1
|
Huang Z, Jia K, Tan Y, Yu Y, Xiao W, Zhou X, Yi J, Zhang C. Advances in cardiac organoid research: implications for cardiovascular disease treatment. Cardiovasc Diabetol 2025; 24:25. [PMID: 39827092 PMCID: PMC11743075 DOI: 10.1186/s12933-025-02598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Globally, cardiovascular diseases remain among the leading causes of mortality, highlighting the urgent need for innovative research models. Consequently, the development of accurate models that simulate cardiac function holds significant scientific and clinical value for both disease research and therapeutic interventions. Cardiac organoids, which are three-dimensional structures derived from the induced differentiation of stem cells, are particularly promising. These organoids not only replicate the autonomous beating and essential electrophysiological properties of the heart but are also widely employed in studies related to cardiac diseases, drug efficacy testing, and regenerative medicine. This review comprehensively surveys the various fabrication techniques used to create cardiac organoids and their diverse applications in modeling a range of cardiac diseases. We emphasize the role of advanced technologies in enhancing the maturation and functionality of cardiac cells, ensuring that these models closely resemble native cardiac tissue. Furthermore, we discuss monitoring techniques and evaluation parameters critical for assessing the performance of cardiac organoids, considering the complex interactions within multi-organ systems. This approach is vital for enhancing precision and efficiency in drug development, allowing for more effective therapeutic strategies. Ultimately, this review aims to provide a thorough and innovative perspective on both fundamental research and clinical treatment of cardiovascular diseases, offering insights that could pave the way for future advancements in understanding and addressing these prevalent health challenges.
Collapse
Affiliation(s)
- Ziteng Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yadan Tan
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yang Yu
- Department of Cardiology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wudian Xiao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
2
|
Wang Y, Hou Y, Hao T, Garcia-Contreras M, Li G, Cretoiu D, Xiao J. Model construction and clinical therapeutic potential of engineered cardiac organoids for cardiovascular diseases. BIOMATERIALS TRANSLATIONAL 2024; 5:337-354. [PMID: 39872935 PMCID: PMC11764187 DOI: 10.12336/biomatertransl.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/08/2024] [Accepted: 10/08/2024] [Indexed: 01/30/2025]
Abstract
Cardiovascular diseases cause significant morbidity and mortality worldwide. Engineered cardiac organoids are being developed and used to replicate cardiac tissues supporting cardiac morphogenesis and development. These organoids have applications in drug screening, cardiac disease models and regenerative medicine. Therefore, a thorough understanding of cardiac organoids and a comprehensive overview of their development are essential for cardiac tissue engineering. This review summarises different types of cardiac organoids used to explore cardiac function, including those based on co-culture, aggregation, scaffolds, and geometries. The self-assembly of monolayers, multilayers and aggravated cardiomyocytes forms biofunctional cell aggregates in cardiac organoids, elucidating the formation mechanism of scaffold-free cardiac organoids. In contrast, scaffolds such as decellularised extracellular matrices, three-dimensional hydrogels and bioprinting techniques provide a supportive framework for cardiac organoids, playing a crucial role in cardiac development. Different geometries are engineered to create cardiac organoids, facilitating the investigation of intrinsic communication between cardiac organoids and biomechanical pathways. Additionally, this review emphasises the relationship between cardiac organoids and the cardiac system, and evaluates their clinical applications. This review aims to provide valuable insights into the study of three-dimensional cardiac organoids and their clinical potential.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, School of Medicine, Shanghai University, Shanghai, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, China
| | - Yan Hou
- Cardiac Regeneration and Ageing Lab, School of Medicine, Shanghai University, Shanghai, China
| | - Tian Hao
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marta Garcia-Contreras
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dragos Cretoiu
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Materno-Fetal Assistance Excellence Unit, Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest, Romania
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, School of Medicine, Shanghai University, Shanghai, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
3
|
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater 2024; 40:88-103. [PMID: 38962658 PMCID: PMC11220467 DOI: 10.1016/j.bioactmat.2024.05.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiovascular disease is a significant cause of death in humans. Various models are necessary for the study of cardiovascular diseases, but once cellular and animal models have some defects, such as insufficient fidelity. As a new technology, organoid has certain advantages and has been used in many applications in the study of cardiovascular diseases. This article aims to summarize the application of organoid platforms in cardiovascular diseases, including organoid construction schemes, modeling, and application of cardiovascular organoids. Advances in cardiovascular organoid research have provided many models for different cardiovascular diseases in a variety of areas, including myocardium, blood vessels, and valves. Physiological and pathological models of different diseases, drug research models, and methods for evaluating and promoting the maturation of different kinds of organ tissues are provided for various cardiovascular diseases, including cardiomyopathy, myocardial infarction, and atherosclerosis. This article provides a comprehensive overview of the latest research progress in cardiovascular organ tissues, including construction protocols for cardiovascular organoid tissues and their evaluation system, different types of disease models, and applications of cardiovascular organoid models in various studies. The problems and possible solutions in organoid development are summarized.
Collapse
Affiliation(s)
- Xingchao Du
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| |
Collapse
|
4
|
Chen EZ, Kannan S, Murphy S, Farid M, Kwon C. Protocol for quantifying stem-cell-derived cardiomyocyte maturity using transcriptomic entropy score. STAR Protoc 2024; 5:103083. [PMID: 38781077 PMCID: PMC11145390 DOI: 10.1016/j.xpro.2024.103083] [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: 12/08/2023] [Revised: 03/15/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
The inability to quantify cardiomyocyte (CM) maturation remains a significant barrier to evaluating the effects of ongoing efforts to produce adult-like CMs from pluripotent stem cells (PSCs). Here, we present a protocol to quantify stem-cell-derived CM maturity using a single-cell RNA sequencing-based metric "entropy score." We describe steps for generating an entropy score using customized R code. This tool can be used to quantify maturation levels of PSC-CMs and potentially other cell types. For complete details on the use and execution of this protocol, please refer to Kannan et al.1.
Collapse
Affiliation(s)
- Elaine Zhelan Chen
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Michael Farid
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA.
| |
Collapse
|
5
|
Tahara S, Sharma S, de Faria FCC, Sarchet P, Tomasello L, Rentsch S, Karna R, Calore F, Pollock RE. Comparison of three-dimensional cell culture techniques of dedifferentiated liposarcoma and their integration with future research. Front Cell Dev Biol 2024; 12:1362696. [PMID: 38500686 PMCID: PMC10945377 DOI: 10.3389/fcell.2024.1362696] [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: 12/28/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Background: Dedifferentiated liposarcoma is a formidable sarcoma subtype due to its high local recurrence rate and resistance to medical treatment. While 2D cell cultures are still commonly used, 3D cell culture systems have emerged as a promising alternative, particularly scaffold-based techniques that enable the creation of 3D models with more accurate cell-stroma interactions. Objective: To investigate how 3D structures with or without the scaffold existence would affect liposarcoma cell lines growth morphologically and biologically. Methods: Lipo246 and Lipo863 cell lines were cultured in 3D using four different methods; Matrigel® ECM scaffold method, Collagen ECM scaffold method, ULA plate method and Hanging drop method, in addition to conventional 2D cell culture methods. All samples were processed for histopathological analysis (HE, IHC and DNAscope™), Western blot, and qPCR; moreover, 3D collagen-based models were treated with different doses of SAR405838, a well-known inhibitor of MDM2, and cell viability was assessed in comparison to 2D model drug response. Results: Regarding morphology, cell lines behaved differently comparing the scaffold-based and scaffold-free methods. Lipo863 formed spheroids in Matrigel® but not in collagen, while Lipo246 did not form spheroids in either collagen or Matrigel®. On the other hand, both cell lines formed spheroids using scaffold-free methods. All samples retained liposarcoma characteristic, such as high level of MDM2 protein expression and MDM2 DNA amplification after being cultivated in 3D. 3D collagen samples showed higher cell viability after SAR40538 treatment than 2D models, while cells sensitive to the drug died by apoptosis or necrosis. Conclusion: Our results prompt us to extend our investigation by applying our 3D models to further oncological relevant applications, which may help address unresolved questions about dedifferentiated liposarcoma biology.
Collapse
Affiliation(s)
- Sayumi Tahara
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Soumya Sharma
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Fernanda Costas Casal de Faria
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Patricia Sarchet
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Luisa Tomasello
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sydney Rentsch
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Roma Karna
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Raphael E. Pollock
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
6
|
Zhang Y, Liu K, He H, Xiao H, Fang Z, Chen X, Li H. Innovative explorations: unveiling the potential of organoids for investigating environmental pollutant exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:16256-16273. [PMID: 38342830 DOI: 10.1007/s11356-024-32256-7] [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: 09/21/2023] [Accepted: 01/25/2024] [Indexed: 02/13/2024]
Abstract
As the economy rapidly develops, chemicals are widely produced and used. This has exacerbated the problems associated with environmental pollution, raising the need for efficient toxicological evaluation techniques to investigate the toxic effects and mechanisms of toxicity of environmental pollutants. The progress in the techniques of cell culture in three dimensions has resulted in the creation of models that are more relevant in terms of biology and physiology. This enables researchers to study organ development, toxicology, and drug screening. Adult stem cells (ASCs) and induced pluripotent stem cells (iPSCs) can be obtained from various mammalian tissues, including cancerous and healthy tissues. Such stem cells exhibit a significant level of tissue memory and ability to self-assemble. When cultivated in 3D in vitro environments, the resulting organoids demonstrate a remarkable capacity to recapitulate the cellular composition and function of organs in vivo. Recently, many tumors' tissue-derived organoids have been widely used in research on tumor pathogenesis, drug development, precision medicine, and other fields, including those derived from colon cancer, cholangiocarcinoma, liver cancer, and gastric cancer. However, the application of organoid models for evaluating the toxicity of environmental pollutants is still in its infancy. This review introduces the characteristics of the toxicity responses of organoid models upon exposure to pollutants from the perspectives of organoid characteristics, tissue types, and their applications in toxicology; discusses the feasibility of using organoid models in evaluating the toxicity of pollutants; and provides a reference for future toxicological studies on environmental pollutants based on organoid models.
Collapse
Affiliation(s)
- Yuanhang Zhang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Kai Liu
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huan He
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China
| | - Hui Xiao
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Zhihong Fang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Xianxian Chen
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huiming Li
- School of Environment, Nanjing Normal University, Nanjing, 210023, China.
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China.
| |
Collapse
|
7
|
Yang Y, Yang H, Kiskin FN, Zhang JZ. The new era of cardiovascular research: revolutionizing cardiovascular research with 3D models in a dish. MEDICAL REVIEW (2021) 2024; 4:68-85. [PMID: 38515776 PMCID: PMC10954298 DOI: 10.1515/mr-2023-0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/18/2024] [Indexed: 03/23/2024]
Abstract
Cardiovascular research has heavily relied on studies using patient samples and animal models. However, patient studies often miss the data from the crucial early stage of cardiovascular diseases, as obtaining primary tissues at this stage is impracticable. Transgenic animal models can offer some insights into disease mechanisms, although they usually do not fully recapitulate the phenotype of cardiovascular diseases and their progression. In recent years, a promising breakthrough has emerged in the form of in vitro three-dimensional (3D) cardiovascular models utilizing human pluripotent stem cells. These innovative models recreate the intricate 3D structure of the human heart and vessels within a controlled environment. This advancement is pivotal as it addresses the existing gaps in cardiovascular research, allowing scientists to study different stages of cardiovascular diseases and specific drug responses using human-origin models. In this review, we first outline various approaches employed to generate these models. We then comprehensively discuss their applications in studying cardiovascular diseases by providing insights into molecular and cellular changes associated with cardiovascular conditions. Moreover, we highlight the potential of these 3D models serving as a platform for drug testing to assess drug efficacy and safety. Despite their immense potential, challenges persist, particularly in maintaining the complex structure of 3D heart and vessel models and ensuring their function is comparable to real organs. However, overcoming these challenges could revolutionize cardiovascular research. It has the potential to offer comprehensive mechanistic insights into human-specific disease processes, ultimately expediting the development of personalized therapies.
Collapse
Affiliation(s)
- Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Fedir N. Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Joe Z. Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| |
Collapse
|
8
|
Majumder J, Torr EE, Aisenbrey EA, Lebakken CS, Favreau PF, Richards WD, Yin Y, Chang Q, Murphy WL. Human induced pluripotent stem cell-derived planar neural organoids assembled on synthetic hydrogels. J Tissue Eng 2024; 15:20417314241230633. [PMID: 38361535 PMCID: PMC10868488 DOI: 10.1177/20417314241230633] [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: 10/30/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024] Open
Abstract
The tailorable properties of synthetic polyethylene glycol (PEG) hydrogels make them an attractive substrate for human organoid assembly. Here, we formed human neural organoids from iPSC-derived progenitor cells in two distinct formats: (i) cells seeded on a Matrigel surface; and (ii) cells seeded on a synthetic PEG hydrogel surface. Tissue assembly on synthetic PEG hydrogels resulted in three dimensional (3D) planar neural organoids with greater neuronal diversity, greater expression of neurovascular and neuroinflammatory genes, and reduced variability when compared with tissues assembled upon Matrigel. Further, our 3D human tissue assembly approach occurred in an open cell culture format and created a tissue that was sufficiently translucent to allow for continuous imaging. Planar neural organoids formed on PEG hydrogels also showed higher expression of neural, vascular, and neuroinflammatory genes when compared to traditional brain organoids grown in Matrigel suspensions. Further, planar neural organoids contained functional microglia that responded to pro-inflammatory stimuli, and were responsive to anti-inflammatory drugs. These results demonstrate that the PEG hydrogel neural organoids can be used as a physiologically relevant in vitro model of neuro-inflammation.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth E Torr
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth A Aisenbrey
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | - Yanhong Yin
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Medical Genetics and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
9
|
An Y, He Y, Ge N, Guo J, Yang F, Sun S. Organoids to Remodel SARS-CoV-2 Research: Updates, Limitations and Perspectives. Aging Dis 2023; 14:1677-1699. [PMID: 37196111 PMCID: PMC10529756 DOI: 10.14336/ad.2023.0209] [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: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 05/19/2023] Open
Abstract
The novel COVID-19 pneumonia caused by the SARS-CoV-2 virus poses a significant threat to human health. Scientists have made significant efforts to control this virus, consequently leading to the development of novel research methods. Traditional animal and 2D cell line models might not be suitable for large-scale applications in SARS-CoV-2 research owing to their limitations. As an emerging modelling method, organoids have been applied in the study of various diseases. Their advantages include their ability to closely mirror human physiology, ease of cultivation, low cost, and high reliability; thus, they are considered to be a suitable choice to further the research on SARS-CoV-2. During the course of various studies, SARS-CoV-2 was shown to infect a variety of organoid models, exhibiting changes similar to those observed in humans. This review summarises the various organoid models used in SARS-CoV-2 research, revealing the molecular mechanisms of viral infection and exploring the drug screening tests and vaccine research that have relied on organoid models, hence illustrating the role of organoids in remodelling SARS-CoV-2 research.
Collapse
Affiliation(s)
- Yucheng An
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Nan Ge
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Jintao Guo
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Fan Yang
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
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
|
11
|
Muniyandi P, O’Hern C, Popa MA, Aguirre A. Biotechnological advances and applications of human pluripotent stem cell-derived heart models. Front Bioeng Biotechnol 2023; 11:1214431. [PMID: 37560538 PMCID: PMC10407810 DOI: 10.3389/fbioe.2023.1214431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
In recent years, significant biotechnological advancements have been made in engineering human cardiac tissues and organ-like models. This field of research is crucial for both basic and translational research due to cardiovascular disease being the leading cause of death in the developed world. Additionally, drug-associated cardiotoxicity poses a major challenge for drug development in the pharmaceutical and biotechnological industries. Progress in three-dimensional cell culture and microfluidic devices has enabled the generation of human cardiac models that faithfully recapitulate key aspects of human physiology. In this review, we will discuss 3D pluripotent stem cell (PSC)-models of the human heart, such as engineered heart tissues and organoids, and their applications in disease modeling and drug screening.
Collapse
Affiliation(s)
- Priyadharshni Muniyandi
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Colin O’Hern
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Mirel Adrian Popa
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
12
|
Dow LP, Parmar T, Marchetti MC, Pruitt BL. Engineering tools for quantifying and manipulating forces in epithelia. BIOPHYSICS REVIEWS 2023; 4:021303. [PMID: 38510344 PMCID: PMC10903508 DOI: 10.1063/5.0142537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/20/2023] [Indexed: 03/22/2024]
Abstract
The integrity of epithelia is maintained within dynamic mechanical environments during tissue development and homeostasis. Understanding how epithelial cells mechanosignal and respond collectively or individually is critical to providing insight into developmental and (patho)physiological processes. Yet, inferring or mimicking mechanical forces and downstream mechanical signaling as they occur in epithelia presents unique challenges. A variety of in vitro approaches have been used to dissect the role of mechanics in regulating epithelia organization. Here, we review approaches and results from research into how epithelial cells communicate through mechanical cues to maintain tissue organization and integrity. We summarize the unique advantages and disadvantages of various reduced-order model systems to guide researchers in choosing appropriate experimental systems. These model systems include 3D, 2D, and 1D micromanipulation methods, single cell studies, and noninvasive force inference and measurement techniques. We also highlight a number of in silico biophysical models that are informed by in vitro and in vivo observations. Together, a combination of theoretical and experimental models will aid future experiment designs and provide predictive insight into mechanically driven behaviors of epithelial dynamics.
Collapse
Affiliation(s)
| | - Toshi Parmar
- Department of Physics, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | | | | |
Collapse
|
13
|
Alhejailan RS, Garoffolo G, Raveendran VV, Pesce M. Cells and Materials for Cardiac Repair and Regeneration. J Clin Med 2023; 12:jcm12103398. [PMID: 37240504 DOI: 10.3390/jcm12103398] [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: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
After more than 20 years following the introduction of regenerative medicine to address the problem of cardiac diseases, still questions arise as to the best cell types and materials to use to obtain effective clinical translation. Now that it is definitively clear that the heart does not have a consistent reservoir of stem cells that could give rise to new myocytes, and that there are cells that could contribute, at most, with their pro-angiogenic or immunomodulatory potential, there is fierce debate on what will emerge as the winning strategy. In this regard, new developments in somatic cells' reprogramming, material science and cell biophysics may be of help, not only for protecting the heart from the deleterious consequences of aging, ischemia and metabolic disorders, but also to boost an endogenous regeneration potential that seems to be lost in the adulthood of the human heart.
Collapse
Affiliation(s)
- Reem Saud Alhejailan
- Cell Biology Department, King's Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Vineesh Vimala Raveendran
- Cell Biology Department, King's Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| |
Collapse
|
14
|
Chun YW, Miyamoto M, Williams CH, Neitzel LR, Silver-Isenstadt M, Cadar AG, Fuller DT, Fong DC, Liu H, Lease R, Kim S, Katagiri M, Durbin MD, Wang KC, Feaster TK, Sheng CC, Neely MD, Sreenivasan U, Cortes-Gutierrez M, Finn AV, Schot R, Mancini GMS, Ament SA, Ess KC, Bowman AB, Han Z, Bichell DP, Su YR, Hong CC. Impaired Reorganization of Centrosome Structure Underlies Human Infantile Dilated Cardiomyopathy. Circulation 2023; 147:1291-1303. [PMID: 36970983 PMCID: PMC10133173 DOI: 10.1161/circulationaha.122.060985] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND During cardiomyocyte maturation, the centrosome, which functions as a microtubule organizing center in cardiomyocytes, undergoes dramatic structural reorganization where its components reorganize from being localized at the centriole to the nuclear envelope. This developmentally programmed process, referred to as centrosome reduction, has been previously associated with cell cycle exit. However, understanding of how this process influences cardiomyocyte cell biology, and whether its disruption results in human cardiac disease, remains unknown. We studied this phenomenon in an infant with a rare case of infantile dilated cardiomyopathy (iDCM) who presented with left ventricular ejection fraction of 18% and disrupted sarcomere and mitochondria structure. METHODS We performed an analysis beginning with an infant who presented with a rare case of iDCM. We derived induced pluripotent stem cells from the patient to model iDCM in vitro. We performed whole exome sequencing on the patient and his parents for causal gene analysis. CRISPR/Cas9-mediated gene knockout and correction in vitro were used to confirm whole exome sequencing results. Zebrafish and Drosophila models were used for in vivo validation of the causal gene. Matrigel mattress technology and single-cell RNA sequencing were used to characterize iDCM cardiomyocytes further. RESULTS Whole exome sequencing and CRISPR/Cas9 gene knockout/correction identified RTTN, the gene encoding the centrosomal protein RTTN (rotatin), as the causal gene underlying the patient's condition, representing the first time a centrosome defect has been implicated in a nonsyndromic dilated cardiomyopathy. Genetic knockdowns in zebrafish and Drosophila confirmed an evolutionarily conserved requirement of RTTN for cardiac structure and function. Single-cell RNA sequencing of iDCM cardiomyocytes showed impaired maturation of iDCM cardiomyocytes, which underlie the observed cardiomyocyte structural and functional deficits. We also observed persistent localization of the centrosome at the centriole, contrasting with expected programmed perinuclear reorganization, which led to subsequent global microtubule network defects. In addition, we identified a small molecule that restored centrosome reorganization and improved the structure and contractility of iDCM cardiomyocytes. CONCLUSIONS This study is the first to demonstrate a case of human disease caused by a defect in centrosome reduction. We also uncovered a novel role for RTTN in perinatal cardiac development and identified a potential therapeutic strategy for centrosome-related iDCM. Future study aimed at identifying variants in centrosome components may uncover additional contributors to human cardiac disease.
Collapse
Affiliation(s)
- Young Wook Chun
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Matthew Miyamoto
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Charles H. Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Leif R. Neitzel
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Maya Silver-Isenstadt
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Adrian G. Cadar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Daniela T. Fuller
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Daniel C. Fong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Hanhan Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Robert Lease
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sungseek Kim
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Mikako Katagiri
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Matthew D. Durbin
- Division of Neonatology-Perinatology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 26202
| | - Kuo-Chen Wang
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Tromondae K. Feaster
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Calvin C. Sheng
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - M. Diana Neely
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37201
| | - Urmila Sreenivasan
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Marcia Cortes-Gutierrez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aloke V. Finn
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Rachel Schot
- Division of Neonatology-Perinatology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 26202
| | - Grazia M. S. Mancini
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kevin C. Ess
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN37201
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47906
| | - Zhe Han
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - David P. Bichell
- Department of Pediatric Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Yan Ru Su
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| |
Collapse
|
15
|
Yang R, Yu Y. Patient-derived organoids in translational oncology and drug screening. Cancer Lett 2023; 562:216180. [PMID: 37061121 DOI: 10.1016/j.canlet.2023.216180] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Patient-derived organoids (PDO) are a new biomedical research model that can reconstruct phenotypic and genetic characteristics of the original tissue and are useful for research on pathogenesis and drug screening. To introduce the progression in this field, we review the key factors of constructing organoids derived from epithelial tissues and cancers, covering culture medium and matrix, morphological characteristics, genetic profiles, high-throughput drug screening, and application potential. We also discuss the co-culture system of cancer organoids with tumor microenvironment (TME) associated cells. The co-culture system is widely used in evaluating crosstalk of cancer cells with TME components, such as fibroblasts, endothelial cells, immune cells, and microorganisms. The article provides a prospective for standardized cultivation mode, automatic morphological evaluation, and drug sensitivity screening using high-throughput methods.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
16
|
Liu Z, Lami B, Ikonomou L, Gu M. Unlocking the potential of induced pluripotent stem cells for neonatal disease modeling and drug development. Semin Perinatol 2023; 47:151729. [PMID: 37012138 PMCID: PMC10133195 DOI: 10.1016/j.semperi.2023.151729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Neonatal lung and heart diseases, albeit rare, can result in poor quality of life, often require long-term management and/or organ transplantation. For example, Congenital Heart Disease (CHD) is one of the most common type of congenital disabilities, affecting nearly 1% of the newborns, and has complex and multifactorial causes, including genetic predisposition and environmental influences. To develop new strategies for heart and lung regeneration in CHD and neonatal lung disease, human induced pluripotent stem cells (hiPSCs) provide a unique and personalized platform for future cell replacement therapy and high-throughput drug screening. Additionally, given the differentiation potential of iPSCs, cardiac cell types such as cardiomyocytes, endothelial cells, and fibroblasts and lung cell types such Type II alveolar epithelial cells can be derived in a dish to study the fundamental pathology during disease progression. In this review, we discuss the applications of hiPSCs in understanding the molecular mechanisms and cellular phenotypes of CHD (e.g., structural heart defect, congenital valve disease, and congenital channelopathies) and congenital lung diseases, such as surfactant deficiencies and Brain-Lung-Thyroid syndrome. We also provide future directions for generating mature cell types from iPSCs, and more complex hiPSC-based systems using three-dimensional (3D) organoids and tissue-engineering. With these potential advancements, the promise that hiPSCs will deliver new CHD and neonatal lung disease treatments may soon be fulfilled.
Collapse
Affiliation(s)
- Ziyi Liu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Bonny Lami
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Laertis Ikonomou
- Department of Oral Biology, University at Buffalo, The State University of New York, Buffalo, NY, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, The State University of New York, Buffalo, NY, United States; Cell, Gene and Tissue Engineering Center, University at Buffalo, The State University of New York, Buffalo, NY, United States.
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
17
|
Joddar B, Natividad-Diaz SL, Padilla AE, Esparza AA, Ramirez SP, Chambers DR, Ibaroudene H. Engineering approaches for cardiac organoid formation and their characterization. Transl Res 2022; 250:46-67. [PMID: 35995380 PMCID: PMC10370285 DOI: 10.1016/j.trsl.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
Cardiac organoids are 3-dimensional (3D) structures composed of tissue or niche-specific cells, obtained from diverse sources, encapsulated in either a naturally derived or synthetic, extracellular matrix scaffold, and include exogenous biochemical signals such as essential growth factors. The overarching goal of developing cardiac organoid models is to establish a functional integration of cardiomyocytes with physiologically relevant cells, tissues, and structures like capillary-like networks composed of endothelial cells. These organoids used to model human heart anatomy, physiology, and disease pathologies in vitro have the potential to solve many issues related to cardiovascular drug discovery and fundamental research. The advent of patient-specific human-induced pluripotent stem cell-derived cardiovascular cells provide a unique, single-source approach to study the complex process of cardiovascular disease progression through organoid formation and incorporation into relevant, controlled microenvironments such as microfluidic devices. Strategies that aim to accomplish such a feat include microfluidic technology-based approaches, microphysiological systems, microwells, microarray-based platforms, 3D bioprinted models, and electrospun fiber mat-based scaffolds. This article discusses the engineering or technology-driven practices for making cardiac organoid models in comparison with self-assembled or scaffold-free methods to generate organoids. We further discuss emerging strategies for characterization of the bio-assembled cardiac organoids including electrophysiology and machine-learning and conclude with prospective points of interest for engineering cardiac tissues in vitro.
Collapse
Affiliation(s)
- Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Andie E Padilla
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Aibhlin A Esparza
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | | | | |
Collapse
|
18
|
Miyamoto M, Kwon C. Uncovering the origins and lineage markers of human heart fields. Cell Stem Cell 2022; 29:1285-1287. [PMID: 36055186 PMCID: PMC9479643 DOI: 10.1016/j.stem.2022.08.004] [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] [Indexed: 11/29/2022]
Abstract
In this issue of Cell Stem Cell, Yang et al. devise a protocol for induction and differentiation of human heart-field precursors, drawing on inspiration from in vivo development. Intricate computational analyses uncovered conserved factors governing heart-field segregation, which facilitate enhanced study of human heart development and disease in the dish.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Department of Biomedical Engineering, Department of Cell Biology, Heart and Vascular Institute, Johns Hopkins University, Baltimore, MD 21042, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Department of Biomedical Engineering, Department of Cell Biology, Heart and Vascular Institute, Johns Hopkins University, Baltimore, MD 21042, USA.
| |
Collapse
|
19
|
Roacho-Pérez JA, Garza-Treviño EN, Moncada-Saucedo NK, Carriquiry-Chequer PA, Valencia-Gómez LE, Matthews ER, Gómez-Flores V, Simental-Mendía M, Delgado-Gonzalez P, Delgado-Gallegos JL, Padilla-Rivas GR, Islas JF. Artificial Scaffolds in Cardiac Tissue Engineering. Life (Basel) 2022; 12:1117. [PMID: 35892919 PMCID: PMC9331725 DOI: 10.3390/life12081117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/08/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are a leading cause of death worldwide. Current treatments directed at heart repair have several disadvantages, such as a lack of donors for heart transplantation or non-bioactive inert materials for replacing damaged tissue. Because of the natural lack of regeneration of cardiomyocytes, new treatment strategies involve stimulating heart tissue regeneration. The basic three elements of cardiac tissue engineering (cells, growth factors, and scaffolds) are described in this review, with a highlight on the role of artificial scaffolds. Scaffolds for cardiac tissue engineering are tridimensional porous structures that imitate the extracellular heart matrix, with the ability to promote cell adhesion, migration, differentiation, and proliferation. In the heart, there is an important requirement to provide scaffold cellular attachment, but scaffolds also need to permit mechanical contractility and electrical conductivity. For researchers working in cardiac tissue engineering, there is an important need to choose an adequate artificial scaffold biofabrication technique, as well as the ideal biocompatible biodegradable biomaterial for scaffold construction. Finally, there are many suitable options for researchers to obtain scaffolds that promote cell-electrical interactions and tissue repair, reaching the goal of cardiac tissue engineering.
Collapse
Affiliation(s)
- Jorge A. Roacho-Pérez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Nidia K. Moncada-Saucedo
- Servicio de Hematología, University Hospital “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico;
| | - Pablo A. Carriquiry-Chequer
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Laura E. Valencia-Gómez
- Instituto de Ingeniería y Tecnología, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Mexico; (L.E.V.-G.); (V.G.-F.)
| | - Elizabeth Renee Matthews
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA;
| | - Víctor Gómez-Flores
- Instituto de Ingeniería y Tecnología, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Mexico; (L.E.V.-G.); (V.G.-F.)
| | - Mario Simental-Mendía
- Orthopedic Trauma Service, University Hospital “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico;
| | - Paulina Delgado-Gonzalez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Juan Luis Delgado-Gallegos
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Gerardo R. Padilla-Rivas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| | - Jose Francisco Islas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.A.C.-C.); (P.D.-G.); (J.L.D.-G.); (G.R.P.-R.)
| |
Collapse
|
20
|
Lewis‐Israeli YR, Abdelhamid M, Olomu I, Aguirre A. Modeling the Effects of Maternal Diabetes on the Developing Human Heart Using Pluripotent Stem Cell-Derived Heart Organoids. Curr Protoc 2022; 2:e461. [PMID: 35723517 PMCID: PMC9219413 DOI: 10.1002/cpz1.461] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital heart defects (CHD) constitute the most common type of birth defect in humans. Maternal diabetes during the first trimester of pregnancy (pregestational diabetes, or PGD) is one of the most prominent factors contributing to CHD, and is present in a significant population of female patients with diabetes in reproductive age. PGD is challenging to manage clinically due to the extreme sensitivity of the developing embryo to glucose oscillations, and constitutes a critical health problem for the mother and the fetus. The prevalence of PGD-induced CHD is increasing due to the ongoing diabetes epidemic. While studies using animal models and cells in culture have demonstrated that PGD alters critical cellular and developmental processes, the mechanisms remain obscure, and it is unclear to what extent these models recapitulate PGD-induced CHD in humans. Clinical practice precludes direct studies in developing human embryos, further highlighting the need for physiologically relevant models. To bypass many of these technical and ethical limitations, we describe here a human pluripotent stem cell (hPSC)-based method to generate developmentally relevant self-organizing human heart organoids. By using glucose and insulin to mimic the diabetic environment that the embryo faces in PGD, this system allows modeling critical features of PGD in a human system with relevant physiology, structure, and cell types. The protocol starts with the generation of hPSC-derived embryoid bodies in a 96-well plate, followed by a small molecule-based three-step Wnt activation/inhibition/activation strategy. Organoids are then differentiated under healthy (normal insulin and glucose) and diabetic conditions (high insulin and glucose) over time, allowing for the study of the effects of pregestational diabetes on the developing human heart. We also provide an immunofluorescence protocol for comparing, characterizing, and analyzing the differences between the healthy and diabetic organoids, and comment on additional steps for preparing the organoids for analysis by other techniques after differentiation. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Generation of hPSC-derived embryoid bodies Basic Protocol 2: Differentiation of EBs into heart organoids under healthy and diabetes-like conditions Basic Protocol 3: Immunofluorescence and organoid preparation for other assays.
Collapse
Affiliation(s)
- Yonatan R. Lewis‐Israeli
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan
- Department of Biomedical Engineering, College of EngineeringMichigan State UniversityEast LansingMichigan
| | - Mishref Abdelhamid
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human MedicineMichigan State UniversityEast LansingMichigan
| | - Isoken Olomu
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human MedicineMichigan State UniversityEast LansingMichigan
| | - Aitor Aguirre
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan
- Department of Biomedical Engineering, College of EngineeringMichigan State UniversityEast LansingMichigan
| |
Collapse
|
21
|
Pang JKS, Ho BX, Chan WK, Soh BS. Insights to Heart Development and Cardiac Disease Models Using Pluripotent Stem Cell Derived 3D Organoids. Front Cell Dev Biol 2021; 9:788955. [PMID: 34926467 PMCID: PMC8675211 DOI: 10.3389/fcell.2021.788955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Medical research in the recent years has achieved significant progress due to the increasing prominence of organoid technology. Various developed tissue organoids bridge the limitations of conventional 2D cell culture and animal models by recapitulating in vivo cellular complexity. Current 3D cardiac organoid cultures have shown their utility in modelling key developmental hallmarks of heart organogenesis, but the complexity of the organ demands a more versatile model that can investigate more fundamental parameters, such as structure, organization and compartmentalization of a functioning heart. This review will cover the prominence of cardiac organoids in recent research, unpack current in vitro 3D models of the developing heart and look into the prospect of developing physiologically appropriate cardiac organoids with translational applicability. In addition, we discuss some of the limitations of existing cardiac organoid models in modelling embryonic development of the heart and manifestation of cardiac diseases.
Collapse
Affiliation(s)
- Jeremy Kah Sheng Pang
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Beatrice Xuan Ho
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Woon-Khiong Chan
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Abstract
Organoids-cellular aggregates derived from stem or progenitor cells that recapitulate organ function in miniature-are of growing interest in developmental biology and medicine. Organoids have been developed for organs and tissues such as the liver, gut, brain, and pancreas; they are used as organ surrogates to study a wide range of questions in basic and developmental biology, genetic disorders, and therapies. However, many organoids reported to date have been cultured in Matrigel, which is prepared from the secretion of Engelbreth-Holm-Swarm mouse sarcoma cells; Matrigel is complex and poorly defined. This complexity makes it difficult to elucidate Matrigel-specific factors governing organoid development. In this review, we discuss promising Matrigel-free methods for the generation and maintenance of organoids that use decellularized extracellular matrix (ECM), synthetic hydrogels, or gel-forming recombinant proteins.
Collapse
Affiliation(s)
- Mark T Kozlowski
- DEVCOM US Army Research Laboratory, Weapons and Materials Research Directorate, Science of Extreme Materials Division, Polymers Branch, 6300 Rodman Rd. Building 4600, Aberdeen Proving Ground, Aberdeen, MD, 21005, USA.
| | - Christiana J Crook
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
23
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
24
|
Lescroart F, Dumas CE, Adachi N, Kelly RG. Emergence of heart and branchiomeric muscles in cardiopharyngeal mesoderm. Exp Cell Res 2021; 410:112931. [PMID: 34798131 DOI: 10.1016/j.yexcr.2021.112931] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/27/2021] [Accepted: 11/14/2021] [Indexed: 12/17/2022]
Abstract
Branchiomeric muscles of the head and neck originate in a population of cranial mesoderm termed cardiopharyngeal mesoderm that also contains progenitor cells contributing to growth of the embryonic heart. Retrospective lineage analysis has shown that branchiomeric muscles share a clonal origin with parts of the heart, indicating the presence of common heart and head muscle progenitor cells in the early embryo. Genetic lineage tracing and functional studies in the mouse, as well as in Ciona and zebrafish, together with recent experiments using single cell transcriptomics and multipotent stem cells, have provided further support for the existence of bipotent head and heart muscle progenitor cells. Current challenges concern defining where and when such common progenitor cells exist in mammalian embryos and how alternative myogenic derivatives emerge in cardiopharyngeal mesoderm. Addressing these questions will provide insights into mechanisms of cell fate acquisition and the evolution of vertebrate musculature, as well as clinical insights into the origins of muscle restricted myopathies and congenital defects affecting craniofacial and cardiac development.
Collapse
Affiliation(s)
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France.
| |
Collapse
|
25
|
Shewale B, Dubois N. Of form and function: Early cardiac morphogenesis across classical and emerging model systems. Semin Cell Dev Biol 2021; 118:107-118. [PMID: 33994301 PMCID: PMC8434962 DOI: 10.1016/j.semcdb.2021.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022]
Abstract
The heart is the earliest organ to develop during embryogenesis and is remarkable in its ability to function efficiently as it is being sculpted. Cardiac heart defects account for a high burden of childhood developmental disorders with many remaining poorly understood mechanistically. Decades of work across a multitude of model organisms has informed our understanding of early cardiac differentiation and morphogenesis and has simultaneously opened new and unanswered questions. Here we have synthesized current knowledge in the field and reviewed recent developments in the realm of imaging, bioengineering and genetic technology and ex vivo cardiac modeling that may be deployed to generate more holistic models of early cardiac morphogenesis, and by extension, new platforms to study congenital heart defects.
Collapse
Affiliation(s)
- Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
26
|
Tampakakis E, Kwon C. Heart generation and regeneration. Semin Cell Dev Biol 2021; 118:92-93. [PMID: 34304994 DOI: 10.1016/j.semcdb.2021.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Chulan Kwon
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Department of Cell Biology, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|