1
|
Ni B, Ye L, Zhang Y, Hu S, Lei W. Advances in humanoid organoid-based research on inter-organ communications during cardiac organogenesis and cardiovascular diseases. J Transl Med 2025; 23:380. [PMID: 40156006 PMCID: PMC11951738 DOI: 10.1186/s12967-025-06381-x] [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: 11/04/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
The intimate correlation between cardiovascular diseases and other organ pathologies, such as metabolic and kidney diseases, underscores the intricate interactions among these organs. Understanding inter-organ communications is crucial for developing more precise drugs and effective treatments for systemic diseases. While animal models have traditionally been pivotal in studying these interactions, human-induced pluripotent stem cells (hiPSCs) offer distinct advantages when constructing in vitro models. Beyond the conventional two-dimensional co-culture model, hiPSC-derived humanoid organoids have emerged as a substantial advancement, capable of replicating essential structural and functional attributes of internal organs in vitro. This breakthrough has spurred the development of multilineage organoids, assembloids, and organoids-on-a-chip technologies, which allow for enhanced physiological relevance. These technologies have shown great potential for mimicking coordinated organogenesis, exploring disease pathogenesis, and facilitating drug discovery. As the central organ of the cardiovascular system, the heart serves as the focal point of an extensively studied network of interactions. This review focuses on the advancements and challenges of hiPSC-derived humanoid organoids in studying interactions between the heart and other organs, presenting a comprehensive exploration of this cutting-edge approach in systemic disease research.
Collapse
Affiliation(s)
- Baoqiang Ni
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Lingqun Ye
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yan Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Wei Lei
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
2
|
Zhang L, Zhao J, Su C, Wu J, Jiang L, Chi H, Wang Q. Organoid models of ovarian cancer: resolving immune mechanisms of metabolic reprogramming and drug resistance. Front Immunol 2025; 16:1573686. [PMID: 40191206 PMCID: PMC11968360 DOI: 10.3389/fimmu.2025.1573686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Metabolic reprogramming is a hallmark of ovarian cancer, enabling tumor progression, immune evasion and drug resistance. The tumor microenvironment (TME) further shapes metabolic adaptations, enabling cancer cells to withstand hypoxia and nutrient deprivation. While organoid models provide a physiologically relevant platform for studying these processes, they still lack immune and vascular components, limiting their ability to fully recapitulate tumor metabolism and drug responses. In this study, we investigated the key metabolic mechanisms involved in ovarian cancer progression, focusing on glycolysis, lipid metabolism and amino acid metabolism. We integrated metabolomic analyses and drug sensitivity assays to explore metabolic-TME interactions using patient-derived, adult stem cell-derived and iPSC-derived organ tissues. Among these, we found that glycolysis, lipid metabolism and amino acid metabolism play a central role in tumor progression and chemotherapy resistance. We identified methylglyoxal (MGO)-mediated BRCA2 dysfunction as a driver of immune escape, a role for sphingolipid signaling in tumor proliferation and a role for kynurenine metabolism in CD8+ T cell suppression. In addition, PI3K/AKT/mTOR and Wnt/β-catenin pathways promote chemoresistance through metabolic adaptation. By elucidating the link between metabolic reprogramming and immune evasion, this study identifies key metabolic vulnerabilities and potential drug targets in ovarian cancer. Our findings support the development of metabolically targeted therapies and increase the utility of organoid-based precision medicine models.
Collapse
Affiliation(s)
- Lanyue Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jiangnan Zhao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Chunyu Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jianxi Wu
- Department of Preventive Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
3
|
She W, Shen C, Xue Z, Zhang B, Zhang G, Meng Q. Hydrogel Strain Sensors for Integrating Into Dynamic Organ-on-a-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407704. [PMID: 39846814 DOI: 10.1002/smll.202407704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Current hydrogel strain sensors have never been integrated into dynamic organ-on-a-chip (OOC) due to the lack of sensitivity in aqueous cell culture systems. To enhance sensing performance, a novel strain sensor is presented in which the MXene layer is coated on the bottom surface of a pre-stretched anti-swelling hydrogel substrate of di-acrylated Pluronic F127 (F127-DA) and chitosan (CS) for isolation from the cell culture on the top surface. The fabricated strain sensors display high sensitivity (gauge factor of 290.96), a wide sensing range (0-100%), and high repeatability. To demonstrate its application, alveolar epithelial cells are cultivated on the top surface of the hydrogel strain sensor forming alveolar barriers, and then integrated into dynamic lung-on-a-chip (LOC) systems. This system can sensitively monitor normal physiological breathing, pathological inflammation stimulated by lipopolysaccharide (LPS), and alleviated inflammation through drug intervention.
Collapse
Affiliation(s)
- Wenqi She
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chong Shen
- Center for Membrane and Water Science and Technology, Institute of Oceanic and Environmental Chemical Engineering, State Key Lab Base of Green Chemical Synthesis Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zaifei Xue
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Bin Zhang
- Department of Respiratory Disease, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 120070, China
| | - Guoliang Zhang
- Center for Membrane and Water Science and Technology, Institute of Oceanic and Environmental Chemical Engineering, State Key Lab Base of Green Chemical Synthesis Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qin Meng
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
4
|
Wang C, Liu S, Li C, Wang Z, Ming R, Huang L. Monitoring the Cascade of Monocyte-Derived Macrophages to Influenza Virus Infection in Human Alveolus Chips. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60045-60055. [PMID: 39450775 DOI: 10.1021/acsami.4c15125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Respiratory viruses ravage the world and seriously threaten people's health. Despite intense research efforts, the immune mechanism underlying respiratory virus-induced acute lung injury (ALI) and pulmonary fibrosis (PF) has not been fully elucidated. Here, the cascade of monocyte-derived macrophages to influenza A virus infection is monitored on an optimized human alveolus chip to reveal the role of macrophages in the development of ALI and PF. We find that viral infection causes damage to the alveolar air-liquid barrier and the release of inflammatory cytokines, which induce the M0 macrophages to gather and polarize to the M1 phenotype at the damaged site through recruitment, adhesion, migration, and activation, leading to ALI. Afterward, M1 macrophages polarize into the M2 phenotype, and then transform into myofibroblasts, followed by enhanced secretion of various anti-inflammatory cytokines and profibrotic cytokines, to promote PF. Our study provides an insight into the pathogenesis of virus-induced ALI and PF, which will assist in the development of therapeutic strategies and drugs for treating influenza and other respiratory virus infections.
Collapse
Affiliation(s)
- Chenguang Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Shujun Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chuyu Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhongjie Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Ruiqi Ming
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lili Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
- Tangshan Research Institute, Beijing Institute of Technology, Tangshan 063000, P. R. China
| |
Collapse
|
5
|
Wan Y, Ding J, Jia Z, Hong Y, Tian G, Zheng S, Pan P, Wang J, Liang H. Current trends and research topics regarding organoids: A bibliometric analysis of global research from 2000 to 2023. Heliyon 2024; 10:e32965. [PMID: 39022082 PMCID: PMC11253259 DOI: 10.1016/j.heliyon.2024.e32965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
The use of animal models for biological experiments is no longer sufficient for research related to human life and disease. The development of organ tissues has replaced animal models by mimicking the structure, function, development and homeostasis of natural organs. This provides more opportunities to study human diseases such as cancer, infectious diseases and genetic disorders. In this study, bibliometric methods were used to analyze organoid-related articles published over the last 20+ years to identify emerging trends and frontiers in organoid research. A total of 13,143 articles from 4125 institutions in 86 countries or regions were included in the analysis. The number of papers increased steadily over the 20-year period. The United States was the leading country in terms of number of papers and citations. Harvard Medical School had the highest number of papers published. Keyword analysis revealed research trends and focus areas such as organ tissues, stem cells, 3D culture and tissue engineering. In conclusion, this study used bibliometric and visualization methods to explore the field of organoid research and found that organ tissues are receiving increasing attention in areas such as cancer, drug discovery, personalized medicine, genetic disease modelling and gene repair, making them a current research hotspot and a future research trend.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianan Ding
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Zixuan Jia
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guijie Tian
- School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou, China
| | - Shuqian Zheng
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Pinfei Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jieyan Wang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| | - Hui Liang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| |
Collapse
|
6
|
Yan J, Monlong J, Cougoule C, Lacroix-Lamandé S, Wiedemann A. Mapping the scientific output of organoids for animal and human modeling infectious diseases: a bibliometric assessment. Vet Res 2024; 55:81. [PMID: 38926765 PMCID: PMC11210181 DOI: 10.1186/s13567-024-01333-7] [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: 12/01/2023] [Accepted: 04/11/2024] [Indexed: 06/28/2024] Open
Abstract
The escalation of antibiotic resistance, pandemics, and nosocomial infections underscores the importance of research in both animal and human infectious diseases. Recent advancements in three-dimensional tissue cultures, or "organoids", have revolutionized the development of in vitro models for infectious diseases. Our study conducts a bibliometric analysis on the use of organoids in modeling infectious diseases, offering an in-depth overview of this field's current landscape. We examined scientific contributions from 2009 onward that focused on organoids in host‒pathogen interactions using the Web of Science Core Collection and OpenAlex database. Our analysis included temporal trends, reference aging, author, and institutional productivity, collaborative networks, citation metrics, keyword cluster dynamics, and disruptiveness of organoid models. VOSviewer, CiteSpace, and Python facilitated this analytical assessment. The findings reveal significant growth and advancements in organoid-based infectious disease research. Analysis of keywords and impactful publications identified three distinct developmental phases in this area that were significantly influenced by outbreaks of Zika and SARS-CoV-2 viruses. The research also highlights the synergistic efforts between academia and publishers in tackling global pandemic challenges. Through mostly consolidating research efforts, organoids are proving to be a promising tool in infectious disease research for both human and animal infectious disease. Their integration into the field necessitates methodological refinements for better physiological emulation and the establishment of extensive organoid biobanks. These improvements are crucial for fully harnessing the potential of organoids in understanding infectious diseases and advancing the development of targeted treatments and vaccines.
Collapse
Affiliation(s)
- Jin Yan
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China.
- Research Center of Digestive Disease, Central South University, Changsha, China.
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.
| | - Jean Monlong
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Agnès Wiedemann
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.
| |
Collapse
|
7
|
Lee S, Rim YA, Kim J, Lee SH, Park HJ, Kim H, Ahn SJ, Ju JH. Guidelines for Manufacturing and Application of Organoids: Skin. Int J Stem Cells 2024; 17:182-193. [PMID: 38783680 PMCID: PMC11170114 DOI: 10.15283/ijsc24045] [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: 04/08/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
To address the limitations of animal testing, scientific research is increasingly focused on developing alternative testing methods. These alternative tests utilize cells or tissues derived from animals or humans for in vitro testing, as well as artificial tissues and organoids. In western countries, animal testing for cosmetics has been banned, leading to the adoption of artificial skin for toxicity evaluation, such as skin corrosion and irritation assessments. Standard guidelines for skin organoid technology becomes necessary to ensure consistent data and evaluation in replacing animal testing with in vitro methods. These guidelines encompass aspects such as cell sourcing, culture techniques, quality requirements and assessment, storage and preservation, and organoid-based assays.
Collapse
Affiliation(s)
- Seunghee Lee
- Organoid Standards Initiative
- Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Yeri Alice Rim
- Catholic Induced Pluripotent Stem Cell Research Center (CiSTEM), Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Su Hyon Lee
- Organoid Standards Initiative
- Biosolution Co., Ltd., Seoul, Korea
| | - Hye Jung Park
- Organoid Standards Initiative
- CellinCells, Seoul National University Dental Hospital, Seoul, Korea
| | - Hyounwoo Kim
- CellinCells, Seoul National University Dental Hospital, Seoul, Korea
| | - Sun-Ju Ahn
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Ji Hyeon Ju
- Organoid Standards Initiative
- Catholic Induced Pluripotent Stem Cell Research Center (CiSTEM), Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- YiPSCELL Inc., Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
8
|
Brignoli T, Ferrara S, Bertoni G. Emerging In Vitro Models for the Study of Infection and Pathogenesis of Pseudomonas aeruginosa and Testing of Antibacterial Agents. Methods Mol Biol 2024; 2721:233-239. [PMID: 37819526 DOI: 10.1007/978-1-0716-3473-8_16] [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: 10/13/2023]
Abstract
Several animal models have been developed to study infection strategies and pathogenesis of Pseudomonas aeruginosa. Some of these models are also used in preclinical and clinical research. However, these models are increasingly showing their limitations, including in recapitulating human diseases such as cystic fibrosis, which is strongly linked to P. aeruginosa infection. The emerging field of human organoids and organs-on-a-chip is expected to provide answers to the need for in vitro modeling of human diseases. Here, we describe the first recent efforts that will hopefully provide the basis for the development of advanced in vitro models for the study of P. aeruginosa infection and pathogenesis and the testing of antibacterial agents.
Collapse
Affiliation(s)
- Tarcisio Brignoli
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Silvia Ferrara
- Department of Biosciences, Università degli Studi di Milano, Milan, Milano, Italy
| | - Giovanni Bertoni
- Department of Biosciences, Università degli Studi di Milano, Milan, Milano, Italy.
| |
Collapse
|
9
|
She W, Shen C, Ying Y, Meng Q. Fabrication of sac-like hydrogel membranes for replicating curved tissue barriers on chips. LAB ON A CHIP 2023; 24:85-96. [PMID: 38018218 DOI: 10.1039/d3lc00807j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Current organ-on-a-chip (OOC) systems cannot mimic in vivo tissue barriers that feature curved geometries and rhythmic movement. This is due to the lack of a relevant membrane that can reproduce the natural biochemical and physical properties of a basement membrane, especially the characteristic sac-like structure possessed by multiple tissue barriers. To address this challenge, a sac-like hydrogel membrane is fabricated here using a one-step simple methodology inspired by soap bubble formation. Di-acrylated Pluronic® F127 (F127-DA) is a hydrogel that exhibits excellent mechanical properties, stably withstanding rhythmic mechanical stretching and fluid flow for at least 24 h. Using this hydrogel to make a membrane, a complex lung-on-a-chip device is successfully constructed, effectively replicating the alveolar-capillary barrier and demonstrating cellular function under physiological respiratory conditions. This membrane offers a crucial platform for replicating sac-like tissue barriers.
Collapse
Affiliation(s)
- Wenqi She
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| | - Chong Shen
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| | - Yinghua Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Qin Meng
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| |
Collapse
|
10
|
Ren B, Chiaravalloti TR, Belony NL, Romero DI, Chai W, Leon C, Wu L, Lamango NS, Offringa IA, Huang Y. Design and Realization of Lung Organoid Cultures for COVID-19 Applications. Biodes Manuf 2023; 6:646-660. [PMID: 38993804 PMCID: PMC11238720 DOI: 10.1007/s42242-023-00255-1] [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: 04/01/2023] [Accepted: 06/25/2023] [Indexed: 07/13/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been spreading globally and threatening public health. Advanced in vitro models that recapitulate the architecture and functioning of specific tissues and organs are in high demand for COVID-19-related pathology studies and drug screening. Three-dimensional (3D) in vitro cultures such as self-assembled and engineered organoid cultures surpass conventional two-dimensional (2D) cultures and animal models with respect to the increased cellular complexity, better human-relevant environment, and reduced cost, thus presenting as promising platforms for understanding viral pathogenesis and developing new therapeutics. This review highlights the recent advances in self-assembled and engineered organoid technologies that are used for COVID-19 studies. The challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | | | - Nadine L Belony
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Diana I Romero
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Wenxuan Chai
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Christopher Leon
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32611
| | - Nazarius S Lamango
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307
| | - Ite A Offringa
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| |
Collapse
|
11
|
Naderi-Meshkin H, Cornelius VA, Eleftheriadou M, Potel KN, Setyaningsih WAW, Margariti A. Vascular organoids: unveiling advantages, applications, challenges, and disease modelling strategies. Stem Cell Res Ther 2023; 14:292. [PMID: 37817281 PMCID: PMC10566155 DOI: 10.1186/s13287-023-03521-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Understanding mechanisms and manifestations of cardiovascular risk factors, including diabetes, on vascular cells such as endothelial cells, pericytes, and vascular smooth muscle cells, remains elusive partly due to the lack of appropriate disease models. Therefore, here we explore different aspects for the development of advanced 3D in vitro disease models that recapitulate human blood vessel complications using patient-derived induced pluripotent stem cells, which retain the epigenetic, transcriptomic, and metabolic memory of their patient-of-origin. In this review, we highlight the superiority of 3D blood vessel organoids over conventional 2D cell culture systems for vascular research. We outline the key benefits of vascular organoids in both health and disease contexts and discuss the current challenges associated with organoid technology, providing potential solutions. Furthermore, we discuss the diverse applications of vascular organoids and emphasize the importance of incorporating all relevant cellular components in a 3D model to accurately recapitulate vascular pathophysiology. As a specific example, we present a comprehensive overview of diabetic vasculopathy, demonstrating how the interplay of different vascular cell types is critical for the successful modelling of complex disease processes in vitro. Finally, we propose a strategy for creating an organ-specific diabetic vasculopathy model, serving as a valuable template for modelling other types of vascular complications in cardiovascular diseases by incorporating disease-specific stressors and organotypic modifications.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Victoria A Cornelius
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Magdalini Eleftheriadou
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Koray Niels Potel
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Wiwit Ananda Wahyu Setyaningsih
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, D.I. Yogyakarta, 55281, Indonesia
| | - Andriana Margariti
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
12
|
Qiu Y, Lu C, Bao F, Hu G. Design of a multilayer lung chip with multigenerational alveolar ducts to investigate the inhaled particle deposition. LAB ON A CHIP 2023; 23:4302-4312. [PMID: 37691540 DOI: 10.1039/d3lc00253e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
We present the development and application of a multilayer microfluidic lung chip designed to accurately replicate the human respiratory bronchi, providing an innovative platform for controlled particle deposition in the lung. By employing a quantitative control method of fluid velocity through the deformation of an elastic PDMS membrane, this platform mimics the passive breathing process in humans and allows for precise simulation of the respiration cycle. We utilized time-lapse photography of fluorescent particles in a water/glycerol solution to qualitatively observe fluid morphology in the channel, while a chip-aerosol exposure device combined with microscopy imaging was employed to visualise aerosol deposition. Both experimental and numerical simulation results showed that particle concentration decreased towards the distal generations of the lung, and that changes in breathing pattern significantly affected particle deposition trends. Furthermore, we found that increasing the residence time of particles in the channel facilitated deeper particle deposition, achievable by adjusting parameters such as breath-hold time, exhalation time, respiration cycle length, and tidal volume. The proposed microfluidic lung chip device has significant potential for future research in respiratory health and inhaled drug delivery, providing an efficient, cost-effective, and ethical alternative to traditional in vivo studies.
Collapse
Affiliation(s)
- Yan Qiu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China.
| | - Chao Lu
- College of Metrology and Measurement Engineering, China Jiliang University, Hangzhou 310018, China
| | - Fubing Bao
- Zhejiang Provincial Key Laboratory of Flow Measurement Technology, China Jiliang University, Hangzhou 310018, China
| | - Guoqing Hu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
13
|
Santos AK, Scalzo S, de Souza RTV, Santana PHG, Marques BL, Oliveira LF, Filho DM, Kihara AH, da Costa Santiago H, Parreira RC, Birbrair A, Ulrich H, Resende RR. Strategic use of organoids and organs-on-chip as biomimetic tools. Semin Cell Dev Biol 2023; 144:3-10. [PMID: 36192310 DOI: 10.1016/j.semcdb.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022]
Abstract
Organoid development and organ-on-a-chip are technologies based on differentiating stem cells, forming 3D multicellular structures resembling organs and tissues in vivo. Hence, both can be strategically used for disease modeling, drug screening, and host-pathogen studies. In this context, this review highlights the significant advancements in the area, providing technical approaches to organoids and organ-on-a-chip that best imitate in vivo physiology.
Collapse
Affiliation(s)
- Anderson K Santos
- Department of Pediatrics, Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Sérgio Scalzo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Lucas F Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Daniel M Filho
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexander Birbrair
- Departmento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Henning Ulrich
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinópolis, Brazil.
| |
Collapse
|
14
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
15
|
Satta S, Rockwood SJ, Wang K, Wang S, Mozneb M, Arzt M, Hsiai TK, Sharma A. Microfluidic Organ-Chips and Stem Cell Models in the Fight Against COVID-19. Circ Res 2023; 132:1405-1424. [PMID: 37167356 PMCID: PMC10171291 DOI: 10.1161/circresaha.122.321877] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
SARS-CoV-2, the virus underlying COVID-19, has now been recognized to cause multiorgan disease with a systemic effect on the host. To effectively combat SARS-CoV-2 and the subsequent development of COVID-19, it is critical to detect, monitor, and model viral pathogenesis. In this review, we discuss recent advancements in microfluidics, organ-on-a-chip, and human stem cell-derived models to study SARS-CoV-2 infection in the physiological organ microenvironment, together with their limitations. Microfluidic-based detection methods have greatly enhanced the rapidity, accessibility, and sensitivity of viral detection from patient samples. Engineered organ-on-a-chip models that recapitulate in vivo physiology have been developed for many organ systems to study viral pathology. Human stem cell-derived models have been utilized not only to model viral tropism and pathogenesis in a physiologically relevant context but also to screen for effective therapeutic compounds. The combination of all these platforms, along with future advancements, may aid to identify potential targets and develop novel strategies to counteract COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Sandro Satta
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Sarah J. Rockwood
- Stanford University Medical Scientist Training Program, Palo Alto, CA (S.J.R.)
| | - Kaidong Wang
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Shaolei Wang
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tzung K. Hsiai
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
16
|
Induced Pluripotent Stem Cell-Derived Organoids: Their Implication in COVID-19 Modeling. Int J Mol Sci 2023; 24:ijms24043459. [PMID: 36834870 PMCID: PMC9961667 DOI: 10.3390/ijms24043459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a significant global health issue. This novel virus's high morbidity and mortality rates have prompted the scientific community to quickly find the best COVID-19 model to investigate all pathological processes underlining its activity and, more importantly, search for optimal drug therapy with minimal toxicity risk. The gold standard in disease modeling involves animal and monolayer culture models; however, these models do not fully reflect the response to human tissues affected by the virus. However, more physiological 3D in vitro culture models, such as spheroids and organoids derived from induced pluripotent stem cells (iPSCs), could serve as promising alternatives. Different iPSC-derived organoids, such as lung, cardiac, brain, intestinal, kidney, liver, nasal, retinal, skin, and pancreatic organoids, have already shown immense potential in COVID-19 modeling. In the present comprehensive review article, we summarize the current knowledge on COVID-19 modeling and drug screening using selected iPSC-derived 3D culture models, including lung, brain, intestinal, cardiac, blood vessels, liver, kidney, and inner ear organoids. Undoubtedly, according to reviewed studies, organoids are the state-of-the-art approach to COVID-19 modeling.
Collapse
|
17
|
Wang H, Yin F, Li Z, Su W, Li D. Advances of microfluidic lung chips for assessing atmospheric pollutants exposure. ENVIRONMENT INTERNATIONAL 2023; 172:107801. [PMID: 36774736 DOI: 10.1016/j.envint.2023.107801] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Atmospheric pollutants, including particulate matters, nanoparticles, bioaerosols, and some chemicals, have posed serious threats to the environment and the human's health. The lungs are the responsible organs for providing the interface betweenthecirculatory system and the external environment, where pollutant particles can deposit or penetrate into bloodstream circulation. Conventional studies to decipher the mechanismunderlying air pollution and human health are quite limited, due to the lack of reliable models that can reproduce in vivo features of lung tissues after pollutants exposure. In the past decade, advanced near-to-native lung chips, combining cell biology with bioengineered technology, present a new strategy for atmospheric pollutants assessment and narrow the gap between 2D cell culture and in vivo animal models. In this review, the key features of artificial lung chips and the cutting-edge technologies of the lung chip manufacture are introduced. The recent progresses of lung chip technologies for atmospheric pollutants exposure assessment are summarized and highlighted. We further discuss the current challenges and the future opportunities of the development of advanced lung chips and their potential utilities in atmospheric pollutants associated toxicity testing and drug screening.
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
| | - Fangchao Yin
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Zhongyu Li
- College of Life Science, Dalian Minzu University, Dalian 116600, China
| | - Wentao Su
- Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034 Liaoning, China.
| | - Dong Li
- Medical School, Nantong University, Nantong 226001, China.
| |
Collapse
|
18
|
Ni K, Che B, Yang C, Qin Y, Gu R, Wang C, Luo M, Deng L. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Front Pharmacol 2022; 13:1033043. [PMID: 36578545 PMCID: PMC9790924 DOI: 10.3389/fphar.2022.1033043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a never before seen challenge to human health and the world economy. However, it is difficult to widely use conventional animal and cell culture models in understanding the underlying pathological mechanisms of COVID-19, which in turn hinders the development of relevant therapeutic treatments, including drugs. To overcome this challenge, various three-dimensional (3D) pulmonary cell culture models such as organoids are emerging as an innovative toolset for simulating the pathophysiology occurring in the respiratory system, including bronchial airways, alveoli, capillary network, and pulmonary interstitium, which provide a robust and powerful platform for studying the process and underlying mechanisms of SARS-CoV-2 infection among the potential primary targets in the lung. This review introduces the key features of some of these recently developed tools, including organoid, lung-on-a-chip, and 3D bioprinting, which can recapitulate different structural compartments of the lung and lung function, in particular, accurately resembling the human-relevant pathophysiology of SARS-CoV-2 infection in vivo. In addition, the recent progress in developing organoids for alveolar and airway disease modeling and their applications for discovering drugs against SARS-CoV-2 infection are highlighted. These innovative 3D cell culture models together may hold the promise to fully understand the pathogenesis and eventually eradicate the pandemic of COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| |
Collapse
|
19
|
Yu J, Wang K, Zheng D. Brain organoids for addressing COVID-19 challenge. Front Neurosci 2022; 16:1055601. [PMID: 36523428 PMCID: PMC9744798 DOI: 10.3389/fnins.2022.1055601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/02/2022] [Indexed: 09/19/2023] Open
Abstract
COVID-19 is a systemic disease involving multiple organs, and clinically, patients have symptoms of neurological damage to varying degrees. However, we do not have a clear understanding of the relationship between neurological manifestations and viral infection due to the limitations of current in vitro study models. Brain organoids, formed by the differentiation of stem cells under 3D culture conditions, can mimic the structure of tiny cell clusters with neurodevelopmental features in different patients. The paper reviewed the history of brain organoids development, the study of the mechanism of viral infection, the inflammatory response associated with neurological damage, the detection of antiviral drugs, and combined microarray technology to affirm the status of the brain organoid models in the study of COVID-19. In addition, our study continuously improved the model in combination with emerging technologies, to lay a theoretical foundation for clinical application and academic research.
Collapse
Affiliation(s)
- Jin Yu
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan, China
| | - Kailun Wang
- Department of Emergency, Panzhihua Central Hospital, Panzhihua, Sichuan, China
| | - Dalin Zheng
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan, China
| |
Collapse
|
20
|
Bai H, Ingber DE. What Can an Organ-on-a-Chip Teach Us About Human Lung Pathophysiology? Physiology (Bethesda) 2022; 37:0. [PMID: 35658627 PMCID: PMC9394778 DOI: 10.1152/physiol.00012.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022] Open
Abstract
The intertwined relationship between structure and function has been key to understanding human organ physiology and disease pathogenesis. An organ-on-a-chip (organ chip) is a bioengineered microfluidic cell culture device lined by living cells and tissues that recapitulates organ-level functions in vitro. This is accomplished by recreating organ-specific tissue-tissue interfaces and microenvironmental biochemical and mechanical cues while providing dynamic perfusion through endothelium-lined vascular channels. In this review, we discuss how this emerging technology has contributed to the understanding of human lung structure-function relationships at the cell, tissue, and organ levels.
Collapse
Affiliation(s)
- Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts
| |
Collapse
|
21
|
Wang F. Fighting the SARS-CoV-2 Pandemic: Focusing a New Lens on COVID-19. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9879646. [PMID: 35966758 PMCID: PMC9351585 DOI: 10.34133/2022/9879646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Fudi Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Hengyang Medical School, University of South of China, Hengyang 421001, China
| |
Collapse
|