation, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Kiran Kalia
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Rakesh Kumar Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.
| |
Collapse
|
27
|
de Graaf MNS, Vivas A, Kasi DG, van den Hil FE, van den Berg A, van der Meer AD, Mummery CL, Orlova VV. Multiplexed fluidic circuit board for controlled perfusion of 3D blood vessels-on-a-chip. LAB ON A CHIP 2022; 23:168-181. [PMID: 36484766 PMCID: PMC9764810 DOI: 10.1039/d2lc00686c] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/21/2022] [Indexed: 06/11/2023]
Abstract
Three-dimensional (3D) blood vessels-on-a-chip (VoC) models integrate the biological complexity of vessel walls with dynamic microenvironmental cues, such as wall shear stress (WSS) and circumferential strain (CS). However, these parameters are difficult to control and are often poorly reproducible due to the high intrinsic diameter variation of individual 3D-VoCs. As a result, the throughput of current 3D systems is one-channel-at-a-time. Here, we developed a fluidic circuit board (FCB) for simultaneous perfusion of up to twelve 3D-VoCs using a single set of control parameters. By designing the internal hydraulic resistances in the FCB appropriately, it was possible to provide a pre-set WSS to all connected 3D-VoCs, despite significant variation in lumen diameters. Using this FCB, we found that variation of CS or WSS induce morphological changes to human induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) and conclude that control of these parameters using a FCB is necessary to study 3D-VOCs.
Collapse
Affiliation(s)
- Mees N S de Graaf
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Aisen Vivas
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | - Dhanesh G Kasi
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
28
|
van der Velden J, Asselbergs FW, Bakkers J, Batkai S, Bertrand L, Bezzina CR, Bot I, Brundel BJJM, Carrier L, Chamuleau S, Ciccarelli M, Dawson D, Davidson SM, Dendorfer A, Duncker DJ, Eschenhagen T, Fabritz L, Falcão-Pires I, Ferdinandy P, Giacca M, Girao H, Gollmann-Tepeköylü C, Gyongyosi M, Guzik TJ, Hamdani N, Heymans S, Hilfiker A, Hilfiker-Kleiner D, Hoekstra AG, Hulot JS, Kuster DWD, van Laake LW, Lecour S, Leiner T, Linke WA, Lumens J, Lutgens E, Madonna R, Maegdefessel L, Mayr M, van der Meer P, Passier R, Perbellini F, Perrino C, Pesce M, Priori S, Remme CA, Rosenhahn B, Schotten U, Schulz R, Sipido KR, Sluijter JPG, van Steenbeek F, Steffens S, Terracciano CM, Tocchetti CG, Vlasman P, Yeung KK, Zacchigna S, Zwaagman D, Thum T. Animal models and animal-free innovations for cardiovascular research: current status and routes to be explored. Consensus document of the ESC Working Group on Myocardial Function and the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2022; 118:3016-3051. [PMID: 34999816 PMCID: PMC9732557 DOI: 10.1093/cvr/cvab370] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases represent a major cause of morbidity and mortality, necessitating research to improve diagnostics, and to discover and test novel preventive and curative therapies, all of which warrant experimental models that recapitulate human disease. The translation of basic science results to clinical practice is a challenging task, in particular for complex conditions such as cardiovascular diseases, which often result from multiple risk factors and comorbidities. This difficulty might lead some individuals to question the value of animal research, citing the translational 'valley of death', which largely reflects the fact that studies in rodents are difficult to translate to humans. This is also influenced by the fact that new, human-derived in vitro models can recapitulate aspects of disease processes. However, it would be a mistake to think that animal models do not represent a vital step in the translational pathway as they do provide important pathophysiological insights into disease mechanisms particularly on an organ and systemic level. While stem cell-derived human models have the potential to become key in testing toxicity and effectiveness of new drugs, we need to be realistic, and carefully validate all new human-like disease models. In this position paper, we highlight recent advances in trying to reduce the number of animals for cardiovascular research ranging from stem cell-derived models to in situ modelling of heart properties, bioinformatic models based on large datasets, and state-of-the-art animal models, which show clinically relevant characteristics observed in patients with a cardiovascular disease. We aim to provide a guide to help researchers in their experimental design to translate bench findings to clinical routine taking the replacement, reduction, and refinement (3R) as a guiding concept.
Collapse
Grants
- R01 HL150359 NHLBI NIH HHS
- RG/16/14/32397 British Heart Foundation
- FS/18/37/33642 British Heart Foundation
- PG/17/64/33205 British Heart Foundation
- PG/15/88/31780 British Heart Foundation
- FS/RTF/20/30009, NH/19/1/34595, PG/18/35/33786, CS/17/4/32960, PG/15/88/31780, and PG/17/64/33205 British Heart Foundation
- NC/T001488/1 National Centre for the Replacement, Refinement and Reduction of Animals in Research
- PG/18/44/33790 British Heart Foundation
- CH/16/3/32406 British Heart Foundation
- FS/RTF/20/30009 British Heart Foundation
- NWO-ZonMW
- ZonMW and Heart Foundation for the translational research program
- Dutch Cardiovascular Alliance (DCVA)
- Leducq Foundation
- Dutch Research Council
- Association of Collaborating Health Foundations (SGF)
- UCL Hospitals NIHR Biomedical Research Centre, and the DCVA
- Netherlands CardioVascular Research Initiative CVON
- Stichting Hartekind and the Dutch Research Counsel (NWO) (OCENW.GROOT.2019.029)
- National Fund for Scientific Research, Belgium and Action de Recherche Concertée de la Communauté Wallonie-Bruxelles, Belgium
- Netherlands CardioVascular Research Initiative CVON (PREDICT2 and CONCOR-genes projects), the Leducq Foundation
- ERA PerMed (PROCEED study)
- Netherlands Cardiovascular Research Initiative
- Dutch Heart Foundation
- German Centre of Cardiovascular Research (DZHH)
- Chest Heart and Stroke Scotland
- Tenovus Scotland
- Friends of Anchor and Grampian NHS-Endowments
- National Institute for Health Research University College London Hospitals Biomedical Research Centre
- German Centre for Cardiovascular Research
- European Research Council (ERC-AG IndivuHeart), the Deutsche Forschungsgemeinschaft
- European Union Horizon 2020 (REANIMA and TRAINHEART)
- German Ministry of Education and Research (BMBF)
- Centre for Cardiovascular Research (DZHK)
- European Union Horizon 2020
- DFG
- National Research, Development and Innovation Office of Hungary
- Research Excellence Program—TKP; National Heart Program
- Austrian Science Fund
- European Union Commission’s Seventh Framework programme
- CVON2016-Early HFPEF
- CVON She-PREDICTS
- CVON Arena-PRIME
- European Union’s Horizon 2020 research and innovation programme
- Deutsche Forschungsgemeinschaft
- Volkswagenstiftung
- French National Research Agency
- ERA-Net-CVD
- Fédération Française de Cardiologie, the Fondation pour la Recherche Médicale
- French PIA Project
- University Research Federation against heart failure
- Netherlands Heart Foundation
- Dekker Senior Clinical Scientist
- Health Holland TKI-LSH
- TUe/UMCU/UU Alliance Fund
- south African National Foundation
- Cancer Association of South Africa and Winetech
- Netherlands Heart Foundation/Applied & Engineering Sciences
- Dutch Technology Foundation
- Pie Medical Imaging
- Netherlands Organisation for Scientific Research
- Dr. Dekker Program
- Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation
- Dutch Federation of University Medical Centres
- Netherlands Organization for Health Research and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project
- Netherlands Organization for Scientific Research (NWO) (VICI grant); the European Research Council
- Incyte s.r.l. and from Ministero dell’Istruzione, Università e Ricerca Scientifica
- German Center for Cardiovascular Research (Junior Research Group & Translational Research Project), the European Research Council (ERC Starting Grant NORVAS),
- Swedish Heart-Lung-Foundation
- Swedish Research Council
- National Institutes of Health
- Bavarian State Ministry of Health and Care through the research project DigiMed Bayern
- ERC
- ERA-CVD
- Dutch Heart Foundation, ZonMw
- the NWO Gravitation project
- Ministero dell'Istruzione, Università e Ricerca Scientifica
- Regione Lombardia
- Netherlands Organisation for Health Research and Development
- ITN Network Personalize AF: Personalized Therapies for Atrial Fibrillation: a translational network
- MAESTRIA: Machine Learning Artificial Intelligence Early Detection Stroke Atrial Fibrillation
- REPAIR: Restoring cardiac mechanical function by polymeric artificial muscular tissue
- Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)
- European Union H2020 program to the project TECHNOBEAT
- EVICARE
- BRAV3
- ZonMw
- German Centre for Cardiovascular Research (DZHK)
- British Heart Foundation Centre for Cardiac Regeneration
- British Heart Foundation studentship
- NC3Rs
- Interreg ITA-AUS project InCARDIO
- Italian Association for Cancer Research
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, UK
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sandor Batkai
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Luc Bertrand
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Connie R Bezzina
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Ilze Bot
- Heart Center, Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Steven Chamuleau
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Odontology, University of Salerno, Fisciano (SA), Italy
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- University Center of Cardiovascular Sciences and Department of Cardiology, University Heart Center Hamburg, Germany and Institute of Cardiovascular Sciences, University of Birmingham, UK
| | - Ines Falcão-Pires
- UnIC - Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Portugal
| | - Péter Ferdinandy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Henrique Girao
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, Faculty of Medicine, Coimbra, Portugal
- Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | | | - Mariann Gyongyosi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Tomasz J Guzik
- Instutute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Nazha Hamdani
- Division Cardiology, Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Andres Hilfiker
- Department for Cardiothoracic, Transplant, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Comprehensive Cancer Centre, Philipps-Universität Marburg, Germany
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Sébastien Hulot
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Linda W van Laake
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Tim Leiner
- Department of Radiology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27B, 48149 Muenster, Germany
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, 56124 Pisa, Italy
- Department of Internal Medicine, Cardiology Division, University of Texas Medical School in Houston, Houston, TX, USA
| | - Lars Maegdefessel
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500AE Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Filippo Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro cardiologico Monzino, IRCCS, Milan, Italy
| | - Silvia Priori
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, Pavia, Italy
- University of Pavia, Pavia, Italy
| | - Carol Ann Remme
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Bodo Rosenhahn
- Institute for information Processing, Leibniz University of Hanover, 30167 Hannover, Germany
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karin R Sipido
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, Regenerative Medicine Center Utrecht, Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Steenbeek
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center for Clinical and Translational Research (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Patricia Vlasman
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Amsterdam UMC, Vrije Universiteit, Surgery, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Dayenne Zwaagman
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Thomas Thum
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
29
|
Dash BC, Korutla L, Vallabhajosyula P, Hsia HC. Unlocking the Potential of Induced Pluripotent Stem Cells for Wound Healing: The Next Frontier of Regenerative Medicine. Adv Wound Care (New Rochelle) 2022; 11:622-638. [PMID: 34155919 DOI: 10.1089/wound.2021.0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: Nonhealing wounds are a significant burden for the health care system all over the world. Existing treatment options are not enough to promote healing, highlighting the urgent need for improved therapies. In addition, the current advancements in tissue-engineered skin constructs and stem cell-based therapies are facing significant hurdles due to the absence of a renewable source of functional cells. Recent Advances: Induced pluripotent stem cell technology (iPSC) is emerging as a novel tool to develop the next generation of personalized medicine for the treatment of chronic wounds. The iPSC provides unlimited access to various skin cells to generate complex personalized three-dimensional skin constructs for disease modeling and autologous grafts. Furthermore, the iPSC-based therapies can target distinct wound healing phases and have shown accelerating wound closure by enhancing angiogenesis, cell migration, tissue regeneration, and modulating inflammation. Critical Issues: Since the last decade, iPSC has been revolutionizing the field of wound healing and skin tissue engineering. Despite the current progress, safety and heterogeneity among iPSC lines are still major hurdles in addition to the lack of large animal studies. These challenges need to be addressed before translating an iPSC-based therapy to the clinic. Future Directions: Future considerations should be given to performing large animal studies to check the safety and efficiency of iPSC-based therapy in a wound healing setup. Furthermore, strategies should be developed to overcome variation between hiPSC lines, develop an efficient manufacturing process for iPSC-derived products, and generate complex skin constructs with vasculature and skin appendages.
Collapse
Affiliation(s)
- Biraja C Dash
- Department of Surgery (Plastic), Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Laxminarayana Korutla
- Department of Surgery (Cardiac), Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Prashanth Vallabhajosyula
- Department of Surgery (Cardiac), Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Henry C Hsia
- Department of Surgery (Plastic), Yale School of Medicine, Yale University, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
30
|
McCloskey MC, Zhang VZ, Ahmad SD, Walker S, Romanick SS, Awad HA, McGrath JL. Sourcing cells for in vitro models of human vascular barriers of inflammation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:979768. [PMID: 36483299 PMCID: PMC9724237 DOI: 10.3389/fmedt.2022.979768] [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: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 07/20/2023] Open
Abstract
The vascular system plays a critical role in the progression and resolution of inflammation. The contributions of the vascular endothelium to these processes, however, vary with tissue and disease state. Recently, tissue chip models have emerged as promising tools to understand human disease and for the development of personalized medicine approaches. Inclusion of a vascular component within these platforms is critical for properly evaluating most diseases, but many models to date use "generic" endothelial cells, which can preclude the identification of biomedically meaningful pathways and mechanisms. As the knowledge of vascular heterogeneity and immune cell trafficking throughout the body advances, tissue chip models should also advance to incorporate tissue-specific cells where possible. Here, we discuss the known heterogeneity of leukocyte trafficking in vascular beds of some commonly modeled tissues. We comment on the availability of different tissue-specific cell sources for endothelial cells and pericytes, with a focus on stem cell sources for the full realization of personalized medicine. We discuss sources available for the immune cells needed to model inflammatory processes and the findings of tissue chip models that have used the cells to studying transmigration.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samantha S. Romanick
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
31
|
Bulut M, Vila Cuenca M, de Graaf M, van den Hil FE, Mummery CL, Orlova VV. Three-Dimensional Vessels-on-a-Chip Based on hiPSC-derived Vascular Endothelial and Smooth Muscle Cells. Curr Protoc 2022; 2:e564. [PMID: 36250774 DOI: 10.1002/cpz1.564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Blood vessels are composed of endothelial cells (ECs) that form the inner vessel wall and mural cells that cover the ECs to mediate their stabilization. Crosstalk between ECs and VSMCs while the ECs undergo microfluidic flow is vital for the function and integrity of blood vessels. Here, we describe a protocol to generate three-dimensional (3D) engineered vessels-on-chip (VoCs) composed of vascular cells derived from human induced pluripotent stem cells (hiPSCs). We first describe protocols for robust differentiation of vascular smooth muscle cells (hiPSC-VSMCs) from hiPSCs that are effective across multiple hiPSC lines. Second, we describe the fabrication of a simple microfluidic device consisting of a single collagen lumen that can act as a cell scaffold and support fluid flow using the viscous finger patterning (VFP) technique. After the channel is seeded sequentially with hiPSC-derived ECs (hiPSC-ECs) and hiPSC-VSMCs, a stable EC barrier covered by VSMCs lines the collagen lumen. We demonstrate that this 3D VoC model can recapitulate physiological cell-cell interaction and can be perfused under physiological shear stress using a microfluidic pump. The uniform geometry of the vessel lumens allows precise control of flow dynamics. We have thus developed a robust protocol to generate an entirely isogenic hiPSC-derived 3D VoC model, which could be valuable for studying vessel barrier function and physiology in healthy or disease states. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Differentiation of hiPSC-VSMCs Support Protocol 1: Characterization of hiPSC-NCCs and hiPSC-VSMCs Support Protocol 2: Preparation of cryopreserved hiPSC-VSMCs and hiPSC-ECs for VoC culture Basic Protocol 2: Generation of 3D VoC model composed of hiPSC-ECs and hiPSC-VSMCs Support Protocol 3: Structural characterization of 3D VoC model.
Collapse
Affiliation(s)
- Merve Bulut
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marc Vila Cuenca
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mees de Graaf
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Mathur T, Kumar A, Flanagan JM, Jain A. Vascular Transcriptomics: Investigating Endothelial Activation and Vascular Dysfunction Using Blood Outgrowth Endothelial Cells, Organ-Chips, and RNA Sequencing. Curr Protoc 2022; 2:e582. [PMID: 36300922 PMCID: PMC9627633 DOI: 10.1002/cpz1.582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vascular organ-chip or vessel-chip technology has significantly impacted our ability to model microphysiological vasculature. These biomimetic platforms have garnered significant interest from scientists and pharmaceutical companies as drug screening models. However, these models still lack the inclusion of patient-specific vasculature in the form of patient-derived endothelial cells. Blood outgrowth endothelial cells are patient blood-derived endothelial progenitors that have gained interest from the vascular biology community as an autologous endothelial cell alternative and have also been incorporated with the vessel-chip model. Next-generation sequencing techniques like RNA sequencing can further unlock the potential of personalized vessel-chips in discerning patient-specific hallmarks of endothelial dysfunction. Here we present a detailed protocol for (1) isolating blood outgrowth endothelial cells from patient blood samples, (2) culturing them in microfluidic vessel-chips, (3) isolating and preparing RNA from individual vessel-chips for sequencing, and (4) performing differential gene expression and bioinformatics analyses of vascular dysfunction and endothelial activation pathways. This method focuses specifically on identification of pathways and genes involved in vascular homeostasis and pathology, but can easily be adapted for the requirements of other systems. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Isolation of blood outgrowth endothelial cells from patient blood Basic Protocol 2: Culture of blood outgrowth endothelial cells in microfluidic vessel-chips Basic Protocol 3: Isolation of RNA from autologous vessel-chips Basic Protocol 4: Differential gene expression and bioinformatics analyses of endothelial activation pathways.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
| | - Ankit Kumar
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
| | - Jonathan M. Flanagan
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, USA
| |
Collapse
|
33
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|
34
|
Hetzer MW, Bersini S. Beyond Static Pipes: Mechanisms and In Vitro Models of Vascular Aging. Cold Spring Harb Perspect Med 2022; 12:a041180. [PMID: 35101902 PMCID: PMC9310951 DOI: 10.1101/cshperspect.a041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The vascular system is a key player for the maintenance of healthy tissues, suggesting how the physiological decline of blood vessel functionality during aging could be a major contributor of organ degeneration. While basic research studies have begun to pinpoint potential mechanisms of vascular aging, it is now critical to translate them into therapeutically relevant options. Microphysiological systems represent a powerful tool to precisely control which combinations of stimuli are provided to in vitro reconstructed blood vessels and to analyze their functional consequences. After highlighting key aspects of vascular aging, this review discusses in vitro models that are able to recapitulate relevant features of blood vessel damage during aging. Strategies to improve current in vitro systems so that they will more faithfully recapitulate vascular aging are proposed, emphasizing the importance of combining in vivo models with microphysiological systems for an effective translation of vascular aging biomarkers and therapies to the clinical level.
Collapse
Affiliation(s)
- Martin W Hetzer
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Simone Bersini
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
35
|
Vivas A, van den Berg A, Passier R, Odijk M, van der Meer AD. Fluidic circuit board with modular sensor and valves enables stand-alone, tubeless microfluidic flow control in organs-on-chips. LAB ON A CHIP 2022; 22:1231-1243. [PMID: 35178541 PMCID: PMC8922413 DOI: 10.1039/d1lc00999k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/09/2022] [Indexed: 05/13/2023]
Abstract
Organs-on-chips are a unique class of microfluidic in vitro cell culture models, in which the in vivo tissue microenvironment is mimicked. Unfortunately, their widespread use is hampered by their operation complexity and incompatibility with end-user research settings. To address these issues, many commercial and non-commercial platforms have been developed for semi-automated culture of organs-on-chips. However, these organ-on-chip culture platforms each represent a closed ecosystem, with very little opportunity to interchange and integrate components from different platforms or to develop new ones. The translational organ-on-chip platform (TOP) is a multi-institutional effort to develop an open platform for automated organ-on-chip culture and integration of components from various developers. Central to TOP is the fluidic circuit board (FCB), a microfluidic plate with the form factor of a typical well plate. The FCB enables microfluidic control of multiple components like sensors or organ-on-chip devices through an interface based on openly available standards. Here, we report an FCB to integrate commercial and in-house developed components forming a stand-alone flow control system for organs-on-chips. The control system is able to achieve constant and pulsatile flow recirculation through a connected organ-on-chip device. We demonstrate that this system is able to automatically perfuse a heart-on-chip device containing co-cultures of cardiac tissues derived from human pluripotent stem cell-derived cardiomyocytes and monolayers of endothelial cells for five days. Altogether, we conclude that open technology platforms allow the integration of components from different sources to form functional and fit-for-purpose organ-on-chip systems. We anticipate that open platforms will play a central role in catalyzing and maturing further technological development of organ-on-chip culture systems.
Collapse
Affiliation(s)
- Aisen Vivas
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Robert Passier
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Mathieu Odijk
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Andries D van der Meer
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
| |
Collapse
|
36
|
Xu Y, Deng J, Hao S, Wang B. A Potential In Vitro 3D Cell Model to Study Vascular Diseases by Simulating the Vascular Wall Microenvironment and Its Application. Life (Basel) 2022; 12:427. [PMID: 35330178 PMCID: PMC8951029 DOI: 10.3390/life12030427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Current in vitro vascular models are too simple compared with the real vascular environment. In this research, a novel in vitro 3D vascular disease model that simulated the vascular microenvironment was introduced. METHODS This model was mainly established by low shear stress and co-culture of endothelial cells and smooth muscle cells. Characterization and reproduction of the pathological state of the 3D model were determined. The effect of two clinical drugs was verified in this model. The difference of drug screening between a traditional oxidative-damaged cell model and this 3D model was determined by HPLC. RESULTS This model presented many disease markers of vascular diseases: abnormal cellular shape, higher endothelial cell apoptotic rate and smooth muscle cell migration rate, decreased superoxide dismutase level, and increased malondialdehyde and platelet-derived growth factor level. The drugs effectively reduced the disease indices and relieved the damage caused by low shear stress. Compared to the traditional oxidative-damaged cell model, this 3D model screened different active components of Salviae Miltiorrhizae extract, and it is closer to clinical studies. CONCLUSIONS These results suggest that the 3D vascular disease model is a more efficient and selective in vitro study and drug screening platform for vascular diseases than previously reported in vitro vascular disease models.
Collapse
Affiliation(s)
- Yingqian Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Y.X.); (S.H.)
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Jia Deng
- Chongqing Key Laboratory of Natural Medicine Research, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China;
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Y.X.); (S.H.)
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Y.X.); (S.H.)
| |
Collapse
|
37
|
Recent Development of Drug Delivery Systems through Microfluidics: From Synthesis to Evaluation. Pharmaceutics 2022; 14:pharmaceutics14020434. [PMID: 35214166 PMCID: PMC8880124 DOI: 10.3390/pharmaceutics14020434] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 01/04/2023] Open
Abstract
Conventional drug administration usually faces the problems of degradation and rapid excretion when crossing many biological barriers, leading to only a small amount of drugs arriving at pathological sites. Therapeutic drugs delivered by drug delivery systems to the target sites in a controlled manner greatly enhance drug efficacy, bioavailability, and pharmacokinetics with minimal side effects. Due to the distinct advantages of microfluidic techniques, microfluidic setups provide a powerful tool for controlled synthesis of drug delivery systems, precisely controlled drug release, and real-time observation of drug delivery to the desired location at the desired rate. In this review, we present an overview of recent advances in the preparation of nano drug delivery systems and carrier-free drug delivery microfluidic systems, as well as the construction of in vitro models on-a-chip for drug efficiency evaluation of drug delivery systems. We firstly introduce the synthesis of nano drug delivery systems, including liposomes, polymers, and inorganic compounds, followed by detailed descriptions of the carrier-free drug delivery system, including micro-reservoir and microneedle drug delivery systems. Finally, we discuss in vitro models developed on microfluidic devices for the evaluation of drug delivery systems, such as the blood–brain barrier model, vascular model, small intestine model, and so on. The opportunities and challenges of the applications of microfluidic platforms in drug delivery systems, as well as their clinical applications, are also discussed.
Collapse
|
38
|
Chen J, Zhang X, Millican R, Lynd T, Gangasani M, Malhotra S, Sherwood J, Hwang PT, Cho Y, Brott BC, Qin G, Jo H, Yoon YS, Jun HW. Recent Progress in in vitro Models for Atherosclerosis Studies. Front Cardiovasc Med 2022; 8:790529. [PMID: 35155603 PMCID: PMC8829969 DOI: 10.3389/fcvm.2021.790529] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is the primary cause of hardening and narrowing arteries, leading to cardiovascular disease accounting for the high mortality in the United States. For developing effective treatments for atherosclerosis, considerable efforts have been devoted to developing in vitro models. Compared to animal models, in vitro models can provide great opportunities to obtain data more efficiently, economically. Therefore, this review discusses the recent progress in in vitro models for atherosclerosis studies, including traditional two-dimensional (2D) systems cultured on the tissue culture plate, 2D cell sheets, and recently emerged microfluidic chip models with 2D culture. In addition, advanced in vitro three-dimensional models such as spheroids, cell-laden hydrogel constructs, tissue-engineered blood vessels, and vessel-on-a-chip will also be covered. Moreover, the functions of these models are also summarized along with model discussion. Lastly, the future perspectives of this field are discussed.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xixi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Tyler Lynd
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Manas Gangasani
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shubh Malhotra
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | - Younghye Cho
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
- Family Medicine Clinic, Obesity, Metabolism, and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Brigitta C. Brott
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
- Endomimetics, LLC., Birmingham, AL, United States
- Division of Cardiovascular Disease, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gangjian Qin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Young-sup Yoon
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
- Endomimetics, LLC., Birmingham, AL, United States
- *Correspondence: Ho-Wook Jun
| |
Collapse
|
39
|
Abstract
The development of vasculature in vivo is an extremely complex process that requires temporal and spatial coordination between multiple cell types to produce an effective vessel. The formation of vasculature from preexisting blood vessels, known as angiogenesis, plays important roles in several physiological and pathological processes, including wound healing, organ development and growth, ischemia, inflammatory disorders, fibrosis, and cancer. Means to deconstruct these complicated biological systems are necessary to gain mechanistic insight into their development, function, and modulation that can be tested in in vivo models and ultimately the clinic. In this chapter, we will first review the classical in vitro techniques to study angiogenesis. Next, we will explore the exciting recent advances that rely on 3D multicellular systems to more accurately mimic vasculature development in vitro. Finally, we will discuss the applications of in vitro angiogenic methods to study related vasculature phenomena, such as vasculogenic mimicry.
Collapse
Affiliation(s)
- Ralph Francescone
- Cancer Signaling and Epigenetics Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Marvin and Concetta Greenberg, Pancreatic Cancer Institute, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Débora Barbosa Vendramini-Costa
- Cancer Signaling and Epigenetics Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
- Marvin and Concetta Greenberg, Pancreatic Cancer Institute, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
40
|
Moreira A, Müller M, Costa PF, Kohl Y. Advanced In Vitro Lung Models for Drug and Toxicity Screening: The Promising Role of Induced Pluripotent Stem Cells. Adv Biol (Weinh) 2021; 6:e2101139. [PMID: 34962104 DOI: 10.1002/adbi.202101139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/25/2021] [Indexed: 12/24/2022]
Abstract
The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.
Collapse
Affiliation(s)
| | - Michelle Müller
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, Porto, 4200-135, Portugal
| | - Yvonne Kohl
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Postgraduate Course for Toxicology and Environmental Toxicology, Medical Faculty, University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
41
|
Sorrentino S, Polini A, Arima V, Romano A, Quattrini A, Gigli G, Mozetic P, Moroni L. Neurovascular signals in amyotrophic lateral sclerosis. Curr Opin Biotechnol 2021; 74:75-83. [PMID: 34800850 DOI: 10.1016/j.copbio.2021.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022]
Abstract
The neurovascular system (NVS) is a complex anatomic-functional unit that synergically works to maintain organs/tissues homeostasis of the entire body. NVS alterations have recently emerged as a common distinct feature in the pathogenesis of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Despite their undeniable involvement, neurovascular signalling pathways remain still far unknown in ALS. This review underlines the importance of endothelial, mural, and fibroblast cells as novel targets for ALS investigation and identifies in the interplay between neuronal and vascular systems the way to disclose novel molecular mechanisms behind the pathogenesis of ALS.
Collapse
Affiliation(s)
- Stefano Sorrentino
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Arima
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Angelo Quattrini
- San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Giuseppe Gigli
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, via Arnesano, 73100, Lecce, Italy
| | - Pamela Mozetic
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Lorenzo Moroni
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229ER, Maastricht, The Netherlands.
| |
Collapse
|
42
|
Mathur T, Tronolone JJ, Jain A. Comparative Analysis of Blood-Derived Endothelial Cells for Designing Next-Generation Personalized Organ-on-Chips. J Am Heart Assoc 2021; 10:e022795. [PMID: 34743553 PMCID: PMC8751908 DOI: 10.1161/jaha.121.022795] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Organ‐on‐chip technology has accelerated in vitro preclinical research of the vascular system, and a key strength of this platform is its promise to impact personalized medicine by providing a primary human cell–culture environment where endothelial cells are directly biopsied from individual tissue or differentiated through stem cell biotechniques. However, these methods are difficult to adopt in laboratories, and often result in impurity and heterogeneity of cells. This limits the power of organ‐chips in making accurate physiological predictions. In this study, we report the use of blood‐derived endothelial cells as alternatives to primary and induced pluripotent stem cell–derived endothelial cells. Methods and Results Here, the genotype, phenotype, and organ‐chip functional characteristics of blood‐derived outgrowth endothelial cells were compared against commercially available and most used primary endothelial cells and induced pluripotent stem cell–derived endothelial cells. The methods include RNA‐sequencing, as well as criterion standard assays of cell marker expression, growth kinetics, migration potential, and vasculogenesis. Finally, thromboinflammatory responses under shear using vessel‐chips engineered with blood‐derived endothelial cells were assessed. Blood‐derived endothelial cells exhibit the criterion standard hallmarks of typical endothelial cells. There are differences in gene expression profiles between different sources of endothelial cells, but blood‐derived cells are relatively closer to primary cells than induced pluripotent stem cell–derived. Furthermore, blood‐derived endothelial cells are much easier to obtain from individuals and yet, they serve as an equally effective cell source for functional studies and organ‐chips compared with primary cells or induced pluripotent stem cell–derived cells. Conclusions Blood‐derived endothelial cells may be used in preclinical research for developing more robust and personalized next‐generation disease models using organ‐on‐chips.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX
| | - James J Tronolone
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX.,Department of Medical Physiology College of MedicineTexas A&M Health Science Center Bryan TX.,Department of Cardiovascular Sciences Houston Methodist Research Institute Houston TX
| |
Collapse
|
43
|
Youn J, Han H, Park SM, Kim DS. Arterial Internal Elastic Lamina-Inspired Membrane for Providing Biochemical and Structural Cues in Developing Artery-on-a-Chip. ACS Macro Lett 2021; 10:1398-1403. [PMID: 35549015 DOI: 10.1021/acsmacrolett.1c00551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vitro artery models constructed on a membrane-based microfluidic chip, called an artery-on-a-chip, have been spotlighted as a powerful platform for studying arterial physiology. However, due to the use of a flat and porous membrane that cannot mimic the in vivo internal elastic lamina (IEL), the physiological similarity in the phenotypes and the arrangements of the endothelial cells (ECs) and aortic smooth muscle cells (AoSMCs) has been limited in the previously developed artery-on-a-chips. Herein, we developed an innovative membrane mimicking the structures of IEL by utilizing electrospun aligned silk fibroin/polycaprolactone nanofiber membranes. An arterial IEL-mimicking (AIM) membrane was about 5 μm thick and composed of orthogonally aligned nanofibers with a diameter of around 400 nm, which were highly comparable to the IEL. Such structural similarity was found to induce the ECs and SMCs to be elongated and orthogonally aligned as in the in vivo artery. In particular, the SMCs cultured on the AIM membrane maintained a healthy state showing increased αSMA mRNA expression, which was easily lost on the conventional membrane. We constructed an AIM membrane-integrated artery-on-a-chip having an orthogonal arrangement of ECs and SMCs, which was desirable but difficult to be realized with the previous artery-on-a-chip.
Collapse
Affiliation(s)
- Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Hyeonseok Han
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Sang Min Park
- School of Mechanical Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea.,Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, South Korea
| |
Collapse
|
44
|
Dessalles CA, Ramón-Lozano C, Babataheri A, Barakat AI. Luminal flow actuation generates coupled shear and strain in a microvessel-on-chip. Biofabrication 2021; 14. [PMID: 34592728 DOI: 10.1088/1758-5090/ac2baa] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
In the microvasculature, blood flow-derived forces are key regulators of vascular structure and function. Consequently, the development of hydrogel-based microvessel-on-chip systems that strive to mimic thein vivocellular organization and mechanical environment has received great attention in recent years. However, despite intensive efforts, current microvessel-on-chip systems suffer from several limitations, most notably failure to produce physiologically relevant wall strain levels. In this study, a novel microvessel-on-chip based on the templating technique and using luminal flow actuation to generate physiologically relevant levels of wall shear stress and circumferential stretch is presented. Normal forces induced by the luminal pressure compress the surrounding soft collagen hydrogel, dilate the channel, and create large circumferential strain. The fluid pressure gradient in the system drives flow forward and generates realistic pulsatile wall shear stresses. Rigorous characterization of the system reveals the crucial role played by the poroelastic behavior of the hydrogel in determining the magnitudes of the wall shear stress and strain. The experimental measurements are combined with an analytical model of flow in both the lumen and the porous hydrogel to provide an exceptionally versatile user manual for an application-based choice of parameters in microvessels-on-chip. This unique strategy of flow actuation adds a dimension to the capabilities of microvessel-on-chip systems and provides a more general framework for improving hydrogel-basedin vitroengineered platforms.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Clara Ramón-Lozano
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Avin Babataheri
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| |
Collapse
|
45
|
Pun S, Haney LC, Barrile R. Modelling Human Physiology on-Chip: Historical Perspectives and Future Directions. MICROMACHINES 2021; 12:1250. [PMID: 34683301 PMCID: PMC8540847 DOI: 10.3390/mi12101250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 01/09/2023]
Abstract
For centuries, animal experiments have contributed much to our understanding of mechanisms of human disease, but their value in predicting the effectiveness of drug treatments in the clinic has remained controversial. Animal models, including genetically modified ones and experimentally induced pathologies, often do not accurately reflect disease in humans, and therefore do not predict with sufficient certainty what will happen in humans. Organ-on-chip (OOC) technology and bioengineered tissues have emerged as promising alternatives to traditional animal testing for a wide range of applications in biological defence, drug discovery and development, and precision medicine, offering a potential alternative. Recent technological breakthroughs in stem cell and organoid biology, OOC technology, and 3D bioprinting have all contributed to a tremendous progress in our ability to design, assemble and manufacture living organ biomimetic systems that more accurately reflect the structural and functional characteristics of human tissue in vitro, and enable improved predictions of human responses to drugs and environmental stimuli. Here, we provide a historical perspective on the evolution of the field of bioengineering, focusing on the most salient milestones that enabled control of internal and external cell microenvironment. We introduce the concepts of OOCs and Microphysiological systems (MPSs), review various chip designs and microfabrication methods used to construct OOCs, focusing on blood-brain barrier as an example, and discuss existing challenges and limitations. Finally, we provide an overview on emerging strategies for 3D bioprinting of MPSs and comment on the potential role of these devices in precision medicine.
Collapse
Affiliation(s)
- Sirjana Pun
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA; (S.P.); (L.C.H.)
| | - Li Cai Haney
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA; (S.P.); (L.C.H.)
| | - Riccardo Barrile
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA; (S.P.); (L.C.H.)
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45221, USA
| |
Collapse
|
46
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
47
|
Lust ST, Shanahan CM, Shipley RJ, Lamata P, Gentleman E. Design considerations for engineering 3D models to study vascular pathologies in vitro. Acta Biomater 2021; 132:114-128. [PMID: 33652164 PMCID: PMC7611653 DOI: 10.1016/j.actbio.2021.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Many cardiovascular diseases (CVD) are driven by pathological remodelling of blood vessels, which can lead to aneurysms, myocardial infarction, ischaemia and strokes. Aberrant remodelling is driven by changes in vascular cell behaviours combined with degradation, modification, or abnormal deposition of extracellular matrix (ECM) proteins. The underlying mechanisms that drive the pathological remodelling of blood vessels are multifaceted and disease specific; however, unravelling them may be key to developing therapies. Reductionist models of blood vessels created in vitro that combine cells with biomaterial scaffolds may serve as useful analogues to study vascular disease progression in a controlled environment. This review presents the main considerations for developing such in vitro models. We discuss how the design of blood vessel models impacts experimental readouts, with a particular focus on the maintenance of normal cellular phenotypes, strategies that mimic normal cell-ECM interactions, and approaches that foster intercellular communication between vascular cell types. We also highlight how choice of biomaterials, cellular arrangements and the inclusion of mechanical stimulation using fluidic devices together impact the ability of blood vessel models to mimic in vivo conditions. In the future, by combining advances in materials science, cell biology, fluidics and modelling, it may be possible to create blood vessel models that are patient-specific and can be used to develop and test therapies. STATEMENT OF SIGNIFICANCE: Simplified models of blood vessels created in vitro are powerful tools for studying cardiovascular diseases and understanding the mechanisms driving their progression. Here, we highlight the key structural and cellular components of effective models and discuss how including mechanical stimuli allows researchers to mimic native vessel behaviour in health and disease. We discuss the primary methods used to form blood vessel models and their limitations and conclude with an outlook on how blood vessel models that incorporate patient-specific cells and flows can be used in the future for personalised disease modelling.
Collapse
Affiliation(s)
- Suzette T Lust
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Catherine M Shanahan
- School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Rebecca J Shipley
- Institute of Healthcare Engineering and Department of Mechanical Engineering, University College London, London WC1E 7JE, United Kingdom
| | - Pablo Lamata
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
48
|
Engineered 3D vessel-on-chip using hiPSC-derived endothelial- and vascular smooth muscle cells. Stem Cell Reports 2021; 16:2159-2168. [PMID: 34478648 PMCID: PMC8452600 DOI: 10.1016/j.stemcr.2021.08.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Crosstalk between endothelial cells (ECs) and pericytes or vascular smooth muscle cells (VSMCs) is essential for the proper functioning of blood vessels. This balance is disrupted in several vascular diseases but there are few experimental models which recapitulate this vascular cell dialogue in humans. Here, we developed a robust multi-cell type 3D vessel-on-chip (VoC) model based entirely on human induced pluripotent stem cells (hiPSCs). Within a fibrin hydrogel microenvironment, the hiPSC-derived vascular cells self-organized to form stable microvascular networks reproducibly, in which the vessels were lumenized and functional, responding as expected to vasoactive stimulation. Vascular organization and intracellular Ca2+ release kinetics in VSMCs could be quantified using automated image analysis based on open-source software CellProfiler and ImageJ on widefield or confocal images, setting the stage for use of the platform to study vascular (patho)physiology and therapy. 3D VoC formed by hiPSC-ECs and hiPSC-VSMCs Vascular organization in 3D VoC formed by hiPSC-VSMC and primary mural cells Functional responses of hiPSC-VSMCs in 3D VoC Automated analysis of microvascular network morphology and Ca2+ release in VSMCs
Collapse
|
49
|
Stein JM, Mummery CL, Bellin M. Engineered models of the human heart: Directions and challenges. Stem Cell Reports 2021; 16:2049-2057. [PMID: 33338434 PMCID: PMC8452488 DOI: 10.1016/j.stemcr.2020.11.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Human heart (patho)physiology is now widely studied using human pluripotent stem cells, but the immaturity of derivative cardiomyocytes has largely limited disease modeling to conditions associated with mutations in cardiac ion channel genes. Recent advances in tissue engineering and organoids have, however, created new opportunities to study diseases beyond "channelopathies." These synthetic cardiac structures allow quantitative measurement of contraction, force, and other biophysical parameters in three-dimensional configurations, in which the cardiomyocytes in addition become more mature. Multiple cardiac-relevant cell types are also often combined to form organized cardiac tissue mimetic constructs, where cell-cell, cell-extracellular matrix, and paracrine interactions can be mimicked. In this review, we provide an overview of some of the most promising technologies being implemented specifically in personalized heart-on-a-chip models and explore their applications, drawbacks, and potential for future development.
Collapse
Affiliation(s)
- Jeroen M Stein
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands; Department of Applied Stem Cell Technologies, University of Twente, Enschede 7500AE, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands; Department of Biology, University of Padua, Padua 35131, Italy; Veneto Institute of Molecular Medicine, Padua 35129, Italy.
| |
Collapse
|
50
|
Malik M, Yang Y, Fathi P, Mahler GJ, Esch MB. Critical Considerations for the Design of Multi-Organ Microphysiological Systems (MPS). Front Cell Dev Biol 2021; 9:721338. [PMID: 34568333 PMCID: PMC8459628 DOI: 10.3389/fcell.2021.721338] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Identification and approval of new drugs for use in patients requires extensive preclinical studies and clinical trials. Preclinical studies rely on in vitro experiments and animal models of human diseases. The transferability of drug toxicity and efficacy estimates to humans from animal models is being called into question. Subsequent clinical studies often reveal lower than expected efficacy and higher drug toxicity in humans than that seen in animal models. Microphysiological systems (MPS), sometimes called organ or human-on-chip models, present a potential alternative to animal-based models used for drug toxicity screening. This review discusses multi-organ MPS that can be used to model diseases and test the efficacy and safety of drug candidates. The translation of an in vivo environment to an in vitro system requires physiologically relevant organ scaling, vascular dimensions, and appropriate flow rates. Even small changes in those parameters can alter the outcome of experiments conducted with MPS. With many MPS devices being developed, we have outlined some established standards for designing MPS devices and described techniques to validate the devices. A physiologically realistic mimic of the human body can help determine the dose response and toxicity effects of a new drug candidate with higher predictive power.
Collapse
Affiliation(s)
- Mridu Malik
- Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| | - Yang Yang
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
- Department of Chemical Engineering, University of Maryland, College Park, College Park, MD, United States
| | - Parinaz Fathi
- Department of Bioengineering, Materials Science and Engineering, and Beckman Institute, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| | - Mandy B. Esch
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| |
Collapse
|