1
|
Singh J, Ruhoff AM, Ashok D, Wise SG, Waterhouse A. Engineering advanced in vitro models of endothelial dysfunction. Trends Biotechnol 2025:S0167-7799(25)00089-7. [PMID: 40187930 DOI: 10.1016/j.tibtech.2025.03.004] [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: 08/29/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Endothelial dysfunction is an important initiator of cardiovascular disease, the leading cause of death globally, and often manifests in arterial regions with disturbed blood flow. Experimental model advances have crucially helped unravel physiological mechanisms. While in vivo models provide a dynamic environment, they often fail to mimic human physiology precisely and face significant ethical barriers. Advanced in vitro models, including organs-on-chips and bioreactors, combine human cells and blood flow to accurately replicate endothelial dysfunction. Newer models have enhanced scalability and accuracy, with organs-on-chips commonly outperforming standard preclinical methods. Importantly, recent endothelial dysfunction discoveries leverage dynamic models to identify and evaluate clinically promising therapeutics. Here, we examine these developments and explore opportunities to develop next-generation in vitro models of endothelial dysfunction.
Collapse
Affiliation(s)
- Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Ruhoff
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Deepu Ashok
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Steven G Wise
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
2
|
Zhong J, Gao RR, Zhang X, Yang JX, Liu Y, Ma J, Chen Q. Dissecting endothelial cell heterogeneity with new tools. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:10. [PMID: 40121354 PMCID: PMC11929667 DOI: 10.1186/s13619-025-00223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The formation of a blood vessel network is crucial for organ development and regeneration. Over the past three decades, the central molecular mechanisms governing blood vessel growth have been extensively studied. Recent evidence indicates that vascular endothelial cells-the specialized cells lining the inner surface of blood vessels-exhibit significant heterogeneity to meet the specific needs of different organs. This review focuses on the current understanding of endothelial cell heterogeneity, which includes both intra-organ and inter-organ heterogeneity. Intra-organ heterogeneity encompasses arterio-venous and tip-stalk endothelial cell specialization, while inter-organ heterogeneity refers to organ-specific transcriptomic profiles and functions. Advances in single-cell RNA sequencing (scRNA-seq) have enabled the identification of new endothelial subpopulations and the comparison of gene expression patterns across different subsets of endothelial cells. Integrating scRNA-seq with other high-throughput sequencing technologies promises to deepen our understanding of endothelial cell heterogeneity at the epigenetic level and in a spatially resolved context. To further explore human endothelial cell heterogeneity, vascular organoids offer powerful tools for studying gene function in three-dimensional culture systems and for investigating endothelial-tissue interactions using human cells. Developing organ-specific vascular organoids presents unique opportunities to unravel inter-organ endothelial cell heterogeneity and its implications for human disease. Emerging technologies, such as scRNA-seq and vascular organoids, are poised to transform our understanding of endothelial cell heterogeneity and pave the way for innovative therapeutic strategies to address human vascular diseases.
Collapse
Affiliation(s)
- Jing Zhong
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Rong-Rong Gao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China
| | - Xin Zhang
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jia-Xin Yang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Jinjin Ma
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- The Institute of Future Health, South China of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Qi Chen
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China.
| |
Collapse
|
3
|
Mei X, Yang Z, Wang X, Shi A, Blanchard J, Elahi F, Kang H, Orive G, Zhang YS. Integrating microfluidic and bioprinting technologies: advanced strategies for tissue vascularization. LAB ON A CHIP 2025; 25:764-786. [PMID: 39775452 DOI: 10.1039/d4lc00280f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tissue engineering offers immense potential for addressing the unmet needs in repairing tissue damage and organ failure. Vascularization, the development of intricate blood vessel networks, is crucial for the survival and functions of engineered tissues. Nevertheless, the persistent challenge of ensuring an ample nutrient supply within implanted tissues remains, primarily due to the inadequate formation of blood vessels. This issue underscores the vital role of the human vascular system in sustaining cellular functions, facilitating nutrient exchange, and removing metabolic waste products. In response to this challenge, new approaches have been explored. Microfluidic devices, emulating natural blood vessels, serve as valuable tools for investigating angiogenesis and allowing the formation of microvascular networks. In parallel, bioprinting technologies enable precise placement of cells and biomaterials, culminating in vascular structures that closely resemble the native vessels. To this end, the synergy of microfluidics and bioprinting has further opened up exciting possibilities in vascularization, encompassing innovations such as microfluidic bioprinting. These advancements hold great promise in regenerative medicine, facilitating the creation of functional tissues for applications ranging from transplantation to disease modeling and drug testing. This review explores the potentially transformative impact of microfluidic and bioprinting technologies on vascularization strategies within the scope of tissue engineering.
Collapse
Affiliation(s)
- Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| | - Ziyi Yang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
- School of Biological Science, University of California Irvine, Irvine, CA 92697, USA
| | - Xiran Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA 92161, USA
| | - Alan Shi
- Brookline High School, Brookline, MA 02445, USA
| | - Joel Blanchard
- Departments of Neurology, Neuroscience, and Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fanny Elahi
- Departments of Neurology, Neuroscience, and Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea.
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, 01007, Spain
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
4
|
Sun Y, Liu J, Zhu L, Huang F, Dong Y, Liu S, Chen S, Ji W, Lu J, Liu L, Li S. Treatment Response to Oncolytic Virus in Patient-Derived Breast Cancer and Hypopharyngeal Cancer Organoids: Evaluation via a Microfluidics Organ-on-a-Chip System. Bioengineering (Basel) 2025; 12:146. [PMID: 40001666 PMCID: PMC11851931 DOI: 10.3390/bioengineering12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and minimal side effects, and it has achieved significant breakthroughs when combined with immunotherapy in recent clinical trials. The development of OVs has become an emerging research focus. PDOs can preserve the heterogeneity of in situ tumor tissues, whereas microfluidic OOC systems can automate and standardize various experimental procedures. These systems have been applied in cutting-edge drug screening and cell therapy experiments; however, their use in functionally complex oncolytic viruses remains to be explored. In this study, we constructed a novel recombinant oncolytic adenovirus, AD4-GHPE, and evaluated OOC systems and PDOs through various functional validations in hypopharyngeal and breast cancer organoids. The results confirmed that AD4-GHPE exhibits three antitumor mechanisms, namely, tumor-specific cytotoxicity, a reduction in programmed death ligand 1 (PD-L1) expression in tumor cells to increase CD8+ T-cell activity, and granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion. The evaluation system combining OOC systems and PDOs was efficient and reliable, providing personalized OV treatment recommendations for patients and offering industrialized and standardized research ideas for the development of OVs.
Collapse
Affiliation(s)
- Yu Sun
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jiaqi Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Li Zhu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Yanbo Dong
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shuang Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Siyi Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wei Ji
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jingjing Lu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Liangfa Liu
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| |
Collapse
|
5
|
Gonzalez G, Molley TG, LaMontagne E, Balayan A, Holman AR, Engler AJ. Conductive Microfibers Improve Stem Cell-Derived Cardiac Spheroid Maturation. J Biomed Mater Res A 2025; 113:e37856. [PMID: 39719888 PMCID: PMC11703634 DOI: 10.1002/jbm.a.37856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024]
Abstract
Conventional two-dimensional (2D) cardiomyocyte differentiation protocols create cells with limited maturity, which impairs their predictive capacity and has driven interest in three-dimensional (3D) engineered cardiac tissue models of varying maturity and scalability. Cardiac spheroids are attractive high-throughput models that have demonstrated improved functional and transcriptional maturity over conventional 2D differentiations. However, these 3D models still tend to have limited contractile and electrical maturity compared to highly engineered cardiac tissues; hence, we incorporated a library of conductive polymer microfibers in cardiac spheroids to determine if fiber properties could accelerate maturation. Conductive microfibers improved contractility parameters of cardiac spheroids over time versus nonconductive fibers, specifically, when they were short, for example, 5 μm, and when there was moderate fiber mass per spheroid, for example, 20 μg. Spheroids with optimal conductive microfiber length and concentration developed a thicker ring-like perimeter and a less compacted cavity, improving their contractile work compared to control cardiac spheroids. Functional improvements correlated with increased expression of contractility and calcium handling-related cardiac proteins, as well as improved calcium handling abilities and drug response. Taken together, these data suggest that conductive microfibers can improve cardiac spheroid performance to improve cardiac disease modeling.
Collapse
Affiliation(s)
- Gisselle Gonzalez
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego; La Jolla, CA, USA 92093
| | - Thomas G. Molley
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego; La Jolla, CA, USA 92093
| | - Erin LaMontagne
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego; La Jolla, CA, USA 92093
| | - Alis Balayan
- Biomedical Sciences Graduate Program; University of California San Diego; La Jolla, CA, USA 92093
| | - Alyssa R. Holman
- Biomedical Sciences Graduate Program; University of California San Diego; La Jolla, CA, USA 92093
| | - Adam J. Engler
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Graduate Program; University of California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| |
Collapse
|
6
|
Hu Y, Zhu T, Cui H, Cui H. Integrating 3D Bioprinting and Organoids to Better Recapitulate the Complexity of Cellular Microenvironments for Tissue Engineering. Adv Healthc Mater 2025; 14:e2403762. [PMID: 39648636 DOI: 10.1002/adhm.202403762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/16/2024] [Indexed: 12/10/2024]
Abstract
Organoids, with their capacity to mimic the structures and functions of human organs, have gained significant attention for simulating human pathophysiology and have been extensively investigated in the recent past. Additionally, 3D bioprinting, as an emerging bio-additive manufacturing technology, offers the potential for constructing heterogeneous cellular microenvironments, thereby promoting advancements in organoid research. In this review, the latest developments in 3D bioprinting technologies aimed at enhancing organoid engineering are introduced. The commonly used bioprinting methods and materials for organoids, with a particular emphasis on the potential advantages of combining 3D bioprinting with organoids are summarized. These advantages include achieving high cell concentrations to form large cellular aggregates, precise deposition of building blocks to create organoids with complex structures and functions, and automation and high throughput to ensure reproducibility and standardization in organoid culture. Furthermore, this review provides an overview of relevant studies from recent years and discusses the current limitations and prospects for future development.
Collapse
Affiliation(s)
- Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tong Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haijun Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
7
|
Wang H, Zhu W, Xu C, Su W, Li Z. Engineering organoids-on-chips for drug testing and evaluation. Metabolism 2025; 162:156065. [PMID: 39522593 DOI: 10.1016/j.metabol.2024.156065] [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: 07/01/2024] [Revised: 09/21/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Organoids-on-chips is an emerging innovative integration of stem cell-derived organoids with advanced organ-on-chip technology, providing a novel platform for the in vitro construction of biomimetic micro-physiological systems. The synergistic merger transcends the limitations of traditional drug screening and safety assessment methodologies, such as 2D cell cultures and animal models. In this review, we examine the prevailing challenges and prerequisites of preclinical models utilized for drug screening and safety evaluations. We highlighted the salient features and merits of organoids-on-chip, elucidating their capability to authentically replicate human physiology, thereby addressing contemporary impediments. We comprehensively overviewed the recent endeavors where organoids-on-chips have been harnessed for drug screening and safety assessment and delved into potential opportunities and challenges for evolving sophisticated, near-physiological organoids-on-chips. Based on current achievements, we further discuss how to enhance the practicality of organoids-on-chips and accelerate the translation from preclinical to clinical stages in healthcare and industry by utilizing multidisciplinary convergent innovation.
Collapse
Affiliation(s)
- Hui Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wan Zhu
- Shanghai General Hospital, Shanghai 200080, China
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University Medical Center, New York 10032, USA
| | - Wentao Su
- Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, Liaoning, China.
| | - Zhongyu Li
- College of Life Science, Dalian Minzu University, Dalian 116600, China.
| |
Collapse
|
8
|
Liu KZ, Tian G, Ko ACT, Geissler M, Malic L, Moon BU, Clime L, Veres T. Microfluidic methods for the diagnosis of acute respiratory tract infections. Analyst 2024; 150:9-33. [PMID: 39440426 DOI: 10.1039/d4an00957f] [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: 10/25/2024]
Abstract
Acute respiratory tract infections (ARTIs) are caused by sporadic or pandemic outbreaks of viral or bacterial pathogens, and continue to be a considerable socioeconomic burden for both developing and industrialized countries alike. Diagnostic methods and technologies serving as the cornerstone for disease management, epidemiological tracking, and public health interventions are evolving continuously to keep up with the demand for higher sensitivity, specificity and analytical throughput. Microfluidics is becoming a key technology in these developments as it allows for integrating, miniaturizing and automating bioanalytical assays at an unprecedented scale, reducing sample and reagent consumption and improving diagnostic performance in terms of sensitivity, throughput and response time. In this article, we describe relevant ARTIs-pneumonia, influenza, severe acute respiratory syndrome, and coronavirus disease 2019-along with their pathogenesis. We provide a summary of established methods for disease diagnosis, involving nucleic acid amplification techniques, antigen detection, serological testing as well as microbial culture. This is followed by a short introduction to microfluidics and how flow is governed at low volume and reduced scale using centrifugation, pneumatic pumping, electrowetting, capillary action, and propagation in porous media through wicking, for each of these principles impacts the design, functioning and performance of diagnostic tools in a particular way. We briefly cover commercial instruments that employ microfluidics for use in both laboratory and point-of-care settings. The main part of the article is dedicated to emerging methods deriving from the use of miniaturized, microfluidic systems for ARTI diagnosis. Finally, we share our thoughts on future perspectives and the challenges associated with validation, approval, and adaptation of microfluidic-based systems.
Collapse
Affiliation(s)
- Kan-Zhi Liu
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Ganghong Tian
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Alex C-T Ko
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Matthias Geissler
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Lidija Malic
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Byeong-Ui Moon
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Liviu Clime
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Teodor Veres
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| |
Collapse
|
9
|
Blazeski A, Garcia-Cardena G, Kamm RD. Advancing Cardiac Organoid Engineering Through Application of Biophysical Forces. IEEE Rev Biomed Eng 2024; PP:211-230. [PMID: 40030454 DOI: 10.1109/rbme.2024.3514378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cardiac organoids represent an important bioengineering opportunity in the development of models to study human heart pathophysiology. By incorporating multiple cardiac cell types in three-dimensional culture and developmentally-guided biochemical signaling, cardiac organoids recapitulate numerous features of heart tissue. However, cardiac tissue also experiences a variety of mechanical forces as the heart develops and over the course of each contraction cycle. It is now clear that these forces impact cellular specification, phenotype, and function, and should be incorporated into the engineering of cardiac organoids in order to generate better models. In this review, we discuss strategies for engineering cardiac organoids and report the effects of organoid design on the function of cardiac cells. We then discuss the mechanical environment of the heart, including forces arising from tissue elasticity, contraction, blood flow, and stretch, and report on efforts to mimic these biophysical cues in cardiac organoids. Finally, we review emerging areas of cardiac organoid research, for the study of cardiac development, the formation of multi-organ models, and the simulation of the effects of spaceflight on cardiac tissue and consider how these investigations might benefit from the inclusion of mechanical cues.
Collapse
|
10
|
van Doorn ECH, Amesz JH, Manintveld OC, de Groot NMS, Essers J, Shin SR, Taverne YJHJ. Advancing 3D Engineered In Vitro Models for Heart Failure Research: Key Features and Considerations. Bioengineering (Basel) 2024; 11:1220. [PMID: 39768038 PMCID: PMC11673263 DOI: 10.3390/bioengineering11121220] [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: 10/08/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025] Open
Abstract
Heart failure is characterized by intricate myocardial remodeling that impairs the heart's pumping and/or relaxation capacity, ultimately reducing cardiac output. It represents a major public health burden, given its high prevalence and associated morbidity and mortality rates, which continue to challenge healthcare systems worldwide. Despite advancements in medical science, there are no treatments that address the disease at its core. The development of three-dimensional engineered in vitro models that closely mimic the (patho)physiology and drug responses of the myocardium has the potential to revolutionize our insights and uncover new therapeutic avenues. Key aspects of these models include the precise replication of the extracellular matrix structure, cell composition, micro-architecture, mechanical and electrical properties, and relevant physiological and pathological stimuli, such as fluid flow, mechanical load, electrical signal propagation, and biochemical cues. Additionally, to fully capture heart failure and its diversity in vivo, it is crucial to consider factors such as age, gender, interactions with other organ systems and external influences-thereby recapitulating unique patient and disease phenotypes. This review details these model features and their significance in heart failure research, with the aim of enhancing future platforms that will deepen our understanding of the disease and facilitate the development of novel, effective therapies.
Collapse
Affiliation(s)
- Elisa C. H. van Doorn
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (E.C.H.v.D.); (J.H.A.)
- Department of Cardiology, Cardiovascular Institute, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (O.C.M.); (N.M.S.d.G.)
| | - Jorik H. Amesz
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (E.C.H.v.D.); (J.H.A.)
- Department of Cardiology, Cardiovascular Institute, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (O.C.M.); (N.M.S.d.G.)
| | - Olivier C. Manintveld
- Department of Cardiology, Cardiovascular Institute, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (O.C.M.); (N.M.S.d.G.)
| | - Natasja M. S. de Groot
- Department of Cardiology, Cardiovascular Institute, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (O.C.M.); (N.M.S.d.G.)
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus Medical Centre, 3015 GD Rotterdam, The Netherlands;
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA;
| | - Yannick J. H. J. Taverne
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (E.C.H.v.D.); (J.H.A.)
| |
Collapse
|
11
|
Landau S, Okhovatian S, Zhao Y, Liu C, Shakeri A, Wang Y, Ramsay K, Kieda J, Jiang R, Radisic M. Bioengineering vascularization. Development 2024; 151:dev204455. [PMID: 39611864 PMCID: PMC11698057 DOI: 10.1242/dev.204455] [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: 11/30/2024]
Abstract
This Review explores the rapidly evolving field of bioengineered vasculature, a key area of focus in tissue engineering and regenerative medicine. The broad relevance of this topic is attributed to its impacts on a wide range of biological processes, enabling studies in tissue development, fundamental biology and drug discovery, and the applications in tissue engineering and regenerative medicine. We outline the design criteria for bioengineered vasculature and the methodologies for constructing these systems by self-assembly and in microfluidics, organs-on-a-chip and macroscale tubular systems that often rely on biofabrication approaches such as 3D printing. We discuss existing challenges in developing functional vasculature that closely mirrors its native equivalent, including achieving hierarchical branching with organ and vessel-specific endothelial and supporting cells, providing perusable vasculature within organoids and scaling the systems for implantation and direct vascular anastomosis.
Collapse
Affiliation(s)
- Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto M5G 1X6, ON, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Kaitlyn Ramsay
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto M5S 3E5, ON, Canada
- Terence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, ON, Canada
| |
Collapse
|
12
|
Tong L, Cui W, Zhang B, Fonseca P, Zhao Q, Zhang P, Xu B, Zhang Q, Li Z, Seashore-Ludlow B, Yang Y, Si L, Lundqvist A. Patient-derived organoids in precision cancer medicine. MED 2024; 5:1351-1377. [PMID: 39341206 DOI: 10.1016/j.medj.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024]
Abstract
Organoids are three-dimensional (3D) cultures, normally derived from stem cells, that replicate the complex structure and function of human tissues. They offer a physiologically relevant model to address important questions in cancer research. The generation of patient-derived organoids (PDOs) from various human cancers allows for deeper insights into tumor heterogeneity and spatial organization. Additionally, interrogating non-tumor stromal cells increases the relevance in studying the tumor microenvironment, thereby enhancing the relevance of PDOs in personalized medicine. PDOs mark a significant advancement in cancer research and patient care, signifying a shift toward more innovative and patient-centric approaches. This review covers aspects of PDO cultures to address the modeling of the tumor microenvironment, including extracellular matrices, air-liquid interface and microfluidic cultures, and organ-on-chip. Specifically, the role of PDOs as preclinical models in gene editing, molecular profiling, drug testing, and biomarker discovery and their potential for guiding personalized treatment in clinical practice are discussed.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Weiyingqi Cui
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Boya Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Pedro Fonseca
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Qian Zhao
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Ping Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Beibei Xu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qisi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Wagner KT, Lu RXZ, Landau S, Shawky SA, Zhao Y, Bodenstein DF, Jiménez Vargas LF, Jiang R, Okhovatian S, Wang Y, Liu C, Vosoughi D, Gustafson D, Fish JE, Cummins CL, Radisic M. Endothelial extracellular vesicles enhance vascular self-assembly in engineered human cardiac tissues. Biofabrication 2024; 16:045037. [PMID: 39226913 PMCID: PMC11409464 DOI: 10.1088/1758-5090/ad76d9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/03/2024] [Indexed: 09/05/2024]
Abstract
The fabrication of complex and stable vasculature in engineered cardiac tissues represents a significant hurdle towards building physiologically relevant models of the heart. Here, we implemented a 3D model of cardiac vasculogenesis, incorporating endothelial cells (EC), stromal cells, and human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) in a fibrin hydrogel. The presence of CMs disrupted vessel formation in 3D tissues, resulting in the upregulation of endothelial activation markers and altered extracellular vesicle (EV) signaling in engineered tissues as determined by the proteomic analysis of culture supernatant. miRNA sequencing of CM- and EC-secreted EVs highlighted key EV-miRNAs that were postulated to play differing roles in cardiac vasculogenesis, including the let-7 family and miR-126-3p in EC-EVs. In the absence of CMs, the supplementation of CM-EVs to EC monolayers attenuated EC migration and proliferation and resulted in shorter and more discontinuous self-assembling vessels when applied to 3D vascular tissues. In contrast, supplementation of EC-EVs to the tissue culture media of 3D vascularized cardiac tissues mitigated some of the deleterious effects of CMs on vascular self-assembly, enhancing the average length and continuity of vessel tubes that formed in the presence of CMs. Direct transfection validated the effects of the key EC-EV miRNAs let-7b-5p and miR-126-3p in improving the maintenance of continuous vascular networks. EC-EV supplementation to biofabricated cardiac tissues and microfluidic devices resulted in tissue vascularization, illustrating the use of this approach in the engineering of enhanced, perfusable, microfluidic models of the myocardium.
Collapse
Affiliation(s)
- Karl T Wagner
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Rick X Z Lu
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Sarah A Shawky
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, ON M5S 3M2, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - David F Bodenstein
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
| | - Luis Felipe Jiménez Vargas
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Daniel Vosoughi
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Dakota Gustafson
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital,University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, ON M5S 3M2, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
14
|
Su R, Ai Y, Wang J, Wu L, Sun H, Ding M, Xie R, Liang Q. Engineered Microfibers for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5823-5840. [PMID: 39145987 DOI: 10.1021/acsabm.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Hydrogel microfibers are hydrogel materials engineered into fiber structures. Techniques such as wet spinning, microfluidic spinning, and 3D bioprinting are often used to prepare microfibers due to their ability to precisely control the size, morphology, and structure of the microfibers. Microfibers with different structural morphologies have different functions; they provide a flow-through culture environment for cells to improve viability, and can also be used to induce the differentiation of cells such as skeletal muscle and cardiac muscle cells to eventually form functional organs in vitro through special morphologies. This Review introduces recent advances in microfluidics, 3D bioprinting, and wet spinning in the preparation of microfibers, focusing on the materials and fabrication methods. The applications of microfibers in tissue engineering are highlighted by summarizing their contributions in engineering biomimetic blood vessels, vascularized tissues, bone, heart, pancreas, kidney, liver, and fat. Furthermore, applications of engineered fibers in tissue repair and drug screening are also discussed.
Collapse
Affiliation(s)
- Riguga Su
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Jingyu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ruoxiao Xie
- Department of Materials, Design and Manufacturing Engineering, School of Engineering, University of Liverpool, Liverpool L69 3BX, U.K
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
15
|
Gurwitz D, Steeg R. Enriching iPSC research diversity: Harnessing human biobank collections for improved ethnic representation. Drug Dev Res 2024; 85:e22227. [PMID: 38943497 DOI: 10.1002/ddr.22227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
Biobanks of human biosamples and cell lines are indispensable for biomedical research on human health and disease and for drug development projects. Many human cell line biobanks worldwide hold collections of lymphoblastoid cell lines (LCLs), representing thousands of affected and control donors from diverse ethnic/ancestry groups. In recent years, induced human pluripotent stem cells (iPSCs) and differentiated human cells derived from these iPSCs have become indispensable for applied biomedical research. Establishing iPSCs remains a laborious and costly step towards generating differentiated human cells. To address this research need, several non-profit and commercial biobanks have established iPSC collections for distribution to researchers, thereby serving as a resource for generating differentiated human cells. The most common starting materials for generation of iPSCs are a skin biopsy for harvesting fibroblasts, or a blood sample for collection of peripheral blood mononuclear cells. However untapped resources include the large established collections of biobanked human LCLs which can be reprogrammed to iPSCs using a variety of published protocols including the use of non-integrating episomal vectors. Many biobanks curate LCLs from diverse ethnic/ancestry populations, an aspect largely absent in most established iPSC biobanks which tend to primarily reflect populations from developed countries. Here, we call upon researchers across the breadth of iPSC research to tap the unique resource of existing and diverse human LCL collections for establishing biobanked iPSC panels that better represent the varied human ethnic (and hence genomic) diversity, thereby benefiting precision medicine and drug development research on a global scale.
Collapse
Affiliation(s)
- David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Rachel Steeg
- European Bank for Induced Pluripotent Stem Cells, Fraunhofer UK Research Ltd, Glasgow, UK
| |
Collapse
|
16
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
17
|
Lim J, Fang HW, Bupphathong S, Sung PC, Yeh CE, Huang W, Lin CH. The Edifice of Vasculature-On-Chips: A Focused Review on the Key Elements and Assembly of Angiogenesis Models. ACS Biomater Sci Eng 2024; 10:3548-3567. [PMID: 38712543 PMCID: PMC11167599 DOI: 10.1021/acsbiomaterials.3c01978] [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: 12/29/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
The conception of vascularized organ-on-a-chip models provides researchers with the ability to supply controlled biological and physical cues that simulate the in vivo dynamic microphysiological environment of native blood vessels. The intention of this niche research area is to improve our understanding of the role of the vasculature in health or disease progression in vitro by allowing researchers to monitor angiogenic responses and cell-cell or cell-matrix interactions in real time. This review offers a comprehensive overview of the essential elements, including cells, biomaterials, microenvironmental factors, microfluidic chip design, and standard validation procedures that currently govern angiogenesis-on-a-chip assemblies. In addition, we emphasize the importance of incorporating a microvasculature component into organ-on-chip devices in critical biomedical research areas, such as tissue engineering, drug discovery, and disease modeling. Ultimately, advances in this area of research could provide innovative solutions and a personalized approach to ongoing medical challenges.
Collapse
Affiliation(s)
- Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsu-Wei Fang
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Po-Chan Sung
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
18
|
Gokhan I, Blum TS, Campbell SG. Engineered heart tissue: Design considerations and the state of the art. BIOPHYSICS REVIEWS 2024; 5:021308. [PMID: 38912258 PMCID: PMC11192576 DOI: 10.1063/5.0202724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Originally developed more than 20 years ago, engineered heart tissue (EHT) has become an important tool in cardiovascular research for applications such as disease modeling and drug screening. Innovations in biomaterials, stem cell biology, and bioengineering, among other fields, have enabled EHT technologies to recapitulate many aspects of cardiac physiology and pathophysiology. While initial EHT designs were inspired by the isolated-trabecula culture system, current designs encompass a variety of formats, each of which have unique strengths and limitations. In this review, we describe the most common EHT formats, and then systematically evaluate each aspect of their design, emphasizing the rational selection of components for each application.
Collapse
Affiliation(s)
| | - Thomas S. Blum
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | | |
Collapse
|
19
|
Lu RXZ, Zhao Y, Radisic M. The emerging role of heart-on-a-chip systems in delineating mechanisms of SARS-CoV-2-induced cardiac dysfunction. Bioeng Transl Med 2024; 9:e10581. [PMID: 38818123 PMCID: PMC11135153 DOI: 10.1002/btm2.10581] [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: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 06/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a major global health concern since its emergence in 2019, with over 680 million confirmed cases as of April 2023. While COVID-19 has been strongly associated with the development of cardiovascular complications, the specific mechanisms by which viral infection induces myocardial dysfunction remain largely controversial as studies have shown that the severe acute respiratory syndrome coronavirus-2 can lead to heart failure both directly, by causing damage to the heart cells, and indirectly, by triggering an inflammatory response throughout the body. In this review, we summarize the current understanding of potential mechanisms that drive heart failure based on in vitro studies. We also discuss the significance of three-dimensional heart-on-a-chip technology in the context of the current and future pandemics.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Yimu Zhao
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
| | - Milica Radisic
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of Chemical Engineering and Applied ChemistryUniversity of TorontoTorontoOntarioCanada
- Terence Donnelly Centre for Cellular & Biomolecular ResearchUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
20
|
Kieda J, Shakeri A, Landau S, Wang EY, Zhao Y, Lai BF, Okhovatian S, Wang Y, Jiang R, Radisic M. Advances in cardiac tissue engineering and heart-on-a-chip. J Biomed Mater Res A 2024; 112:492-511. [PMID: 37909362 PMCID: PMC11213712 DOI: 10.1002/jbm.a.37633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Recent advances in both cardiac tissue engineering and hearts-on-a-chip are grounded in new biomaterial development as well as the employment of innovative fabrication techniques that enable precise control of the mechanical, electrical, and structural properties of the cardiac tissues being modelled. The elongated structure of cardiomyocytes requires tuning of substrate properties and application of biophysical stimuli to drive its mature phenotype. Landmark advances have already been achieved with induced pluripotent stem cell-derived cardiac patches that advanced to human testing. Heart-on-a-chip platforms are now commonly used by a number of pharmaceutical and biotechnology companies. Here, we provide an overview of cardiac physiology in order to better define the requirements for functional tissue recapitulation. We then discuss the biomaterials most commonly used in both cardiac tissue engineering and heart-on-a-chip, followed by the discussion of recent representative studies in both fields. We outline significant challenges common to both fields, specifically: scalable tissue fabrication and platform standardization, improving cellular fidelity through effective tissue vascularization, achieving adult tissue maturation, and ultimately developing cryopreservation protocols so that the tissues are available off the shelf.
Collapse
Affiliation(s)
- Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Fook Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Lu RXZ, Rafatian N, Zhao Y, Wagner KT, Beroncal EL, Li B, Lee C, Chen J, Churcher E, Vosoughi D, Liu C, Wang Y, Baker A, Trahtemberg U, Li B, Pierro A, Andreazza AC, dos Santos CC, Radisic M. Cardiac tissue model of immune-induced dysfunction reveals the role of free mitochondrial DNA and the therapeutic effects of exosomes. SCIENCE ADVANCES 2024; 10:eadk0164. [PMID: 38536913 PMCID: PMC10971762 DOI: 10.1126/sciadv.adk0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024]
Abstract
Despite tremendous progress in the development of mature heart-on-a-chip models, human cell-based models of myocardial inflammation are lacking. Here, we bioengineered a vascularized heart-on-a-chip with circulating immune cells to model severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced acute myocarditis. We observed hallmarks of coronavirus disease (COVID-19)-induced myocardial inflammation, as the presence of immune cells augmented the secretion of proinflammatory cytokines, triggered progressive impairment of contractile function, and altered intracellular calcium transients. An elevation of circulating cell-free mitochondrial DNA (ccf-mtDNA) was measured first in the heart-on-a-chip and then validated in COVID-19 patients with low left ventricular ejection fraction, demonstrating that mitochondrial damage is an important pathophysiological hallmark of inflammation-induced cardiac dysfunction. Leveraging this platform in the context of SARS-CoV-2-induced myocardial inflammation, we established that administration of endothelial cell-derived exosomes effectively rescued the contractile deficit, normalized calcium handling, elevated the contraction force, and reduced the ccf-mtDNA and cytokine release via Toll-like receptor-nuclear factor κB signaling axis.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Naimeh Rafatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Karl T. Wagner
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Erika L. Beroncal
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bo Li
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Carol Lee
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Eryn Churcher
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Daniel Vosoughi
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Andrew Baker
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Uriel Trahtemberg
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Galilee Medical Center, Nahariya, Israel
| | - Bowen Li
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Agostino Pierro
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ana C. Andreazza
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Claudia C. dos Santos
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Mitochondrial Innovation Initiative, MITO2i, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3D5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1
| |
Collapse
|
22
|
Ko J, Song J, Choi N, Kim HN. Patient-Derived Microphysiological Systems for Precision Medicine. Adv Healthc Mater 2024; 13:e2303161. [PMID: 38010253 PMCID: PMC11469251 DOI: 10.1002/adhm.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Patient-derived microphysiological systems (P-MPS) have emerged as powerful tools in precision medicine that provide valuable insight into individual patient characteristics. This review discusses the development of P-MPS as an integration of patient-derived samples, including patient-derived cells, organoids, and induced pluripotent stem cells, into well-defined MPSs. Emphasizing the necessity of P-MPS development, its significance as a nonclinical assessment approach that bridges the gap between traditional in vitro models and clinical outcomes is highlighted. Additionally, guidance is provided for engineering approaches to develop microfluidic devices and high-content analysis for P-MPSs, enabling high biological relevance and high-throughput experimentation. The practical implications of the P-MPS are further examined by exploring the clinically relevant outcomes obtained from various types of patient-derived samples. The construction and analysis of these diverse samples within the P-MPS have resulted in physiologically relevant data, paving the way for the development of personalized treatment strategies. This study describes the significance of the P-MPS in precision medicine, as well as its unique capacity to offer valuable insights into individual patient characteristics.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano TechnologyGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
| | - Jiyoung Song
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
23
|
Esparza A, Jimenez N, Borrego EA, Browne S, Natividad-Diaz SL. Review: Human stem cell-based 3D in vitro angiogenesis models for preclinical drug screening applications. Mol Biol Rep 2024; 51:260. [PMID: 38302762 PMCID: PMC10834608 DOI: 10.1007/s11033-023-09048-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 11/06/2023] [Indexed: 02/03/2024]
Abstract
Vascular diseases are the underlying pathology in many life-threatening illnesses. Human cellular and molecular mechanisms involved in angiogenesis are complex and difficult to study in current 2D in vitro and in vivo animal models. Engineered 3D in vitro models that incorporate human pluripotent stem cell (hPSC) derived endothelial cells (ECs) and supportive biomaterials within a dynamic microfluidic platform provide a less expensive, more controlled, and reproducible platform to better study angiogenic processes in response to external chemical or physical stimulus. Current studies to develop 3D in vitro angiogenesis models aim to establish single-source systems by incorporating hPSC-ECs into biomimetic extracellular matrices (ECM) and microfluidic devices to create a patient-specific, physiologically relevant platform that facilitates preclinical study of endothelial cell-ECM interactions, vascular disease pathology, and drug treatment pharmacokinetics. This review provides a detailed description of the current methods used for the directed differentiation of human stem cells to endothelial cells and their use in engineered 3D in vitro angiogenesis models that have been developed within the last 10 years.
Collapse
Affiliation(s)
- Aibhlin Esparza
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Nicole Jimenez
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Edgar A Borrego
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Shane Browne
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA.
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
24
|
Pahuja A, Goux Corredera I, Moya-Rull D, Garreta E, Montserrat N. Engineering physiological environments to advance kidney organoid models from human pluripotent stem cells. Curr Opin Cell Biol 2024; 86:102306. [PMID: 38194750 DOI: 10.1016/j.ceb.2023.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/11/2024]
Abstract
During embryogenesis, the mammalian kidney arises because of reciprocal interactions between the ureteric bud (UB) and the metanephric mesenchyme (MM), driving UB branching and nephron induction. These morphogenetic processes involve a series of cellular rearrangements that are tightly controlled by gene regulatory networks and signaling cascades. Here, we discuss how kidney developmental studies have informed the definition of procedures to obtain kidney organoids from human pluripotent stem cells (hPSCs). Moreover, bioengineering techniques have emerged as potential solutions to externally impose controlled microenvironments for organoid generation from hPSCs. Next, we summarize some of these advances with major focus On recent works merging hPSC-derived kidney organoids (hPSC-kidney organoids) with organ-on-chip to develop robust models for drug discovery and disease modeling applications. We foresee that, in the near future, coupling of different organoid models through bioengineering approaches will help advancing to recreate organ-to-organ crosstalk to increase our understanding on kidney disease progression in the human context and search for new therapeutics.
Collapse
Affiliation(s)
- Anisha Pahuja
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Daniel Moya-Rull
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; University of Barcelona, 08028 Barcelona, Spain.
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
25
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
26
|
Shakeri A, Wang Y, Zhao Y, Landau S, Perera K, Lee J, Radisic M. Engineering Organ-on-a-Chip Systems for Vascular Diseases. Arterioscler Thromb Vasc Biol 2023; 43:2241-2255. [PMID: 37823265 PMCID: PMC10842627 DOI: 10.1161/atvbaha.123.318233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Vascular diseases, such as atherosclerosis and thrombosis, are major causes of morbidity and mortality worldwide. Traditional in vitro models for studying vascular diseases have limitations, as they do not fully recapitulate the complexity of the in vivo microenvironment. Organ-on-a-chip systems have emerged as a promising approach for modeling vascular diseases by incorporating multiple cell types, mechanical and biochemical cues, and fluid flow in a microscale platform. This review provides an overview of recent advancements in engineering organ-on-a-chip systems for modeling vascular diseases, including the use of microfluidic channels, ECM (extracellular matrix) scaffolds, and patient-specific cells. We also discuss the limitations and future perspectives of organ-on-a-chip for modeling vascular diseases.
Collapse
Affiliation(s)
- Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Ying Wang
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Yimu Zhao
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Shira Landau
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Kevin Perera
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jonguk Lee
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- KITE - Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
27
|
Wang YT, Meng XT. A review of the evidence to support electrical stimulation -induced vascularization in engineered tissue. Regen Ther 2023; 24:237-244. [PMID: 37534238 PMCID: PMC10393514 DOI: 10.1016/j.reth.2023.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/25/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Abstract
Tissue engineering presents a promising solution for regenerative medicine and the success depends on the supply of oxygen/nutrients to the cells by rapid vascularization. More and more technologies are being developed to facilitate vascularization of engineered tissues. In this review, we indicated that a regulatory system which influences all angiogenesis associated cells to achieve their desired functional state is ideal for the construction of vascularized engineered tissues in vitro. We presented the evidence that electrical stimulation (ES) enhances the synergistic promotion of co-cultured angiogenesis associated cells and its potential regulatory mechanisms, highlighted the potential advantages of a combination of mesenchymal stem cells (MSCs), endothelial cells (ECs) and ES to achieve tissue vascularization, with particular emphasis on the different biological pathways of ES-regulated ECs. Finally, we proposed the future direction of using ES to reconstruct engineered tissue blood vessels, pointed out the potential advantages and disadvantages of ES application on tissue vascularization.
Collapse
Affiliation(s)
- Ying-tong Wang
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiao-ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
28
|
Okhovatian S, Shakeri A, Huyer LD, Radisic M. Elastomeric Polyesters in Cardiovascular Tissue Engineering and Organs-on-a-Chip. Biomacromolecules 2023; 24:4511-4531. [PMID: 37639715 PMCID: PMC10915885 DOI: 10.1021/acs.biomac.3c00387] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cardiovascular tissue constructs provide unique design requirements due to their functional responses to substrate mechanical properties and cyclic stretching behavior of cardiac tissue that requires the use of durable elastic materials. Given the diversity of polyester synthesis approaches, an opportunity exists to develop a new class of biocompatible, elastic, and immunomodulatory cardiovascular polymers. Furthermore, elastomeric polyester materials have the capability to provide tailored biomechanical synergy with native tissue and hence reduce inflammatory response in vivo and better support tissue maturation in vitro. In this review, we highlight underlying chemistry and design strategies of polyester elastomers optimized for cardiac tissue scaffolds. The major advantages of these materials such as their tunable elasticity, desirable biodegradation, and potential for incorporation of bioactive compounds are further expanded. Unique fabrication methods using polyester materials such as micromolding, 3D stamping, electrospinning, laser ablation, and 3D printing are discussed. Moreover, applications of these biomaterials in cardiovascular organ-on-a-chip devices and patches are analyzed. Finally, we outline unaddressed challenges in the field that need further study to enable the impactful translation of soft polyesters to clinical applications.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Locke Davenport Huyer
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
29
|
Coliaie P, Bhawnani RR, Ali R, Kelkar MS, Korde A, Langston M, Liu C, Nazemifard N, Patience DB, Rosenbaum T, Skliar D, Nere NK, Singh MR. Snap-on Adaptor for Microtiter Plates to Enable Continuous-Flow Microfluidic Screening and Harvesting of Crystalline Materials. ACS OMEGA 2023; 8:41502-41511. [PMID: 37969966 PMCID: PMC10633872 DOI: 10.1021/acsomega.3c05478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/05/2023] [Indexed: 11/17/2023]
Abstract
Microtiter plate assay is a conventional and standard tool for high-throughput (HT) screening that allows the synthesis, harvesting, and analysis of crystals. The microtiter plate screening assays require a small amount of solute in each experiment, which is adequate for a solid-state crystal analysis such as X-ray diffraction (XRD) or Raman spectroscopy. Despite the advantages of these high-throughput assays, their batch operational nature results in a continuous decrease in supersaturation due to crystal nucleation and growth. Continuous-flow microfluidic mixer devices have evolved as an alternate technique for efficiently screening crystals under controlled supersaturation. However, such a microfluidic device requires a minimum of two inlets per micromixer to create cyclonic flow, thereby creating physical limitations for implementing such a device for HT screening. Additionally, the monolithic design of these microfluidic devices makes it challenging to harvest crystals for post-screening analysis. Here, we develop a snap-on adapter that can be reversibly attached to a microtiter plate and convert it into a continuous-flow microfluidic mixer device. The integration of the snap-on adapter with a flow distributor and concentration gradient generator provides greater control over screening conditions while minimizing the number of independent inlets and pumps required. The three-dimensional (3D)-printed snap-on adaptor is plugged into a 24-well plate assay to demonstrate salt screening of naproxen crystals. Different naproxen salts are crystallized using four different salt formers (SFs)-sodium hydroxide, potassium hydroxide, pyridine, and arginine-and four different solvents-ethanol, methanol, isopropyl alcohol, and deionized water. The wells are further inspected under an optical microscope to identify their morphological forms and yields. The crystals are then harvested for solid-state characterization using XRD and Fourier transform infrared spectroscopy, followed by measurement of their dissolution rates. The flexibility of the snap-on adapter to fit on a wide range of microtiter plates and the ease in harvesting and analyzing crystals postscreening are two significant advantages that make this device versatile for various applications.
Collapse
Affiliation(s)
- Paria Coliaie
- Department
of Chemical Engineering, University of Illinois
at Chicago, Chicago, Illinois 60607, United States
| | - Rajan R. Bhawnani
- Department
of Chemical Engineering, University of Illinois
at Chicago, Chicago, Illinois 60607, United States
| | - Rabia Ali
- Department
of Chemical Engineering, University of Illinois
at Chicago, Chicago, Illinois 60607, United States
| | - Manish S. Kelkar
- Center
of Excellence for Isolation & Separation Technologies (CoExIST), Process R&D, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Akshay Korde
- Center
of Excellence for Isolation & Separation Technologies (CoExIST), Process R&D, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Marianne Langston
- Pharmaceutics
Research—Analytical Development, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts 02139, United States
| | - Chengxiang Liu
- Pharmaceutical
Development, Biogen, Cambridge, Massachusetts 02142, United States
| | - Neda Nazemifard
- Pharmaceutics
Research—Analytical Development, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts 02139, United States
| | - Daniel B. Patience
- Chemical
Process Development, Biogen, Cambridge, Massachusetts 02142, United States
| | - Tamar Rosenbaum
- Bristol-Myers
Squibb Co., Drug Product Science & Technology, New Brunswick, New Jersey 08901, United States
| | - Dimitri Skliar
- Bristol
Myers Squibb Co., Chemical & Synthetic Development, New Brunswick, New Jersey 08901, United States
| | - Nandkishor K. Nere
- Department
of Chemical Engineering, University of Illinois
at Chicago, Chicago, Illinois 60607, United States
- Center
of Excellence for Isolation & Separation Technologies (CoExIST), Process R&D, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Meenesh R. Singh
- Department
of Chemical Engineering, University of Illinois
at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
30
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
31
|
Lu RXZ, Rafatian N, Zhao Y, Wagner KT, Beroncal EL, Li B, Lee C, Chen J, Churcher E, Vosoughi D, Wang Y, Baker A, Trahtemberg U, Li B, Pierro A, Andreazza AC, Dos Santos CC, Radisic M. Heart-on-a-chip model of immune-induced cardiac dysfunction reveals the role of free mitochondrial DNA and therapeutic effects of endothelial exosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552495. [PMID: 37609237 PMCID: PMC10441383 DOI: 10.1101/2023.08.09.552495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Cardiovascular disease continues to take more human lives than all cancer combined, prompting the need for improved research models and treatment options. Despite a significant progress in development of mature heart-on-a-chip models of fibrosis and cardiomyopathies starting from induced pluripotent stem cells (iPSCs), human cell-based models of myocardial inflammation are lacking. Here, we bioengineered a vascularized heart-on-a-chip system with circulating immune cells to model SARS-CoV-2-induced acute myocarditis. Briefly, we observed hallmarks of COVID-19-induced myocardial inflammation in the heart-on-a-chip model, as the presence of immune cells augmented the expression levels of proinflammatory cytokines, triggered progressive impairment of contractile function and altered intracellular calcium transient activities. An elevation of circulating cell-free mitochondrial DNA (ccf-mtDNA) was measured first in the in vitro heart-on-a-chip model and then validated in COVID-19 patients with low left ventricular ejection fraction (LVEF), demonstrating that mitochondrial damage is an important pathophysiological hallmark of inflammation induced cardiac dysfunction. Leveraging this platform in the context of SARS-CoV-2 induced myocardial inflammation, we established that administration of human umbilical vein-derived EVs effectively rescued the contractile deficit, normalized intracellular calcium handling, elevated the contraction force and reduced the ccf- mtDNA and chemokine release via TLR-NF-kB signaling axis.
Collapse
|
32
|
Santos AC, Nader G, El Soufi El Sabbagh D, Urban K, Attisano L, Carlen PL. Treating Hyperexcitability in Human Cerebral Organoids Resulting from Oxygen-Glucose Deprivation. Cells 2023; 12:1949. [PMID: 37566028 PMCID: PMC10416870 DOI: 10.3390/cells12151949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Human cerebral organoids resemble the 3D complexity of the human brain and have the potential to augment current drug development pipelines for neurological disease. Epilepsy is a complex neurological condition characterized by recurrent seizures. A third of people with epilepsy do not respond to currently available pharmaceutical drugs, and there is not one drug that treats all subtypes; thus, better models of epilepsy are needed for drug development. Cerebral organoids may be used to address this unmet need. In the present work, human cerebral organoids are used along with electrophysiological methods to explore oxygen-glucose deprivation as a hyperexcitability agent. This activity is investigated in its response to current antiseizure drugs. Furthermore, the mechanism of action of the drug candidates is probed with qPCR and immunofluorescence. The findings demonstrate OGD-induced hyperexcitable changes in the cerebral organoid tissue, which is treated with cannabidiol and bumetanide. There is evidence for NKCC1 and KCC2 gene expression, as well as other genes and proteins involved in the complex development of GABAergic signaling. This study supports the use of organoids as a platform for modelling cerebral cortical hyperexcitability that could be extended to modelling epilepsy and used for drug discovery.
Collapse
Affiliation(s)
- Alexandra C. Santos
- Krembil Research Institute, University Health Network, Toronto, ON M5S 0T8, Canada (P.L.C.)
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - George Nader
- Krembil Research Institute, University Health Network, Toronto, ON M5S 0T8, Canada (P.L.C.)
| | - Dana El Soufi El Sabbagh
- Krembil Research Institute, University Health Network, Toronto, ON M5S 0T8, Canada (P.L.C.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
- Donnelly Centre, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Peter L. Carlen
- Krembil Research Institute, University Health Network, Toronto, ON M5S 0T8, Canada (P.L.C.)
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
33
|
Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. NATURE REVIEWS BIOENGINEERING 2023; 1:1-15. [PMID: 37359774 PMCID: PMC10173243 DOI: 10.1038/s44222-023-00063-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 06/20/2023]
Abstract
Biomedical research is undergoing a paradigm shift towards approaches centred on human disease models owing to the notoriously high failure rates of the current drug development process. Major drivers for this transition are the limitations of animal models, which, despite remaining the gold standard in basic and preclinical research, suffer from interspecies differences and poor prediction of human physiological and pathological conditions. To bridge this translational gap, bioengineered human disease models with high clinical mimicry are being developed. In this Review, we discuss preclinical and clinical studies that benefited from these models, focusing on organoids, bioengineered tissue models and organs-on-chips. Furthermore, we provide a high-level design framework to facilitate clinical translation and accelerate drug development using bioengineered human disease models.
Collapse
Affiliation(s)
- Anna Loewa
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - James J. Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
- Department of Mathematics, University of British Columbia, Vancouver, BC Canada
| | - Sarah Hedtrich
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Center of Biological Design, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC Canada
- Max-Delbrück Center for Molecular Medicine (MCD), Helmholtz Association, Berlin, Germany
| |
Collapse
|
34
|
Sunildutt N, Parihar P, Chethikkattuveli Salih AR, Lee SH, Choi KH. Revolutionizing drug development: harnessing the potential of organ-on-chip technology for disease modeling and drug discovery. Front Pharmacol 2023; 14:1139229. [PMID: 37180709 PMCID: PMC10166826 DOI: 10.3389/fphar.2023.1139229] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
The inefficiency of existing animal models to precisely predict human pharmacological effects is the root reason for drug development failure. Microphysiological system/organ-on-a-chip technology (organ-on-a-chip platform) is a microfluidic device cultured with human living cells under specific organ shear stress which can faithfully replicate human organ-body level pathophysiology. This emerging organ-on-chip platform can be a remarkable alternative for animal models with a broad range of purposes in drug testing and precision medicine. Here, we review the parameters employed in using organ on chip platform as a plot mimic diseases, genetic disorders, drug toxicity effects in different organs, biomarker identification, and drug discoveries. Additionally, we address the current challenges of the organ-on-chip platform that should be overcome to be accepted by drug regulatory agencies and pharmaceutical industries. Moreover, we highlight the future direction of the organ-on-chip platform parameters for enhancing and accelerating drug discoveries and personalized medicine.
Collapse
Affiliation(s)
- Naina Sunildutt
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | - Pratibha Parihar
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | | | - Sang Ho Lee
- College of Pharmacy, Jeju National University, Jeju, Republic of Korea
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
35
|
Cao C, Lu X, Guo X, Zhao H, Gao Y. Patient-derived models: Promising tools for accelerating the clinical translation of breast cancer research findings. Exp Cell Res 2023; 425:113538. [PMID: 36871856 DOI: 10.1016/j.yexcr.2023.113538] [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: 12/12/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Breast cancer has become the highest incidence of cancer in women. It was extensively and deeply studied by biologists and medical workers worldwide. However, the meaningful results in lab researches cannot be realized in clinical, and a part of new drugs in clinical experiments do not obtain as good results as the preclinical researches. It is urgently that promote a kind of breast cancer research models that can get study results closer to the physiological condition of the human body. Patient-derived models (PDMs) originating from clinical tumor, contain primary elements of tumor and maintain key clinical features of tumor. So they are promising research models to facilitate laboratory researches translate to clinical application, and predict the treatment outcome of patients. In this review, we summarize the establishment of PDMs of breast cancer, reviewed the application of PDMs in clinical translational researches and personalized precision medicine with breast cancer as an example, to improve the understanding of PDMs among researchers and clinician, facilitate them to use PDMs on a large scale of breast cancer researches and promote the clinical translation of laboratory research and new drug development.
Collapse
Affiliation(s)
- Changqing Cao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China; State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Xiyan Lu
- Department of Outpatient, The Second Affiliated Hospital of Air Force Medical University, China
| | - Xinyan Guo
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Huadong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China.
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China.
| |
Collapse
|
36
|
Melzer MK, Resheq Y, Navaee F, Kleger A. The application of pancreatic cancer organoids for novel drug discovery. Expert Opin Drug Discov 2023; 18:429-444. [PMID: 36945198 DOI: 10.1080/17460441.2023.2194627] [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: 03/23/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma presents with a dismal prognosis. Personalized therapy is urgently warranted to overcome the treatment limitations of the "one-size-fits-all" scheme. Organoids have emerged as fundamental novel tools to study tumor biology and heterogeneity, hence overcoming limitations of other model systems by better-reflecting tissue heterogeneity and recapitulating in-vivo processes. Besides their crucial role in basic research, they have evolved as tools for translational drug discovery and patient stratification. AREAS COVERED This review highlights the achievements of an organoid-based drug investigation and discovery. The authors present an overview of studies using organoids for drug testing. Further, they pinpoint studies correlating the in vitro prediction of organoids to the actual patient`s response. Furthermore, the authors describe novel model systems and take a thorough overlook of microfluidic chips, synthetic matrices, multicellular systems, bioprinting, and stem cell-derived pancreatic organoid systems. EXPERT OPINION Organoid systems promise great potential for future clinical applications. Indeed, they may be implemented into informed decision-making for guiding therapies. However, validation by randomized trials is mandatory. Additionally, organoids in combination with other cellular compartments may be exploited for drug discovery by studying niche-tumor interaction. Yet, several precautions must be kept in mind, such as standardization and reproducibility.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Yazid Resheq
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Fatemeh Navaee
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
- Core Facility Organoids, Ulm University, Ulm, Germany
| |
Collapse
|
37
|
Zhao Y, Wang EY, Lai FBL, Cheung K, Radisic M. Organs-on-a-chip: a union of tissue engineering and microfabrication. Trends Biotechnol 2023; 41:410-424. [PMID: 36725464 PMCID: PMC9985977 DOI: 10.1016/j.tibtech.2022.12.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 02/03/2023]
Abstract
We review the emergence of the new field of organ-on-a-chip (OOAC) engineering, from the parent fields of tissue engineering and microfluidics. We place into perspective the tools and capabilities brought into the OOAC field by early tissue engineers and microfluidics experts. Liver-on-a-chip and heart-on-a-chip are used as two case studies of systems that heavily relied on tissue engineering techniques and that were amongst the first OOAC models to be implemented, motivated by the need to better assess toxicity to human tissues in preclinical drug development. We review current challenges in OOAC that often stem from the same challenges in the parent fields, such as stable vascularization and drug absorption.
Collapse
Affiliation(s)
- Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fook B L Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Krisco Cheung
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.
| |
Collapse
|
38
|
Soto Veliz D, Lin K, Sahlgren C. Organ-on-a-chip technologies for biomedical research and drug development: A focus on the vasculature. SMART MEDICINE 2023; 2:e20220030. [PMID: 37089706 PMCID: PMC7614466 DOI: 10.1002/smmd.20220030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/20/2023] [Indexed: 04/25/2023]
Abstract
Current biomedical models fail to replicate the complexity of human biology. Consequently, almost 90% of drug candidates fail during clinical trials after decades of research and billions of investments in drug development. Despite their physiological similarities, animal models often misrepresent human responses, and instead, trigger ethical and societal debates regarding their use. The overall aim across regulatory entities worldwide is to replace, reduce, and refine the use of animal experimentation, a concept known as the Three Rs principle. In response, researchers develop experimental alternatives to improve the biological relevance of in vitro models through interdisciplinary approaches. This article highlights the emerging organ-on-a-chip technologies, also known as microphysiological systems, with a focus on models of the vasculature. The cardiovascular system transports all necessary substances, including drugs, throughout the body while in charge of thermal regulation and communication between other organ systems. In addition, we discuss the benefits, limitations, and challenges in the widespread use of new biomedical models. Coupled with patient-derived induced pluripotent stem cells, organ-on-a-chip technologies are the future of drug discovery, development, and personalized medicine.
Collapse
Affiliation(s)
- Diosangeles Soto Veliz
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Kai‐Lan Lin
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Cecilia Sahlgren
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
39
|
Yu C, Kang R, Tang D. Organoids Models of Pancreatic Duct Adenocarcinoma. Methods Mol Biol 2023; 2712:45-60. [PMID: 37578695 DOI: 10.1007/978-1-0716-3433-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Three-dimensional (3D) organoid culture is a laboratory technique used to grow and study miniature organs that mimic the structure and function of real organs in the human body. Organoids are created from stem cells or tissue samples and are grown in a 3D matrix that allows them to self-organize into a complex, three-dimensional structure. Organoids are valuable tools for studying human biology and disease, including cancer. Pancreatic ductal adenocarcinoma (PDAC) still has the worst survival rate of common malignancies, despite recent advances in cancer treatment. Preclinical studies have shown that impaired cell death pathways, including apoptosis, necroptosis, ferroptosis, pyroptosis, and alkaliptosis, promote PDAC development. Organoid models are now widely used in the study of pancreatic cancer biology, including cell death machinery. This chapter provides step-by-step protocols for generating human or mice PDAC organoids in a 3D Matrigel system.
Collapse
Affiliation(s)
- Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
40
|
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: 43] [Impact Index Per Article: 14.3] [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
|
41
|
Liu C, Campbell SB, Li J, Bannerman D, Pascual-Gil S, Kieda J, Wu Q, Herman PR, Radisic M. High Throughput Omnidirectional Printing of Tubular Microstructures from Elastomeric Polymers. Adv Healthc Mater 2022; 11:e2201346. [PMID: 36165232 PMCID: PMC9742311 DOI: 10.1002/adhm.202201346] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/09/2022] [Indexed: 01/28/2023]
Abstract
Bioelastomers are extensively used in biomedical applications due to their desirable mechanical strength, tunable properties, and chemical versatility; however, three-dimensional (3D) printing bioelastomers into microscale structures has proven elusive. Herein, a high throughput omnidirectional printing approach via coaxial extrusion is described that fabricates perfusable elastomeric microtubes of unprecedently small inner diameter (350-550 µm) and wall thickness (40-60 µm). The versatility of this approach is shown through the printing of two different polymeric elastomers, followed by photocrosslinking and removal of the fugitive inner phase. Designed experiments are used to tune the microtube dimensions and stiffness to match that of native ex vivo rat vasculature. This approach affords the fabrication of multiple biomimetic shapes resembling cochlea and kidney glomerulus and affords facile, high-throughput generation of perfusable structures that can be seeded with endothelial cells for biomedical applications. Post-printing laser micromachining is performed to generate micro-sized holes (520 µm) in the tube wall to tune microstructure permeability. Importantly, for organ-on-a-chip applications, the described approach takes only 3.6 min to print microtubes (without microholes) over an entire 96-well plate device, in contrast to comparable hole-free structures that take between 1.5 and 6.5 days to fabricate using a manual 3D stamping approach.
Collapse
Affiliation(s)
- Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Scott B. Campbell
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jianzhao Li
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Dawn Bannerman
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Simon Pascual-Gil
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Peter R. Herman
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Caballero D, Reis RL, Kundu SC. Boosting the Clinical Translation of Organ-on-a-Chip Technology. Bioengineering (Basel) 2022; 9:549. [PMID: 36290517 PMCID: PMC9598310 DOI: 10.3390/bioengineering9100549] [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: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
Organ-on-a-chip devices have become a viable option for investigating critical physiological events and responses; this technology has matured substantially, and many systems have been reported for disease modeling or drug screening over the last decade. Despite the wide acceptance in the academic community, their adoption by clinical end-users is still a non-accomplished promise. The reasons behind this difficulty can be very diverse but most likely are related to the lack of predictive power, physiological relevance, and reliability necessary for being utilized in the clinical area. In this Perspective, we briefly discuss the main attributes of organ-on-a-chip platforms in academia and how these characteristics impede their easy translation to the clinic. We also discuss how academia, in conjunction with the industry, can contribute to boosting their adoption by proposing novel design concepts, fabrication methods, processes, and manufacturing materials, improving their standardization and versatility, and simplifying their manipulation and reusability.
Collapse
Affiliation(s)
- David Caballero
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| | - Subhas C. Kundu
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| |
Collapse
|
43
|
Terrassoux L, Claux H, Bacari S, Meignan S, Furlan A. A Bloody Conspiracy. Blood Vessels and Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194581. [PMID: 36230504 PMCID: PMC9558972 DOI: 10.3390/cancers14194581] [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: 08/18/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The tumor microenvironment has risen over the last years as a significant contributor to the failure of antitumoral strategies due to its numerous pro-tumorigenic activities. In this review, we focused on two features of this microenvironment, namely angiogenesis and immunity, which have been the targets of therapies to tackle tumors via its microenvironmental part over the last decade. Increasing our knowledge of the complex interactions within this ecosystem is mandatory to optimize these therapeutic approaches. The development of innovative experimental models is of great help in reaching this goal. Abstract Cancer progression occurs in concomitance with a profound remodeling of the cellular microenvironment. Far from being a mere passive event, the re-orchestration of interactions between the various cell types surrounding tumors highly contributes to the progression of the latter. Tumors notably recruit and stimulate the sprouting of new blood vessels through a process called neo-angiogenesis. Beyond helping the tumor cope with an increased metabolic demand associated with rapid growth, this also controls the metastatic dissemination of cancer cells and the infiltration of immune cells in the tumor microenvironment. To decipher this critical interplay for the clinical progression of tumors, the research community has developed several valuable models in the last decades. This review offers an overview of the various instrumental solutions currently available, including microfluidic chips, co-culture models, and the recent rise of organoids. We highlight the advantages of each technique and the specific questions they can address to better understand the tumor immuno-angiogenic ecosystem. Finally, we discuss this development field’s fundamental and applied perspectives.
Collapse
Affiliation(s)
- Lisa Terrassoux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Hugo Claux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Salimata Bacari
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Samuel Meignan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Alessandro Furlan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
44
|
Ma X, Li H, Zhu S, Hong Z, Kong W, Yuan Q, Wu R, Pan Z, Zhang J, Chen Y, Wang X, Wang K. Angiorganoid: vitalizing the organoid with blood vessels. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R44-R57. [PMID: 35994010 PMCID: PMC9513648 DOI: 10.1530/vb-22-0001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022]
Abstract
The emergence of the organoid simulates the native organs and this mini organ offers an excellent platform for probing multicellular interaction, disease modeling and drug discovery. Blood vessels constitute the instructive vascular niche which is indispensable for organ development, function and regeneration. Therefore, it is expected that the introduction of infiltrated blood vessels into the organoid might further pump vitality and credibility into the system. While the field is emerging and growing with new concepts and methodologies, this review aims at presenting various sources of vascular ingredients for constructing vascularized organoids and the paired methodology including de- and recellularization, bioprinting and microfluidics. Representative vascular organoids corresponding to specific tissues are also summarized and discussed to elaborate on the next generation of organoid development.
Collapse
Affiliation(s)
- Xiaojing Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Hongfei Li
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Shuntian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zixuan Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Weijing Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Qihang Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Runlong Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York, USA
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
45
|
Günther C, Winner B, Neurath MF, Stappenbeck TS. Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut 2022; 71:1892-1908. [PMID: 35636923 PMCID: PMC9380493 DOI: 10.1136/gutjnl-2021-326560] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
We are entering an era of medicine where increasingly sophisticated data will be obtained from patients to determine proper diagnosis, predict outcomes and direct therapies. We predict that the most valuable data will be produced by systems that are highly dynamic in both time and space. Three-dimensional (3D) organoids are poised to be such a highly valuable system for a variety of gastrointestinal (GI) diseases. In the lab, organoids have emerged as powerful systems to model molecular and cellular processes orchestrating natural and pathophysiological human tissue formation in remarkable detail. Preclinical studies have impressively demonstrated that these organs-in-a-dish can be used to model immunological, neoplastic, metabolic or infectious GI disorders by taking advantage of patient-derived material. Technological breakthroughs now allow to study cellular communication and molecular mechanisms of interorgan cross-talk in health and disease including communication along for example, the gut-brain axis or gut-liver axis. Despite considerable success in culturing classical 3D organoids from various parts of the GI tract, some challenges remain to develop these systems to best help patients. Novel platforms such as organ-on-a-chip, engineered biomimetic systems including engineered organoids, micromanufacturing, bioprinting and enhanced rigour and reproducibility will open improved avenues for tissue engineering, as well as regenerative and personalised medicine. This review will highlight some of the established methods and also some exciting novel perspectives on organoids in the fields of gastroenterology. At present, this field is poised to move forward and impact many currently intractable GI diseases in the form of novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Stem Cell Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Zhu S, Abudupataer M, Yan S, Wang C, Wang L, Zhu K. Construction of a high-throughput aorta smooth muscle-on-a-chip for thoracic aortic aneurysm drug screening. Biosens Bioelectron 2022; 218:114747. [DOI: 10.1016/j.bios.2022.114747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/02/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
|
47
|
Huang X, Wang P, Liu J, Xu F, Liu C, Xu Z, Hou Z, Ye F. Patterning High-Resolution Microstructures on Thermoplastics by Ceramic Nanoparticles Filled Epoxy Coated Molds for Duplicating Nature-Derived Functional Surfaces. ACS APPLIED MATERIALS & INTERFACES 2022; 14:28270-28279. [PMID: 35680478 DOI: 10.1021/acsami.2c04277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Patterning high-resolution microstructures on thermoplastic substrates is of fundamental importance for the commercialization of microfluidics, advanced functional surfaces, and optical elements. Though many methods are developed to fabricate micropatterned plastic devices with 100 μm resolution, they suffer substantially higher cost or lower productivity when the resolution of the micropatterns is to be further improved. Here, we develop low-cost molds consisting of thin ceramic-filled-epoxy composite coatings on steel substrates. By virtue of the loaded ZrO2 nanoparticle fillers, the enhanced mechanical and thermal properties of the composite molds enable the epoxy microstructures to survive harsh conditions in conventional thermoplastic processing methods including hot embossing, imprinting, and mold injection. With the ceramic-filled-epoxy coated molds, we are able to improve the fabrication resolution of microstructures on plastics to 10 μm with unprecedented low-cost and excellent durability.
Collapse
Affiliation(s)
- Xing Huang
- School of Engineering, Zhejiang University City College, Hangzhou, 310015, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, China
| | - Pengfei Wang
- School of Engineering, Zhejiang University City College, Hangzhou, 310015, China
| | - Junfeng Liu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, China
| | - Fangmin Xu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, China
| | - Cong Liu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhongbin Xu
- School of Engineering, Zhejiang University City College, Hangzhou, 310015, China
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, China
- Ningbo Research Institute and Institute of Robotics, Zhejiang University, Ningbo 315100, China
| | - Zhanglin Hou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Fangfu Ye
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| |
Collapse
|
48
|
Rajasekar S, Lin DSY, Zhang F, Sotra A, Boshart A, Clotet-Freixas S, Liu A, Hirota JA, Ogawa S, Konvalinka A, Zhang B. Subtractive manufacturing with swelling induced stochastic folding of sacrificial materials for fabricating complex perfusable tissues in multi-well plates. LAB ON A CHIP 2022; 22:1929-1942. [PMID: 35383790 DOI: 10.1039/d1lc01141c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Organ-on-a-chip systems that recapitulate tissue-level functions have been proposed to improve in vitro-in vivo correlation in drug development. Significant progress has been made to control the cellular microenvironment with mechanical stimulation and fluid flow. However, it has been challenging to introduce complex 3D tissue structures due to the physical constraints of microfluidic channels or membranes in organ-on-a-chip systems. Inspired by 4D bioprinting, we develop a subtractive manufacturing technique where a flexible sacrificial material can be patterned on a 2D surface, swell and shape change when exposed to aqueous hydrogel, and subsequently degrade to produce perfusable networks in a natural hydrogel matrix that can be populated with cells. The technique is applied to fabricate organ-specific vascular networks, vascularized kidney proximal tubules, and terminal lung alveoli in a customized 384-well plate and then further scaled to a 24-well plate format to make a large vascular network, vascularized liver tissues, and for integration with ultrasound imaging. This biofabrication method eliminates the physical constraints in organ-on-a-chip systems to incorporate complex ready-to-perfuse tissue structures in an open-well design.
Collapse
Affiliation(s)
- Shravanthi Rajasekar
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
| | - Dawn S Y Lin
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
| | - Feng Zhang
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Alexander Sotra
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Alex Boshart
- Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Renal Transplant Program, Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Sergi Clotet-Freixas
- Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Renal Transplant Program, Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Amy Liu
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Jeremy A Hirota
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
- Department of Medicine, Division of Respirology, McMaster University, 1200 Main St W, Hamilton, ON, L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
| | - Shinichiro Ogawa
- McEwen Stem Cell Institute, University Health Network, MaRS Center, 101 College St, Toronto, Ontario, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, MaRS Center, 101 College St, Toronto, Ontario, M5G 1L7 Canada
- Liver Transplant Program, Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Ana Konvalinka
- Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Renal Transplant Program, Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, MaRS Center, 101 College St, Toronto, Ontario, M5G 1L7 Canada
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| |
Collapse
|
49
|
Shang L, Ye F, Li M, Zhao Y. Spatial confinement toward creating artificial living systems. Chem Soc Rev 2022; 51:4075-4093. [PMID: 35502858 DOI: 10.1039/d1cs01025e] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lifeforms are regulated by many physicochemical factors, and these factors could be controlled to play a role in the construction of artificial living systems. Among these factors, spatial confinement is an important one, which mediates biological behaviors at multiscale levels and participates in the biomanufacturing processes accordingly. This review describes how spatial confinement, as a fundamental biological phenomenon, provides cues for the construction of artificial living systems. Current knowledge about the role of spatial confinement in mediating individual cell behavior, collective cellular behavior, and tissue-level behavior are categorized. Endeavors on the synthesis of biomacromolecules, artificial cells, engineered tissues, and organoids in spatially confined bioreactors are then emphasized. After that, we discuss the cutting-edge applications of spatially confined artificial living systems in biomedical fields. Finally, we conclude by assessing the remaining challenges and future trends in the context of fundamental science, technical improvement, and practical applications.
Collapse
Affiliation(s)
- Luoran Shang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China. .,Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China. .,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| | - Ming Li
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China. .,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| |
Collapse
|
50
|
Ronaldson-Bouchard K, Baldassarri I, Tavakol DN, Graney PL, Samaritano M, Cimetta E, Vunjak-Novakovic G. Engineering complexity in human tissue models of cancer. Adv Drug Deliv Rev 2022; 184:114181. [PMID: 35278521 PMCID: PMC9035134 DOI: 10.1016/j.addr.2022.114181] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
Major progress in the understanding and treatment of cancer have tremendously improved our knowledge of this complex disease and improved the length and quality of patients' lives. Still, major challenges remain, in particular with respect to cancer metastasis which still escapes effective treatment and remains responsible for 90% of cancer related deaths. In recent years, the advances in cancer cell biology, oncology and tissue engineering converged into the engineered human tissue models of cancer that are increasingly recapitulating many aspects of cancer progression and response to drugs, in a patient-specific context. The complexity and biological fidelity of these models, as well as the specific questions they aim to investigate, vary in a very broad range. When selecting and designing these experimental models, the fundamental question is "how simple is complex enough" to accomplish a specific goal of cancer research. Here we review the state of the art in developing and using the human tissue models in cancer research and developmental drug screening. We describe the main classes of models providing different levels of biological fidelity and complexity, discuss their advantages and limitations, and propose a framework for designing an appropriate model for a given study. We close by outlining some of the current needs, opportunities and challenges in this rapidly evolving field.
Collapse
Affiliation(s)
- Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Maria Samaritano
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Elisa Cimetta
- Department of Industrial Engineering, University of Padua, Via Marzolo 9, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; College of Dental Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA.
| |
Collapse
|