1
|
Yoshida A, Baba K, Takahashi H, Nagese K, Shimizu T. One-step fabrication of 3D-aligned human skeletal muscle tissue and measurement of contractile force for preclinical drug testing. Mater Today Bio 2025; 31:101456. [PMID: 39896285 PMCID: PMC11783003 DOI: 10.1016/j.mtbio.2025.101456] [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: 07/12/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Human muscle tissue models are critical to understanding the complex physiology of skeletal muscle in studies of drug discovery, development, and toxicity profiling in the human body. However, due to the challenges in in vitro maturation of human muscle cells, few research groups developing their own tissue engineering techniques have succeeded in producing contractile human muscle tissues. Moreover, a more sophisticated method is necessary to measure contractile forces generated by the muscle tissues for preclinical studies in muscle physiology and drug discovery. Although a few research groups have established their own tissue model systems that measure contractile force, they require multi-step fabrication processes to produce human muscle tissues sufficiently functional to be able to measure the contractile forces. To improve the usability of our tissue model system, this study focused on simplifying the tissue engineering approach to produce a practical muscle tissue model. In this study, muscle satellite cells were simply mixed with a combination of fibrinogen, thrombin, and Matrigel before gel formation. The presence of muscle satellite cells induces gel compaction and spontaneously induces unidirectional stretching of the gel, resulting in the muscle satellite cells being aligned three-dimensionally with the direction of stretching. Furthermore, this gel environment promotes the maturation of the human muscle progenitor cells into aligned myofibers, also provides the tissue with an elastic platform for muscle contraction, and allows the attachment of the muscle tissue to a device for measurement of contractile force. Therefore, this one-step tissue fabrication allowed us to produce 3D-aligned human muscle tissues and this tissue model is ready to use for the measurement of contractile forces. In fact, the muscle contractions created by electrical and chemical stimulation were quantitatively determined using our measurement system. In addition, the impact of some representative drugs on this muscle tissue were able to be monitored in real-time throughout the changes in contractile forces. In conclusion, our tissue model system, produced by a simple fabrication method, can be used for preclinical in vitro studies in muscle physiology and drug discovery.
Collapse
Affiliation(s)
- Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kazuki Baba
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kenichi Nagese
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
- Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| |
Collapse
|
2
|
Li Z, Kovács P, Friec AL, Jensen BN, Nygaard JV, Chen M. Mechanical memory based biofabrication of hierarchical elastic cardiac tissue. Biofabrication 2024; 17:015013. [PMID: 39437832 DOI: 10.1088/1758-5090/ad89fd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
Mimicking the multilayered, anisotropic, elastic structure of cardiac tissues for controlled guidiance of 3D cellular orientation is essential in designing bionic scaffolds for cardiac tissue biofabrication. Here, a hierarchically organized, anisotropic, wavy and conductive polycaprolactone/Au scaffold was created in a facile fashion based on mechanical memory during fabrication. The bionic 3D scaffold shows good biocompatibility, excellent biomimetic mechanical properties that guide myoblast alignment, support the hyperelastic behavior observed in native cardiac muscle tissue, and promote myotube maturation, which holds potential for cardiac muscle engineering and the establishment of anin vitroculture platform for drug screening.
Collapse
Affiliation(s)
- Zhitong Li
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150000, Heilongjiang, People's Republic of China
| | - Panna Kovács
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | - Alice Le Friec
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | | | - Jens Vinge Nygaard
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | - Menglin Chen
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Zhang X, Zhao G, Ma T, Simmons CA, Santerre JP. A critical review on advances and challenges of bioprinted cardiac patches. Acta Biomater 2024; 189:1-24. [PMID: 39374681 DOI: 10.1016/j.actbio.2024.09.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Myocardial infarction (MI), which causes irreversible myocardium necrosis, affects 0.25 billion people globally and has become one of the most significant epidemics of our time. Over the past few years, bioprinting has moved beyond a concept of simply incorporating cells into biomaterials, to strategically defining the microenvironment (e.g., architecture, biomolecular signalling, mechanical stimuli, etc.) within which the cells are printed. Among the different bioprinting applications, myocardial repair is a field that has seen some of the most significant advances towards the management of the repaired tissue microenvironment. This review critically assesses the most recent biomedical innovations being carried out in cardiac patch bioprinting, with specific considerations given to the biomaterial design parameters, growth factors/cytokines, biomechanical and bioelectrical conditioning, as well as innovative biomaterial-based "4D" bioprinting (3D scaffold structure + temporal morphology changes) of myocardial tissues, immunomodulation and sustained delivery systems used in myocardium bioprinting. Key challenges include the ability to generate large quantities of cardiac cells, achieve high-density capillary networks, establish biomaterial designs that are comparable to native cardiac extracellular matrix, and manage the sophisticated systems needed for combining cardiac tissue microenvironmental cues while simultaneously establishing bioprinting technologies yielding both high-speed and precision. This must be achieved while considering quality assurance towards enabling reproducibility and clinical translation. Moreover, this manuscript thoroughly discussed the current clinical translational hurdles and regulatory issues associated with the post-bioprinting evaluation, storage, delivery and implantation of the bioprinted myocardial patches. Overall, this paper provides insights into how the clinical feasibility and important regulatory concerns may influence the design of the bioink (biomaterials, cell sources), fabrication and post-fabrication processes associated with bioprinting of the cardiac patches. This paper emphasizes that cardiac patch bioprinting requires extensive collaborations from imaging and 3D modelling technical experts, biomaterial scientists, additive manufacturing experts and healthcare professionals. Further, the work can also guide the field of cardiac patch bioprinting moving forward, by shedding light on the potential use of robotics and automation to increase productivity, reduce financial cost, and enable standardization and true commercialization of bioprinted cardiac patches. STATEMENT OF SIGNIFICANCE: The manuscript provides a critical review of important themes currently pursued for heart patch bioprinting, including critical biomaterial design parameters, physiologically-relevant cardiac tissue stimulations, and newly emerging cardiac tissue bioprinting strategies. This review describes the limited number of studies, to date in the literature, that describe systemic approaches to combine multiple design parameters, including capabilities to yield high-density capillary networks, establish biomaterial composite designs similar to native cardiac extracellular matrix, and incorporate cardiac tissue microenvironmental cues, while simultaneously establishing bioprinting technologies that yield high-speed and precision. New tools such as artificial intelligence may provide the analytical power to consider multiple design parameters and identify an optimized work-flow(s) for enabling the clinical translation of bioprinted cardiac patches.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - Guangtao Zhao
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Tianyi Ma
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Craig A Simmons
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - J Paul Santerre
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
4
|
Padmanaban AM, Ganesan K, Ramkumar KM. A Co-Culture System for Studying Cellular Interactions in Vascular Disease. Bioengineering (Basel) 2024; 11:1090. [PMID: 39593750 PMCID: PMC11591305 DOI: 10.3390/bioengineering11111090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are leading causes of morbidity and mortality globally, characterized by complications such as heart failure, atherosclerosis, and coronary artery disease. The vascular endothelium, forming the inner lining of blood vessels, plays a pivotal role in maintaining vascular homeostasis. The dysfunction of endothelial cells contributes significantly to the progression of CVDs, particularly through impaired cellular communication and paracrine signaling with other cell types, such as smooth muscle cells and macrophages. In recent years, co-culture systems have emerged as advanced in vitro models for investigating these interactions and mimicking the pathological environment of CVDs. This review provides an in-depth analysis of co-culture models that explore endothelial cell dysfunction and the role of cellular interactions in the development of vascular diseases. It summarizes recent advancements in multicellular co-culture models, their physiological and therapeutic relevance, and the insights they provide into the molecular mechanisms underlying CVDs. Additionally, we evaluate the advantages and limitations of these models, offering perspectives on how they can be utilized for the development of novel therapeutic strategies and drug testing in cardiovascular research.
Collapse
Affiliation(s)
- Abirami M. Padmanaban
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| |
Collapse
|
5
|
Fang W, Yu Z, Gao G, Yang M, Du X, Wang Y, Fu Q. Light-based 3D bioprinting technology applied to repair and regeneration of different tissues: A rational proposal for biomedical applications. Mater Today Bio 2024; 27:101135. [PMID: 39040222 PMCID: PMC11262185 DOI: 10.1016/j.mtbio.2024.101135] [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: 02/02/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
3D bioprinting technology, a subset of 3D printing technology, is currently witnessing widespread utilization in tissue repair and regeneration endeavors. In particular, light-based 3D bioprinting technology has garnered significant interest and favor. Central to its successful implementation lies the judicious selection of photosensitive polymers. Moreover, by fine-tuning parameters such as light irradiation time, choice of photoinitiators and crosslinkers, and their concentrations, the properties of the scaffolds can be tailored to suit the specific requirements of the targeted tissue repair sites. In this comprehensive review, we provide an overview of commonly utilized bio-inks suitable for light-based 3D bioprinting, delving into the distinctive characteristics of each material. Furthermore, we delineate strategies for bio-ink selection tailored to diverse repair locations, alongside methods for optimizing printing parameters. Ultimately, we present a coherent synthesis aimed at enhancing the practical application of light-based 3D bioprinting technology in tissue engineering, while also addressing current challenges and future prospects.
Collapse
Affiliation(s)
- Wenzhuo Fang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Zhenwei Yu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Guo Gao
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ming Yang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Xuan Du
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Qiang Fu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| |
Collapse
|
6
|
Florido MHC, Ziats NP. Endothelial dysfunction and cardiovascular diseases: The role of human induced pluripotent stem cells and tissue engineering. J Biomed Mater Res A 2024; 112:1286-1304. [PMID: 38230548 DOI: 10.1002/jbm.a.37669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Cardiovascular disease (CVD) remains to be the leading cause of death globally today and therefore the need for the development of novel therapies has become increasingly important in the cardiovascular field. The mechanism(s) behind the pathophysiology of CVD have been laboriously investigated in both stem cell and bioengineering laboratories. Scientific breakthroughs have paved the way to better mimic cell types of interest in recent years, with the ability to generate any cell type from reprogrammed human pluripotent stem cells. Mimicking the native extracellular matrix using both organic and inorganic biomaterials has allowed full organs to be recapitulated in vitro. In this paper, we will review techniques from both stem cell biology and bioengineering which have been fruitfully combined and have fueled advances in the cardiovascular disease field. We will provide a brief introduction to CVD, reviewing some of the recent studies as related to the role of endothelial cells and endothelial cell dysfunction. Recent advances and the techniques widely used in both bioengineering and stem cell biology will be discussed, providing a broad overview of the collaboration between these two fields and their overall impact on tissue engineering in the cardiovascular devices and implications for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mary H C Florido
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Nicholas P Ziats
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Biomedical Engineering and Anatomy, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Khodayari H, Khodayari S, Rezaee M, Rezaeiani S, Alipour Choshali M, Erfanian S, Muhammadnejad A, Nili F, Pourmehran Y, Pirjani R, Rajabi S, Aghdami N, Nebigil-Désaubry C, Wang K, Mahmoodzadeh H, Pahlavan S. Promotion of cardiac microtissue assembly within G-CSF-enriched collagen I-cardiogel hybrid hydrogel. Regen Biomater 2024; 11:rbae072. [PMID: 38974665 PMCID: PMC11226883 DOI: 10.1093/rb/rbae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Tissue engineering as an interdisciplinary field of biomedical sciences has raised many hopes in the treatment of cardiovascular diseases as well as development of in vitro three-dimensional (3D) cardiac models. This study aimed to engineer a cardiac microtissue using a natural hybrid hydrogel enriched by granulocyte colony-stimulating factor (G-CSF), a bone marrow-derived growth factor. Cardiac ECM hydrogel (Cardiogel: CG) was mixed with collagen type I (ColI) to form the hybrid hydrogel, which was tested for mechanical and biological properties. Three cell types (cardiac progenitor cells, endothelial cells and cardiac fibroblasts) were co-cultured in the G-CSF-enriched hybrid hydrogel to form a 3D microtissue. ColI markedly improved the mechanical properties of CG in the hybrid form with a ratio of 1:1. The hybrid hydrogel demonstrated acceptable biocompatibility and improved retention of encapsulated human foreskin fibroblasts. Co-culture of three cell types in G-CSF enriched hybrid hydrogel, resulted in a faster 3D structure shaping and a well-cellularized microtissue with higher angiogenesis compared to growth factor-free hybrid hydrogel (control). Immunostaining confirmed the presence of CD31+ tube-like structures as well as vimentin+ cardiac fibroblasts and cTNT+ human pluripotent stem cells-derived cardiomyocytes. Bioinformatics analysis of signaling pathways related to the G-CSF receptor in cardiovascular lineage cells, identified target molecules. The in silico-identified STAT3, as one of the major molecules involved in G-CSF signaling of cardiac tissue, was upregulated in G-CSF compared to control. The G-CSF-enriched hybrid hydrogel could be a promising candidate for cardiac tissue engineering, as it facilitates tissue formation and angiogenesis.
Collapse
Affiliation(s)
- Hamid Khodayari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 13145-871, Iran
| | - Saeed Khodayari
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Malihe Rezaee
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Siamak Rezaeiani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Mahmoud Alipour Choshali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Saiedeh Erfanian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Ahad Muhammadnejad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Fatemeh Nili
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Yasaman Pourmehran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 13145-871, Iran
| | - Reihaneh Pirjani
- Obstetrics and Gynecology Department, Arash Women’s Hospital, Tehran University of Medical Sciences, Tehran 1653915981, Iran
| | - Sarah Rajabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Naser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 19395-4644, Iran
| | - Canan Nebigil-Désaubry
- Institute National de le santé et de la recherce médicale, INSERM, University of Strasbourg, UMR 1260-Regenerative Nanomedicine, CRBS, Central of Research in biomedicine of Strasbourg, Strasbourg 90032, France
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Habibollah Mahmoodzadeh
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| |
Collapse
|
8
|
Bravo-Olín J, Martínez-Carreón SA, Francisco-Solano E, Lara AR, Beltran-Vargas NE. Analysis of the role of perfusion, mechanical, and electrical stimulation in bioreactors for cardiac tissue engineering. Bioprocess Biosyst Eng 2024; 47:767-839. [PMID: 38643271 DOI: 10.1007/s00449-024-03004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 03/13/2024] [Indexed: 04/22/2024]
Abstract
Since cardiovascular diseases (CVDs) are globally one of the leading causes of death, of which myocardial infarction (MI) can cause irreversible damage and decrease survivors' quality of life, novel therapeutics are needed. Current approaches such as organ transplantation do not fully restore cardiac function or are limited. As a valuable strategy, tissue engineering seeks to obtain constructs that resemble myocardial tissue, vessels, and heart valves using cells, biomaterials as scaffolds, biochemical and physical stimuli. The latter can be induced using a bioreactor mimicking the heart's physiological environment. An extensive review of bioreactors providing perfusion, mechanical and electrical stimulation, as well as the combination of them is provided. An analysis of the stimulations' mechanisms and modes that best suit cardiac construct culture is developed. Finally, we provide insights into bioreactor configuration and culture assessment properties that need to be elucidated for its clinical translation.
Collapse
Affiliation(s)
- Jorge Bravo-Olín
- Biological Engineering Undergraduate Program, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Sabina A Martínez-Carreón
- Biological Engineering Undergraduate Program, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Emmanuel Francisco-Solano
- Natural Science and Engineering Graduate Program, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Alvaro R Lara
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000, Aarhus, Denmark
| | - Nohra E Beltran-Vargas
- Process and Technology Department, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México.
| |
Collapse
|
9
|
Min S, Kim S, Sim WS, Choi YS, Joo H, Park JH, Lee SJ, Kim H, Lee MJ, Jeong I, Cui B, Jo SH, Kim JJ, Hong SB, Choi YJ, Ban K, Kim YG, Park JU, Lee HA, Park HJ, Cho SW. Versatile human cardiac tissues engineered with perfusable heart extracellular microenvironment for biomedical applications. Nat Commun 2024; 15:2564. [PMID: 38519491 PMCID: PMC10960018 DOI: 10.1038/s41467-024-46928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
Engineered human cardiac tissues have been utilized for various biomedical applications, including drug testing, disease modeling, and regenerative medicine. However, the applications of cardiac tissues derived from human pluripotent stem cells are often limited due to their immaturity and lack of functionality. Therefore, in this study, we establish a perfusable culture system based on in vivo-like heart microenvironments to improve human cardiac tissue fabrication. The integrated culture platform of a microfluidic chip and a three-dimensional heart extracellular matrix enhances human cardiac tissue development and their structural and functional maturation. These tissues are comprised of cardiovascular lineage cells, including cardiomyocytes and cardiac fibroblasts derived from human induced pluripotent stem cells, as well as vascular endothelial cells. The resultant macroscale human cardiac tissues exhibit improved efficacy in drug testing (small molecules with various levels of arrhythmia risk), disease modeling (Long QT Syndrome and cardiac fibrosis), and regenerative therapy (myocardial infarction treatment). Therefore, our culture system can serve as a highly effective tissue-engineering platform to provide human cardiac tissues for versatile biomedical applications.
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suran Kim
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Cellartgen, Seoul, 03722, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi Jeong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Baofang Cui
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seok Beom Hong
- Department of Thoracic and Cardiovascular Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 03312, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, 999077, Hong Kong
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Cellartgen, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
10
|
Gao H, Wang Z, Yang F, Wang X, Wang S, Zhang Q, Liu X, Sun Y, Kong J, Yao J. Graphene-integrated mesh electronics with converged multifunctionality for tracking multimodal excitation-contraction dynamics in cardiac microtissues. Nat Commun 2024; 15:2321. [PMID: 38485708 PMCID: PMC10940632 DOI: 10.1038/s41467-024-46636-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
Cardiac microtissues provide a promising platform for disease modeling and developmental studies, which require the close monitoring of the multimodal excitation-contraction dynamics. However, no existing assessing tool can track these multimodal dynamics across the live tissue. We develop a tissue-like mesh bioelectronic system to track these multimodal dynamics. The mesh system has tissue-level softness and cell-level dimensions to enable stable embedment in the tissue. It is integrated with an array of graphene sensors, which uniquely converges both bioelectrical and biomechanical sensing functionalities in one device. The system achieves stable tracking of the excitation-contraction dynamics across the tissue and throughout the developmental process, offering comprehensive assessments for tissue maturation, drug effects, and disease modeling. It holds the promise to provide more accurate quantification of the functional, developmental, and pathophysiological states in cardiac tissues, creating an instrumental tool for improving tissue engineering and studies.
Collapse
Affiliation(s)
- Hongyan Gao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Zhien Wang
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Xiaoyu Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Siqi Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Quan Zhang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Xiaomeng Liu
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Jing Kong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jun Yao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
11
|
Iwoń Z, Krogulec E, Kierlańczyk A, Baranowska P, Łopianiak I, Wojasiński M, Jastrzębska E. Improving rodents and humans cardiac cell maturity in vitrothrough polycaprolactone and polyurethane nanofibers. Biomed Mater 2024; 19:025031. [PMID: 38290152 DOI: 10.1088/1748-605x/ad240a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
Currently, numerous studies are conducted using nanofibers as a scaffold for culture cardiac cells; however, there still needs to be more research evaluating the impact of the physicochemical properties of polymer nanofibers on the structure and function of cardiac cells. We have studied how poly(ϵ-caprolactone) and polyurethane nanofibrous mats with different physicochemical properties influence the viability, morphology, orientation, and maturation of cardiac cells. For this purpose, the cells taken from different species were used. They were rat ventricular cardiomyoblasts (H9c2), mouse atrial cardiomyocytes (CMs) (HL-1), and human ventricular CMs. Based on the results, it can be concluded that cardiac cells cultured on nanofibers exhibit greater maturity in terms of orientation, morphology, and gene expression levels compared to cells cultured on polystyrene plates. Additionally, the physicochemical properties of nanofibers affecting the functionality of cardiac cells from different species and different parts of the heart were evaluated. These studies can support research on understanding and explaining mechanisms leading to cellular maturity present in the heart and the selection of nanofibers that will effectively help the maturation of CMs.
Collapse
Affiliation(s)
- Zuzanna Iwoń
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | | | - Aleksandra Kierlańczyk
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Patrycja Baranowska
- Centre for Advanced Materials and Technologies, CEZAMAT Warsaw University of Technology, Warsaw, Poland
| | - Iwona Łopianiak
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Michal Wojasiński
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- Centre for Advanced Materials and Technologies, CEZAMAT Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
12
|
Chen X, Liu S, Han M, Long M, Li T, Hu L, Wang L, Huang W, Wu Y. Engineering Cardiac Tissue for Advanced Heart-On-A-Chip Platforms. Adv Healthc Mater 2024; 13:e2301338. [PMID: 37471526 DOI: 10.1002/adhm.202301338] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Cardiovascular disease is a major cause of mortality worldwide, and current preclinical models including traditional animal models and 2D cell culture models have limitations in replicating human native heart physiology and response to drugs. Heart-on-a-chip (HoC) technology offers a promising solution by combining the advantages of cardiac tissue engineering and microfluidics to create in vitro 3D cardiac models, which can mimic key aspects of human microphysiological systems and provide controllable microenvironments. Herein, recent advances in HoC technologies are introduced, including engineered cardiac microtissue construction in vitro, microfluidic chip fabrication, microenvironmental stimulation, and real-time feedback systems. The development of cardiac tissue engineering methods is focused for 3D microtissue preparation, advanced strategies for HoC fabrication, and current applications of these platforms. Major challenges in HoC fabrication are discussed and the perspective on the potential for these platforms is provided to advance research and clinical applications.
Collapse
Affiliation(s)
- Xinyi Chen
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sitian Liu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingying Han
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Meng Long
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Li
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lanlan Hu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ling Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
13
|
Shang Y, Liu R, Gan J, Yang Y, Sun L. Construction of cardiac fibrosis for biomedical research. SMART MEDICINE 2023; 2:e20230020. [PMID: 39188350 PMCID: PMC11235890 DOI: 10.1002/smmd.20230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 08/28/2024]
Abstract
Cardiac remodeling is critical for effective tissue recuperation, nevertheless, excessive formation and deposition of extracellular matrix components can result in the onset of cardiac fibrosis. Despite the emergence of novel therapies, there are still no lifelong therapeutic solutions for this issue. Understanding the detrimental cardiac remodeling may aid in the development of innovative treatment strategies to prevent or reverse fibrotic alterations in the heart. Further combining the latest understanding of disease pathogenesis with cardiac tissue engineering has provided the conversion of basic laboratory studies into the therapy of cardiac fibrosis patients as an increasingly viable prospect. This review presents the current main mechanisms and the potential tissue engineering of cardiac fibrosis. Approaches using biomedical materials-based cardiac constructions are reviewed to consider key issues for simulating in vitro cardiac fibrosis, outlining a future perspective for preclinical applications.
Collapse
Affiliation(s)
- Yixuan Shang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yuzhi Yang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
14
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
15
|
Roshanravan N, Ghaffari S, Bastani S, Pahlavan S, Asghari S, Doustvandi MA, Jalilzadeh- Razin S, Dastouri M. Human cardiac organoids: A recent revolution in disease modeling and regenerative medicine. J Cardiovasc Thorac Res 2023; 15:68-72. [PMID: 37654821 PMCID: PMC10466470 DOI: 10.34172/jcvtr.2023.31830] [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: 04/04/2023] [Accepted: 06/10/2023] [Indexed: 09/02/2023] Open
Abstract
Three-dimensional (3D) myocardial tissues for studying human heart biology, physiology and pharmacology have recently received lots of attention. Organoids as 3D mini-organs are created from multiple cell types (i.e. induced pluripotent stem cells (iPSCs) or embryonic stem cells (ESCs)) with other supporting co-cultured cells such as endothelial cells or fibroblasts. Cardiac organoid culture technologies are bringing about significant advances in organ research and allows for the establishment of tissue regeneration and disease modeling. The present review provides an overview of the recent advances in human cardiac organoid platforms in disease biology and for cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Bastani
- Department of Immunology, Leiden University Medical Science, Leiden, Netherlands
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samira Asghari
- University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | | | - Sepideh Jalilzadeh- Razin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Dastouri
- Ankara University Biotechnology Institute and SISBIYOTEK Advanced Research Unit, Gumusdere Yerleskesi, Kecioren, Ankara, Turkey
| |
Collapse
|
16
|
Wolfe JT, He W, Kim MS, Liang HL, Shradhanjali A, Jurkiewicz H, Freudinger BP, Greene AS, LaDisa JF, Tayebi L, Mitchell ME, Tomita-Mitchell A, Tefft BJ. 3D-bioprinting of patient-derived cardiac tissue models for studying congenital heart disease. Front Cardiovasc Med 2023; 10:1162731. [PMID: 37293290 PMCID: PMC10247285 DOI: 10.3389/fcvm.2023.1162731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Congenital heart disease is the leading cause of death related to birth defects and affects 1 out of every 100 live births. Induced pluripotent stem cell technology has allowed for patient-derived cardiomyocytes to be studied in vitro. An approach to bioengineer these cells into a physiologically accurate cardiac tissue model is needed in order to study the disease and evaluate potential treatment strategies. Methods To accomplish this, we have developed a protocol to 3D-bioprint cardiac tissue constructs comprised of patient-derived cardiomyocytes within a hydrogel bioink based on laminin-521. Results Cardiomyocytes remained viable and demonstrated appropriate phenotype and function including spontaneous contraction. Contraction remained consistent during 30 days of culture based on displacement measurements. Furthermore, tissue constructs demonstrated progressive maturation based on sarcomere structure and gene expression analysis. Gene expression analysis also revealed enhanced maturation in 3D constructs compared to 2D cell culture. Discussion This combination of patient-derived cardiomyocytes and 3D-bioprinting represents a promising platform for studying congenital heart disease and evaluating individualized treatment strategies.
Collapse
Affiliation(s)
- Jayne T. Wolfe
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
| | - Wei He
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
| | - Min-Su Kim
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Huan-Ling Liang
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Akankshya Shradhanjali
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
| | - Hilda Jurkiewicz
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
| | | | | | - John F. LaDisa
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
- Department of Pediatrics - Section of Cardiology, Children’s Wisconsin, Milwaukee, WI, United States
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI, United States
| | - Michael E. Mitchell
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, United States
| | - Aoy Tomita-Mitchell
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brandon J. Tefft
- Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, WI, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
17
|
Belanger K, Koppes AN, Koppes RA. Impact of Non-Muscle Cells on Excitation-Contraction Coupling in the Heart and the Importance of In Vitro Models. Adv Biol (Weinh) 2023; 7:e2200117. [PMID: 36216583 DOI: 10.1002/adbi.202200117] [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: 04/20/2022] [Revised: 08/07/2022] [Indexed: 05/13/2023]
Abstract
Excitation-coupling (ECC) is paramount for coordinated contraction to maintain sufficient cardiac output. The study of ECC regulation has primarily been limited to cardiomyocytes (CMs), which conduct voltage waves via calcium fluxes from one cell to another, eliciting contraction of the atria followed by the ventricles. CMs rapidly transmit ionic flux via gap junction proteins, predominantly connexin 43. While the expression of connexin isoforms has been identified in each of the individual cell populations comprising the heart, the formation of gap junctions with nonmuscle cells (i.e., macrophages and Schwann cells) has gained new attention. Evaluating nonmuscle contributions to ECC in vivo or in situ remains difficult and necessitates the development of simple, yet biomimetic in vitro models to better understand and prevent physiological dysfunction. Standard 2D cell culture often consists of homogenous cell populations and lacks the dynamic mechanical environment of native tissue, confounding the phenotypic and proteomic makeup of these highly mechanosensitive cell populations in prolonged culture conditions. This review will highlight the recent developments and the importance of new microphysiological systems to better understand the complex regulation of ECC in cardiac tissue.
Collapse
Affiliation(s)
- Kirstie Belanger
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
18
|
Abstract
Heart disease is a significant burden on global health care systems and is a leading cause of death each year. To improve our understanding of heart disease, high quality disease models are needed. These will facilitate the discovery and development of new treatments for heart disease. Traditionally, researchers have relied on 2D monolayer systems or animal models of heart disease to elucidate pathophysiology and drug responses. Heart-on-a-chip (HOC) technology is an emerging field where cardiomyocytes among other cell types in the heart can be used to generate functional, beating cardiac microtissues that recapitulate many features of the human heart. HOC models are showing great promise as disease modeling platforms and are poised to serve as important tools in the drug development pipeline. By leveraging advances in human pluripotent stem cell-derived cardiomyocyte biology and microfabrication technology, diseased HOCs are highly tuneable and can be generated via different approaches such as: using cells with defined genetic backgrounds (patient-derived cells), adding small molecules, modifying the cells' environment, altering cell ratio/composition of microtissues, among others. HOCs have been used to faithfully model aspects of arrhythmia, fibrosis, infection, cardiomyopathies, and ischemia, to name a few. In this review, we highlight recent advances in disease modeling using HOC systems, describing instances where these models outperformed other models in terms of reproducing disease phenotypes and/or led to drug development.
Collapse
Affiliation(s)
- Omar Mourad
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Ryan Yee
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Mengyuan Li
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Ajmera Transplant Center (S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
- Department of Laboratory Medicine and Pathobiology (S.S.N.), University of Toronto, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence (S.S.N.), University of Toronto, Canada
| |
Collapse
|
19
|
A Three-Dimensional Engineered Cardiac In Vitro Model: Controlled Alignment of Cardiomyocytes in 3D Microphysiological Systems. Cells 2023; 12:cells12040576. [PMID: 36831243 PMCID: PMC9954012 DOI: 10.3390/cells12040576] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Cardiomyocyte alignment in myocardium tissue plays a significant role in the physiological, electrical, and mechanical functions of the myocardium. It remains, however, difficult to align cardiac cells in a 3D in vitro heart model. This paper proposes a simple method to align cells using microfabricated Polydimethylsiloxane (PDMS) grooves with large dimensions (of up to 350 µm in width), similar to the dimensions of trabeculae carneae, the smallest functional unit of the myocardium. Two cell groups were used in this work; first, H9c2 cells in combination with Nor10 cells for proof of concept, and second, neonatal cardiac cells to investigate the functionality of the 3D model. This model compared the patterned and nonpatterned 3D constructs, as well as the 2D cell cultures, with and without patterns. In addition to alignment, we assessed the functionality of our proposed 3D model by comparing beating rates between aligned and non-aligned structures. In order to assess the practicality of the model, the 3D aligned structures should be demonstrated to be detachable and alignable. This evaluation is crucial to the use of this 3D functional model in future studies related to drug screening, building blocks for tissue engineering, and as a heart-on-chip by integrating microfluidics.
Collapse
|
20
|
Takahashi H, Wakayama H, Nagase K, Shimizu T. Engineered Human Muscle Tissue from Multilayered Aligned Myofiber Sheets for Studies of Muscle Physiology and Predicting Drug Response. SMALL METHODS 2023; 7:e2200849. [PMID: 36562139 DOI: 10.1002/smtd.202200849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/22/2022] [Indexed: 06/17/2023]
Abstract
In preclinical drug testing, human muscle tissue models are critical to understanding the complex physiology, including drug effects in the human body. This study reports that a multilayering approach to cell sheet-based engineering produces an engineered human muscle tissue with sufficient contractile force suitable for measurement. A thermoresponsive micropatterned substrate regulates the biomimetic alignment of myofiber structures enabling the harvest of the aligned myofibers as a single cell sheet. The functional muscle tissue is produced by layering multiple myofiber sheets on a fibrin-based gel. This gel environment promotes myofiber maturation, provides the tissue an elastic platform for contraction, and allows the attachment of a measurement device. Since this multilayering approach is effective in enhancing the contractile ability of the muscle tissue, this muscle tissue generates a significantly high contractile force that can be measured quantitatively. The multilayered muscle tissue shows unidirectional contraction from electrical and chemical stimulation. In addition, their physiological responses to representative drugs can be determined quantitatively in real time by changes in contractile force and fatigue resistance. These physiological properties indicate that the engineered muscle tissue can become a promising tissue model for preclinical in vitro studies in muscle physiology and drug discovery.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Haruno Wakayama
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| |
Collapse
|
21
|
Spedicati M, Ruocco G, Zoso A, Mortati L, Lapini A, Delledonne A, Divieto C, Romano V, Castaldo C, Di Meglio F, Nurzynska D, Carmagnola I, Chiono V. Biomimetic design of bioartificial scaffolds for the in vitro modelling of human cardiac fibrosis. Front Bioeng Biotechnol 2022; 10:983872. [PMID: 36507252 PMCID: PMC9731288 DOI: 10.3389/fbioe.2022.983872] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022] Open
Abstract
In vitro models of pathological cardiac tissue have attracted interest as predictive platforms for preclinical validation of therapies. However, models reproducing specific pathological features, such as cardiac fibrosis size (i.e., thickness and width) and stage of development are missing. This research was aimed at engineering 2D and 3D models of early-stage post-infarct fibrotic tissue (i.e., characterized by non-aligned tissue organization) on bioartificial scaffolds with biomimetic composition, design, and surface stiffness. 2D scaffolds with random nanofibrous structure and 3D scaffolds with 150 µm square-meshed architecture were fabricated from polycaprolactone, surface-grafted with gelatin by mussel-inspired approach and coated with cardiac extracellular matrix (ECM) by 3 weeks culture of human cardiac fibroblasts. Scaffold physicochemical properties were thoroughly investigated. AFM analysis of scaffolds in wet state, before cell culture, confirmed their close surface stiffness to human cardiac fibrotic tissue. Following 3 weeks culture, biomimetic biophysical and biochemical scaffold properties triggered the activation of myofibroblast phenotype. Upon decellularization, immunostaining, SEM and two-photon excitation fluorescence microscopy showed homogeneous decoration of both 2D and 3D scaffolds with cardiac ECM. The versatility of the approach was demonstrated by culturing ventricular or atrial cardiac fibroblasts on scaffolds, thus suggesting the possibility to use the same scaffold platforms to model both ventricular and atrial cardiac fibrosis. In the future, herein developed in vitro models of cardiac fibrotic tissue, reproducing specific pathological features, will be exploited for a fine preclinical tuning of therapies.
Collapse
Affiliation(s)
- Mattia Spedicati
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- POLITO Biomedlab, Politecnico di Torino, Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Gerardina Ruocco
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- POLITO Biomedlab, Politecnico di Torino, Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Alice Zoso
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- POLITO Biomedlab, Politecnico di Torino, Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Leonardo Mortati
- Istituto Nazionale di Ricerca Metrologica (INRIM), Torino, Italy
| | - Andrea Lapini
- Istituto Nazionale di Ricerca Metrologica (INRIM), Torino, Italy
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parma, Italy
| | - Andrea Delledonne
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parma, Italy
| | - Carla Divieto
- Istituto Nazionale di Ricerca Metrologica (INRIM), Torino, Italy
| | - Veronica Romano
- Department of Public Health, University of Naples “Federico II”, Napoli, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples “Federico II”, Napoli, Italy
| | - Franca Di Meglio
- Department of Public Health, University of Naples “Federico II”, Napoli, Italy
| | - Daria Nurzynska
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- POLITO Biomedlab, Politecnico di Torino, Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- POLITO Biomedlab, Politecnico di Torino, Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| |
Collapse
|
22
|
Liang PY, Chang Y, Jin G, Lian X, Bao X. Wnt signaling directs human pluripotent stem cells into vascularized cardiac organoids with chamber-like structures. Front Bioeng Biotechnol 2022; 10:1059243. [PMID: 36466327 PMCID: PMC9715615 DOI: 10.3389/fbioe.2022.1059243] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/04/2022] [Indexed: 10/28/2023] Open
Abstract
Heart diseases are leading cause of death around the world. Given their unique capacity to self-renew and differentiate into all types of somatic cells, human pluripotent stem cells (hPSCs) hold great promise for heart disease modeling and cardiotoxic drug screening. hPSC-derived cardiac organoids are emerging biomimetic models for studying heart development and cardiovascular diseases, but it remains challenging to make mature organoids with a native-like structure in vitro. In this study, temporal modulation of Wnt signaling pathway co-differentiated hPSCs into beating cardiomyocytes and cardiac endothelial-like cells in 3D organoids, resulting in cardiac endothelial-bounded chamber formation. These chambered cardiac organoids exhibited more mature membrane potential compared to cardiac organoids composed of only cardiomyocytes. Furthermore, a better response to toxic drugs was observed in chamber-contained cardiac organoids. In summary, spatiotemporal signaling pathway modulation may lead to more mature cardiac organoids for studying cardiovascular development and diseases.
Collapse
Affiliation(s)
- Po-Yu Liang
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - Yun Chang
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Gyuhyung Jin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Xiaojun Lian
- Department of Biomedical Engineering, The Huck Institutes of the Life Sciences, Department of Biology, The Pennsylvania State University, University Park, PA, United States
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| |
Collapse
|
23
|
Jafari A, Ajji Z, Mousavi A, Naghieh S, Bencherif SA, Savoji H. Latest Advances in 3D Bioprinting of Cardiac Tissues. ADVANCED MATERIALS TECHNOLOGIES 2022; 7:2101636. [PMID: 38044954 PMCID: PMC10691862 DOI: 10.1002/admt.202101636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Indexed: 12/05/2023]
Abstract
Cardiovascular diseases (CVDs) are known as the major cause of death worldwide. In spite of tremendous advancements in medical therapy, the gold standard for CVD treatment is still transplantation. Tissue engineering, on the other hand, has emerged as a pioneering field of study with promising results in tissue regeneration using cells, biological cues, and scaffolds. Three-dimensional (3D) bioprinting is a rapidly growing technique in tissue engineering because of its ability to create complex scaffold structures, encapsulate cells, and perform these tasks with precision. More recently, 3D bioprinting has made its debut in cardiac tissue engineering, and scientists are investigating this technique for development of new strategies for cardiac tissue regeneration. In this review, the fundamentals of cardiac tissue biology, available 3D bioprinting techniques and bioinks, and cells implemented for cardiac regeneration are briefly summarized and presented. Afterwards, the pioneering and state-of-the-art works that have utilized 3D bioprinting for cardiac tissue engineering are thoroughly reviewed. Finally, regulatory pathways and their contemporary limitations and challenges for clinical translation are discussed.
Collapse
Affiliation(s)
- Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Zineb Ajji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Ali Mousavi
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
- Sorbonne University, UTC CNRS UMR 7338, Biomechanics and Bioengineering (BMBI), University of Technology of Compiègne, 60203 Compiègne, France
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02128, United States
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| |
Collapse
|
24
|
Kałużna E, Nadel A, Zimna A, Rozwadowska N, Kolanowski T. Modeling the human heart ex vivo-current possibilities and strive for future applications. J Tissue Eng Regen Med 2022; 16:853-874. [PMID: 35748158 PMCID: PMC9796015 DOI: 10.1002/term.3335] [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: 11/16/2021] [Revised: 04/20/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
Collapse
Affiliation(s)
- Ewelina Kałużna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Nadel
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Zimna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | | | | |
Collapse
|
25
|
Csöbönyeiová M, Beerová N, Klein M, Debreová-Čeháková M, Danišovič Ľ. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci 2022; 23:10314. [PMID: 36142245 PMCID: PMC9499607 DOI: 10.3390/ijms231810314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Because of cardiomyocyte death or dysfunction frequently caused by myocardial infarction (MI), heart failure is a leading cause of morbidity and mortality in modern society. Paradoxically, only limited and non-curative therapies for heart failure or MI are currently available. As a result, over the past two decades research has focused on developing cell-based approaches promoting the regeneration of infarcted tissue. Cell-based therapies for myocardial regeneration include powerful candidates, such as multipotent stem cells (mesenchymal stem cells (MSCs), bone-marrow-derived stem cells, endothelial progenitor cells, and hematopoietic stem cells) and induced pluripotent stem cells (iPSCs). These possess unique properties, such as potency to differentiate into desired cell types, proliferation capacity, and patient specificity. Preclinical and clinical studies have demonstrated modest improvement in the myocardial regeneration and reduced infarcted areas upon transplantation of pluripotent or multipotent stem cells. Another cell population that need to be considered as a potential source for cardiac regeneration are telocytes found in different organs, including the heart. Their therapeutic effect has been studied in various heart pathologies, such as MI, arrhythmias, or atrial amyloidosis. The most recent cell-free therapeutic tool relies on the cardioprotective effect of complex cargo carried by small membrane-bound vesicles-exosomes-released from stem cells via exocytosis. The MSC/iPSC-derived exosomes could be considered a novel exosome-based therapy for cardiovascular diseases thanks to their unique content. There are also other cell-free approaches, e.g., gene therapy, or acellular cardiac patches. Therefore, our review provides the most recent insights into the novel strategies for myocardial repair based on the regenerative potential of different cell types and cell-free approaches.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Nikoleta Beerová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Martin Klein
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Debreová-Čeháková
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
26
|
Zhang Y, Mu W, Zhang Y, He X, Wang Y, Ma H, Zhu T, Li A, Hou Q, Yang W, Ding Y, Ramakrishna S, Li H. Recent Advances in Cardiac Patches: Materials, Preparations, and Properties. ACS Biomater Sci Eng 2022; 8:3659-3675. [PMID: 36037313 DOI: 10.1021/acsbiomaterials.2c00348] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cardiac patches are biomaterials that can be used for transplantation and repair of damaged myocardium by combining seed cells with the ability to form cardiomyocytes and suitable scaffold materials. On the one hand, they provide temporary support to the infarcted area, and on the other hand, they repair the damaged myocardium by delivering cells or bioactive factors to integrate with the host, which have gradually become a hot research topic in recent years. This paper summarizes the structural properties of natural myocardium and reviews the recent research progress of cardiac patches, including the seed cells and scaffold materials used in patch preparation, as well as the main methods of scaffold preparation and the structure properties of various scaffolds. In addition, a comprehensive analysis of the problems faced in the clinical implementation of cardiac patches is presented. Finally, we look forward to the development of cardiac patches and point out that precisely tunable anisotropic tissue engineering scaffolds close to natural myocardial tissue will become an important direction for future research.
Collapse
Affiliation(s)
- Yi Zhang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.,Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wenying Mu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100000, China
| | - Yanping Zhang
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, DK-8000, Denmark
| | - Xuetao He
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yiming Wang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Hongyu Ma
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Tianyang Zhu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Aoyuan Li
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qinzheng Hou
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Weimin Yang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.,Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China.,State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yumei Ding
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Seeram Ramakrishna
- Center for Nanofibers & Nanotechnology, National University of Singapore, Singapore, 119077, Singapore
| | - Haoyi Li
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.,Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China.,State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
27
|
Kowalczewski A, Sakolish C, Hoang P, Liu X, Jacquir S, Rusyn I, Ma Z. Integrating nonlinear analysis and machine learning for human induced pluripotent stem cell-based drug cardiotoxicity testing. J Tissue Eng Regen Med 2022; 16:732-743. [PMID: 35621199 PMCID: PMC9719611 DOI: 10.1002/term.3325] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 01/16/2023]
Abstract
Utilizing recent advances in human induced pluripotent stem cell (hiPSC) technology, nonlinear analysis and machine learning we can create novel tools to evaluate drug-induced cardiotoxicity on human cardiomyocytes. With cardiovascular disease remaining the leading cause of death globally it has become imperative to create effective and modern tools to test the efficacy and toxicity of drugs to combat heart disease. The calcium transient signals recorded from hiPSC-derived cardiomyocytes (hiPSC-CMs) are highly complex and dynamic with great degrees of response characteristics to various drug treatments. However, traditional linear methods often fail to capture the subtle variation in these signals generated by hiPSC-CMs. In this work, we integrated nonlinear analysis, dimensionality reduction techniques and machine learning algorithms for better classifying the contractile signals from hiPSC-CMs in response to different drug exposure. By utilizing extracted parameters from a commercially available high-throughput testing platform, we were able to distinguish the groups with drug treatment from baseline controls, determine the drug exposure relative to IC50 values, and classify the drugs by its unique cardiac responses. By incorporating nonlinear parameters computed by phase space reconstruction, we were able to improve our machine learning algorithm's ability to predict cardiotoxic levels and drug classifications. We also visualized the effects of drug treatment and dosages with dimensionality reduction techniques, t-distributed stochastic neighbor embedding (t-SNE). We have shown that integration of nonlinear analysis and artificial intelligence has proven to be a powerful tool for analyzing cardiotoxicity and classifying toxic compounds through their mechanistic action.
Collapse
Affiliation(s)
- Andrew Kowalczewski
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse NY, USA,BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse NY, USA
| | - Courtney Sakolish
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Plansky Hoang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse NY, USA,BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse NY, USA
| | - Xiyuan Liu
- Department of Mechanical & Aerospace Engineering, Syracuse University, Syracuse NY, USA
| | - Sabir Jacquir
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, Gif-sur-Yvette, France
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse NY, USA,BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse NY, USA,Corresponding author Zhen Ma, PhD. Syracuse University ()
| |
Collapse
|
28
|
Dou W, Malhi M, Cui T, Wang M, Wang T, Shan G, Law J, Gong Z, Plakhotnik J, Filleter T, Li R, Simmons CA, Maynes JT, Sun Y. A Carbon-Based Biosensing Platform for Simultaneously Measuring the Contraction and Electrophysiology of iPSC-Cardiomyocyte Monolayers. ACS NANO 2022; 16:11278-11290. [PMID: 35715006 DOI: 10.1021/acsnano.2c04676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Heart beating is triggered by the generation and propagation of action potentials through the myocardium, resulting in the synchronous contraction of cardiomyocytes. This process highlights the importance of electrical and mechanical coordination in organ function. Investigating the pathogenesis of heart diseases and potential therapeutic actions in vitro requires biosensing technologies which allow for long-term and simultaneous measurement of the contractility and electrophysiology of cardiomyocytes. However, the adoption of current biosensing approaches for functional measurement of in vitro cardiac models is hampered by low sensitivity, difficulties in achieving multifunctional detection, and costly manufacturing processes. Leveraging carbon-based nanomaterials, we developed a biosensing platform that is capable of performing on-chip and simultaneous measurement of contractility and electrophysiology of human induced pluripotent stem-cell-derived cardiomyocyte (iPSC-CM) monolayers. This platform integrates with a flexible thin-film cantilever embedded with a carbon black (CB)-PDMS strain sensor for high-sensitivity contraction measurement and four pure carbon nanotube (CNT) electrodes for the detection of extracellular field potentials with low electrode impedance. Cardiac functional properties including contractile stress, beating rate, beating rhythm, and extracellular field potential were evaluated to quantify iPSC-CM responses to common cardiotropic agents. In addition, an in vitro model of drug-induced cardiac arrhythmia was established to further validate the platform for disease modeling and drug testing.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Teng Cui
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Minyao Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, M5G 1L7, Canada
| | - Tiancong Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Zheyuan Gong
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Julia Plakhotnik
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Tobin Filleter
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Renke Li
- Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, M5G 1L7, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, M5G 1M1, Canada
| | - Jason T Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
- Department of Computer Science, University of Toronto, Toronto, M5T 3A1, Canada
| |
Collapse
|
29
|
Malone A, Gallagher S, Saidi J, Rizq G, O'Dowd E, Vallence D, Hameed A. In vitro benchtop mock circulatory loop for heart failure with preserved ejection fraction emulation. Front Cardiovasc Med 2022; 9:910120. [PMID: 35935659 PMCID: PMC9353029 DOI: 10.3389/fcvm.2022.910120] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/30/2022] [Indexed: 01/08/2023] Open
Abstract
In this work, a novel mock circulatory loop (MCL) is presented that is capable of simulating both healthy cardiac function and Heart Failure with preserved Ejection Fraction (HFpEF). This MCL differs from others presented in the literature as it features two independently actuated heart chambers, representing the left atrium and the left ventricle. This is an important improvement over other designs as it allows for potential HFpEF treatments to be examined, not just in relation to their effect on the left ventricle but also on the left atrium. The aim of this work was to show that novel MCL designs could be developed to allow for testing of new mechanical circulatory support devices for the treatment of HFpEF. Two loop configurations are presented, one featuring hard PVC cylindrical chambers and one that features soft silicone chambers which are anatomically analogous to the native heart. We show that both MCLs are capable of simulating the onset of HFpEF with a sustained increase in diastolic pressure of 62.03% and a sustained decrease in end diastolic volume (EDV) of 14.24%.
Collapse
Affiliation(s)
- Andrew Malone
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, The Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
| | - Sean Gallagher
- Medical Device Design, National College of Art and Design, Dublin, Ireland
| | - Jemil Saidi
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, The Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
| | - Gina Rizq
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, The Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
- School of Medicine, The Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
| | - Enda O'Dowd
- Medical Device Design, National College of Art and Design, Dublin, Ireland
| | - Derek Vallence
- Medical Device Design, National College of Art and Design, Dublin, Ireland
| | - Aamir Hameed
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, The Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Li Y, Ye Z, Zhang J, Zhao Y, Zhu T, Song J, Xu F, Li F. In Situ and Quantitative Monitoring of Cardiac Tissues Using Programmable Scanning Electrochemical Microscopy. Anal Chem 2022; 94:10515-10523. [PMID: 35822575 DOI: 10.1021/acs.analchem.2c01919] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vitro cardiac tissue model holds great potential as a powerful platform for drug screening. Respiratory activity, contraction frequency, and extracellular H2O2 levels are the three key parameters for determining the physiological functions of cardiac tissues, which are technically challenging to be monitored in an in situ and quantitative manner. Herein, we constructed an in vitro cardiac tissue model on polyacrylamide gels and applied a pulsatile electrical field to promote the maturation of the cardiac tissue. Then, we built a scanning electrochemical microscopy (SECM) platform with programmable pulse potentials to in situ characterize the dynamic changes in the respiratory activity, contraction frequency, and extracellular H2O2 level of cardiac tissues under both normal physiological and drug (isoproterenol and propranolol) treatment conditions using oxygen, ferrocenecarboxylic acid (FcCOOH), and H2O2 as the corresponding redox mediators. The SECM results showed that isoproterenol treatment induced enhanced oxygen consumption, accelerated contractile frequency, and increased released H2O2 level, while propranolol treatment induced dynamically decreased oxygen consumption and contractile frequency and no obvious change in H2O2 levels, suggesting the effects of activation and inhibition of β-adrenoceptor on the metabolic and electrophysiological activities of cardiac tissues. Our work realizes the in situ and quantitative monitoring of respiratory activity, contraction frequency, and secreted H2O2 level of living cardiac tissues using SECM for the first time. The programmable SECM methodology can also be used to real-time and quantitatively monitor electrochemical and electrophysiological parameters of cardiac tissues for future drug screening studies.
Collapse
Affiliation(s)
- Yabei Li
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhaoyang Ye
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Junjie Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yuxiang Zhao
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Tong Zhu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Department of Cardiovasology, Xidian Group Hospital, Xi'an, Shaanxi Province 710077, P. R. China
| | - Jingjing Song
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
31
|
Mousavi A, Stefanek E, Jafari A, Ajji Z, Naghieh S, Akbari M, Savoji H. Tissue-engineered heart chambers as a platform technology for drug discovery and disease modeling. BIOMATERIALS ADVANCES 2022; 138:212916. [PMID: 35913255 DOI: 10.1016/j.bioadv.2022.212916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Current drug screening approaches are incapable of fully detecting and characterizing drug effectiveness and toxicity of human cardiomyocytes. The pharmaceutical industry uses mathematical models, cell lines, and in vivo models. Many promising drugs are abandoned early in development, and some cardiotoxic drugs reach humans leading to drug recalls. Therefore, there is an unmet need to have more reliable and predictive tools for drug discovery and screening applications. Biofabrication of functional cardiac tissues holds great promise for developing a faithful 3D in vitro disease model, optimizing drug screening efficiencies enabling precision medicine. Different fabrication techniques including molding, pull spinning and 3D bioprinting were used to develop tissue-engineered heart chambers. The big challenge is to effectively organize cells into tissue with structural and physiological features resembling native tissues. Some advancements have been made in engineering miniaturized heart chambers that resemble a living pump for drug screening and disease modeling applications. Here, we review the currently developed tissue-engineered heart chambers and discuss challenges and prospects.
Collapse
Affiliation(s)
- Ali Mousavi
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Evan Stefanek
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Zineb Ajji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Mohsen Akbari
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada; Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada.
| |
Collapse
|
32
|
Zhang Q, Bei HP, Zhao M, Dong Z, Zhao X. Shedding light on 3D printing: Printing photo-crosslinkable constructs for tissue engineering. Biomaterials 2022; 286:121566. [DOI: 10.1016/j.biomaterials.2022.121566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/25/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022]
|
33
|
Cecen B, Bal-Ozturk A, Yasayan G, Alarcin E, Kocak P, Tutar R, Kozaci LD, Shin SR, Miri AK. Selection of natural biomaterials for micro-tissue and organ-on-chip models. J Biomed Mater Res A 2022; 110:1147-1165. [PMID: 35102687 PMCID: PMC10700148 DOI: 10.1002/jbm.a.37353] [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: 09/06/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
The desired organ in micro-tissue models of organ-on-a-chip (OoC) devices dictates the optimum biomaterials, divided into natural and synthetic biomaterials. They can resemble biological tissues' biological functions and architectures by constructing bioactivity of macromolecules, cells, nanoparticles, and other biological agents. The inclusion of such components in OoCs allows them having biological processes, such as basic biorecognition, enzymatic cleavage, and regulated drug release. In this report, we review natural-based biomaterials that are used in OoCs and their main characteristics. We address the preparation, modification, and characterization methods of natural-based biomaterials and summarize recent reports on their applications in the design and fabrication of micro-tissue models. This article will help bioengineers select the proper biomaterials based on developing new technologies to meet clinical expectations and improve patient outcomes fusing disease modeling.
Collapse
Affiliation(s)
- Berivan Cecen
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Ayca Bal-Ozturk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey
| | - Gokcen Yasayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Polen Kocak
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Leyla Didem Kozaci
- Faculty of Medicine, Department of Medical Biochemistry, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, Massachusetts, USA
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
34
|
Morsink M, Severino P, Luna-Ceron E, Hussain MA, Sobahi N, Shin SR. Effects of electrically conductive nano-biomaterials on regulating cardiomyocyte behavior for cardiac repair and regeneration. Acta Biomater 2022; 139:141-156. [PMID: 34818579 PMCID: PMC11041526 DOI: 10.1016/j.actbio.2021.11.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) represents one of the most prevalent cardiovascular diseases, with a highly relevant and impactful role in public health. Despite the therapeutic advances of the last decades, MI still begets extensive death rates around the world. The pathophysiology of the disease correlates with cardiomyocyte necrosis, caused by an imbalance in the demand of oxygen to cardiac tissues, resulting from obstruction of the coronary flow. To alleviate the severe effects of MI, the use of various biomaterials exhibit vast potential in cardiac repair and regeneration, acting as native extracellular matrices. These hydrogels have been combined with nano sized or functional materials which possess unique electrical, mechanical, and topographical properties that play important roles in regulating phenotypes and the contractile function of cardiomyocytes even in adverse microenvironments. These nano-biomaterials' differential properties have led to substantial healing on in vivo cardiac injury models by promoting fibrotic scar reduction, hemodynamic function preservation, and benign cardiac remodeling. In this review, we discuss the interplay of the unique physical properties of electrically conductive nano-biomaterials, are able to manipulate the phenotypes and the electrophysiological behavior of cardiomyocytes in vitro, and can enhance heart regeneration in vivo. Consequently, the understanding of the decisive roles of the nano-biomaterials discussed in this review could be useful for designing novel nano-biomaterials in future research for cardiac tissue engineering and regeneration. STATEMENT OF SIGNIFICANCE: This study introduced and deciphered the understanding of the role of multimodal cues in recent advances of electrically conductive nano-biomaterials on cardiac tissue engineering. Compared with other review papers, which mainly describe these studies based on various types of electrically conductive nano-biomaterials, in this review paper we mainly discussed the interplay of the unique physical properties (electrical conductivity, mechanical properties, and topography) of electrically conductive nano-biomaterials, which would allow them to manipulate phenotypes and the electrophysiological behavior of cardiomyocytes in vitro and to enhance heart regeneration in vivo. Consequently, understanding the decisive roles of the nano-biomaterials discussed in the review could help design novel nano-biomaterials in future research for cardiac tissue engineering and regeneration.
Collapse
Affiliation(s)
- Margaretha Morsink
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, United States of America; Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, Netherlands; Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, Netherlands
| | - Patrícia Severino
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, United States of America; University of Tiradentes (Unit), Biotechnological Postgraduate Program. Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil; Institute of Technology and Research (ITP), Nanomedicine and Nanotechnology Laboratory (LNMed), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil; Tiradentes Institute, 150 Mt Vernon St, Dorchester, MA 02125, United States of America
| | - Eder Luna-Ceron
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, United States of America
| | - Mohammad A Hussain
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Nebras Sobahi
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, United States of America.
| |
Collapse
|
35
|
Xiang Y, Miller K, Guan J, Kiratitanaporn W, Tang M, Chen S. 3D bioprinting of complex tissues in vitro: state-of-the-art and future perspectives. Arch Toxicol 2022; 96:691-710. [PMID: 35006284 PMCID: PMC8850226 DOI: 10.1007/s00204-021-03212-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The pharmacology and toxicology of a broad variety of therapies and chemicals have significantly improved with the aid of the increasing in vitro models of complex human tissues. Offering versatile and precise control over the cell population, extracellular matrix (ECM) deposition, dynamic microenvironment, and sophisticated microarchitecture, which is desired for the in vitro modeling of complex tissues, 3D bio-printing is a rapidly growing technology to be employed in the field. In this review, we will discuss the recent advancement of printing techniques and bio-ink sources, which have been spurred on by the increasing demand for modeling tactics and have facilitated the development of the refined tissue models as well as the modeling strategies, followed by a state-of-the-art update on the specialized work on cancer, heart, muscle and liver. In the end, the toxicological modeling strategies, substantial challenges, and future perspectives for 3D printed tissue models were explored.
Collapse
Affiliation(s)
- Yi Xiang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Kathleen Miller
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA
| | | | - Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, USA.
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA.
| |
Collapse
|
36
|
Fang Y, Sun W, Zhang T, Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: A review. Biomaterials 2021; 280:121298. [PMID: 34864451 DOI: 10.1016/j.biomaterials.2021.121298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The field of cardiac tissue engineering has advanced over the past decades; however, most research progress has been limited to engineered cardiac tissues (ECTs) at the microscale with minimal geometrical complexities such as 3D strips and patches. Although microscale ECTs are advantageous for drug screening applications because of their high-throughput and standardization characteristics, they have limited translational applications in heart repair and the in vitro modeling of cardiac function and diseases. Recently, researchers have made various attempts to construct engineered cardiac pumps (ECPs) such as chambered ventricles, recapitulating the geometrical complexity of the native heart. The transition from microscale ECTs to ECPs at a translatable scale would greatly accelerate their translational applications; however, researchers are confronted with several major hurdles, including geometrical reconstruction, vascularization, and functional maturation. Therefore, the objective of this paper is to review the recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps. We first review the bioengineering approaches to fabricate ECPs, and then emphasize the unmatched potential of 3D bioprinting techniques. We highlight key advances in bioprinting strategies with high cell density as researchers have begun to realize the critical role that the cell density of non-proliferative cardiomyocytes plays in the cell-cell interaction and functional contracting performance. We summarize the current approaches to engineering vasculatures both at micro- and meso-scales, crucial for the survival of thick cardiac tissues and ECPs. We showcase a variety of strategies developed to enable the functional maturation of cardiac tissues, mimicking the in vivo environment during cardiac development. By highlighting state-of-the-art research, this review offers personal perspectives on future opportunities and trends that may bring us closer to the promise of functional ECPs.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| |
Collapse
|
37
|
Pien N, Pezzoli D, Van Hoorick J, Copes F, Vansteenland M, Albu M, De Meulenaer B, Mantovani D, Van Vlierberghe S, Dubruel P. Development of photo-crosslinkable collagen hydrogel building blocks for vascular tissue engineering applications: A superior alternative to methacrylated gelatin? MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112460. [PMID: 34702535 DOI: 10.1016/j.msec.2021.112460] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022]
Abstract
The present work targets the development of collagen-based hydrogel precursors, functionalized with photo-crosslinkable methacrylamide moieties (COL-MA), for vascular tissue engineering (vTE) applications. The developed materials were physico-chemically characterized in terms of crosslinking kinetics, degree of modification/conversion, swelling behavior, mechanical properties and in vitro cytocompatibility. The collagen derivatives were benchmarked to methacrylamide-modified gelatin (GEL-MA), due to its proven track record in the field of tissue engineering. To the best of our knowledge, this is the first paper in its kind comparing these two methacrylated biopolymers for vTE applications. For both gelatin and collagen, two derivatives with varying degrees of substitutions (DS) were developed by altering the added amount of methacrylic anhydride (MeAnH). This led to photo-crosslinkable derivatives with a DS of 74 and 96% for collagen, and a DS of 73 and 99% for gelatin. The developed derivatives showed high gel fractions (i.e. 74% and 84%, for the gelatin derivatives; 87 and 83%, for the collagen derivatives) and an excellent crosslinking efficiency. Furthermore, the results indicated that the functionalization of collagen led to hydrogels with tunable mechanical properties (i.e. storage moduli of [4.8-9.4 kPa] for the developed COL-MAs versus [3.9-8.4 kPa] for the developed GEL-MAs) along with superior cell-biomaterial interactions when compared to GEL-MA. Moreover, the developed photo-crosslinkable collagens showed superior mechanical properties compared to extracted native collagen. Therefore, the developed photo-crosslinkable collagens demonstrate great potential as biomaterials for vTE applications.
Collapse
Affiliation(s)
- Nele Pien
- Polymer Chemistry & Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281 S4bis, 9000 Gent, Belgium; Laboratory for Biomaterials and Bioengineering, CRC-I, Laval University, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Daniele Pezzoli
- Laboratory for Biomaterials and Bioengineering, CRC-I, Laval University, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Jasper Van Hoorick
- Polymer Chemistry & Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281 S4bis, 9000 Gent, Belgium
| | - Francesco Copes
- Laboratory for Biomaterials and Bioengineering, CRC-I, Laval University, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Margot Vansteenland
- Research Group Food Chemistry and Human Nutrition, Department of Food Safety and Food Quality, Ghent University, Coupure Links 653, Block B, 9000 Gent, Belgium
| | - Madalina Albu
- Department of Collagen Research, National Research & Development Institute for Textiles and Leather, Str. Patrascanu Lucretiu, 16, Bucuresti-Sector 3, Bucuresti 030508, București, Romania
| | - Bruno De Meulenaer
- Research Group Food Chemistry and Human Nutrition, Department of Food Safety and Food Quality, Ghent University, Coupure Links 653, Block B, 9000 Gent, Belgium
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC-I, Laval University, Pavillon Pouliot, Québec G1V 0A6, Canada
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281 S4bis, 9000 Gent, Belgium
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Research Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281 S4bis, 9000 Gent, Belgium.
| |
Collapse
|
38
|
Electromechanical Stimulation of 3D Cardiac Microtissues in a Heart-on-Chip Model. Methods Mol Biol 2021; 2373:133-157. [PMID: 34520011 DOI: 10.1007/978-1-0716-1693-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Modeling human cardiac tissues in vitro is essential to elucidate the biological mechanisms related to the heart physiopathology, possibly paving the way for new treatments. Organs-on-chips have emerged as innovative tools able to recreate tissue-specific microenvironments, guiding the development of miniaturized models and offering the opportunity to directly analyze functional readouts. Here we describe the fabrication and operational procedures for the development of a heart-on-chip model, reproducing cardiac biomimetic microenvironment. The device provides 3D cardiac microtissue with a synchronized electromechanical stimulation to support the tissue development. We additionally describe procedures for characterizing tissue evolution and functionality through immunofluorescence, real time qPCR, calcium imaging and microtissue contractility investigations.
Collapse
|
39
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
40
|
Biomimetic reduced graphene oxide coated collagen scaffold for in situ bone regeneration. Sci Rep 2021; 11:16783. [PMID: 34408206 PMCID: PMC8373942 DOI: 10.1038/s41598-021-96271-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/09/2021] [Indexed: 11/08/2022] Open
Abstract
A variety of bone-related diseases and injures and limitations of traditional regeneration methods require new tissue substitutes. Tissue engineering and regeneration combined with nanomedicine can provide different natural or synthetic and combined scaffolds with bone mimicking properties for implantation in the injured area. In this study, we synthesized collagen (Col) and reduced graphene oxide coated collagen (Col-rGO) scaffolds, and we evaluated their in vitro and in vivo effects on bone tissue repair. Col and Col-rGO scaffolds were synthesized by chemical crosslinking and freeze-drying methods. The surface topography, and the mechanical and chemical properties of scaffolds were characterized, showing three-dimensional (3D) porous scaffolds and successful coating of rGO on Col. The rGO coating enhanced the mechanical strength of Col-rGO scaffolds to a greater extent than Col scaffolds by 2.8 times. Furthermore, Col-rGO scaffolds confirmed that graphene addition induced no cytotoxic effects and enhanced the viability and proliferation of human bone marrow-derived mesenchymal stem cells (hBMSCs) with 3D adherence and expansion. Finally, scaffold implantation into rabbit cranial bone defects for 12 weeks showed increased bone formation, confirmed by Hematoxylin–Eosin (H&E) and alizarin red staining. Overall, the study showed that rGO coating improves Col scaffold properties and could be a promising implant for bone injuries.
Collapse
|
41
|
Guo F, Hall AR, Tape CJ, Ling S, Pointon A. Intra- and intercellular signaling pathways associated with drug-induced cardiac pathophysiology. Trends Pharmacol Sci 2021; 42:675-687. [PMID: 34092416 DOI: 10.1016/j.tips.2021.05.004] [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: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
Cardiac physiology and homeostasis are maintained by the interaction of multiple cell types, via both intra- and intercellular signaling pathways. Perturbations in these signaling pathways induced by oncology therapies can reduce cardiac function, ultimately leading to heart failure. As cancer survival increases, related cardiovascular complications are becoming increasingly prevalent, thus identifying the perturbations and cell signaling drivers of cardiotoxicity is increasingly important. Here, we discuss the homotypic and heterotypic cellular interactions that form the basis of intra- and intercellular cardiac signaling pathways, and how oncological agents disrupt these pathways, leading to heart failure. We also highlight the emerging systems biology techniques that can be applied, enabling a deeper understanding of the intra- and intercellular signaling pathways across multiple cell types associated with cardiovascular toxicity.
Collapse
Affiliation(s)
- Fei Guo
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK; Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Andrew R Hall
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Christopher J Tape
- Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Stephanie Ling
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK.
| |
Collapse
|
42
|
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy.
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | | |
Collapse
|
43
|
Wang Z, Wang L, Li T, Liu S, Guo B, Huang W, Wu Y. 3D bioprinting in cardiac tissue engineering. Am J Cancer Res 2021; 11:7948-7969. [PMID: 34335973 PMCID: PMC8315053 DOI: 10.7150/thno.61621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/06/2021] [Indexed: 12/22/2022] Open
Abstract
Heart disease is the main cause of death worldwide. Because death of the myocardium is irreversible, it remains a significant clinical challenge to rescue myocardial deficiency. Cardiac tissue engineering (CTE) is a promising strategy for repairing heart defects and offers platforms for studying cardiac tissue. Numerous achievements have been made in CTE in the past decades based on various advanced engineering approaches. 3D bioprinting has attracted much attention due to its ability to integrate multiple cells within printed scaffolds with complex 3D structures, and many advancements in bioprinted CTE have been reported recently. Herein, we review the recent progress in 3D bioprinting for CTE. After a brief overview of CTE with conventional methods, the current 3D printing strategies are discussed. Bioink formulations based on various biomaterials are introduced, and strategies utilizing composite bioinks are further discussed. Moreover, several applications including heart patches, tissue-engineered cardiac muscle, and other bionic structures created via 3D bioprinting are summarized. Finally, we discuss several crucial challenges and present our perspective on 3D bioprinting techniques in the field of CTE.
Collapse
|
44
|
Lee NH, Bayaraa O, Zechu Z, Kim HS. Biomaterials-assisted spheroid engineering for regenerative therapy. BMB Rep 2021; 54:356-367. [PMID: 34154700 PMCID: PMC8328824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 04/04/2024] Open
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy. [BMB Reports 2021; 54(7): 356-367].
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
45
|
Charrez B, Charwat V, Siemons B, Finsberg H, Miller EW, Edwards AG, Healy KE. In vitro safety "clinical trial" of the cardiac liability of drug polytherapy. Clin Transl Sci 2021; 14:1155-1165. [PMID: 33786981 PMCID: PMC8212738 DOI: 10.1111/cts.13038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Only a handful of US Food and Drug Administration (FDA) Emergency Use Authorizations exist for drug and biologic therapeutics that treat severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection. Potential therapeutics include repurposed drugs, some with cardiac liabilities. We report on a chronic preclinical drug screening platform, a cardiac microphysiological system (MPS), to assess cardiotoxicity associated with repurposed hydroxychloroquine (HCQ) and azithromycin (AZM) polytherapy in a mock phase I safety clinical trial. The MPS contained human heart muscle derived from induced pluripotent stem cells. The effect of drug response was measured using outputs that correlate with clinical measurements, such as QT interval (action potential duration) and drug-biomarker pairing. Chronic exposure (10 days) of heart muscle to HCQ alone elicited early afterdepolarizations and increased QT interval past 5 days. AZM alone elicited an increase in QT interval from day 7 onward, and arrhythmias were observed at days 8 and 10. Monotherapy results mimicked clinical trial outcomes. Upon chronic exposure to HCQ and AZM polytherapy, we observed an increase in QT interval on days 4-8. Interestingly, a decrease in arrhythmias and instabilities was observed in polytherapy relative to monotherapy, in concordance with published clinical trials. Biomarkers, most of them measurable in patients' serum, were identified for negative effects of monotherapy or polytherapy on tissue contractile function, morphology, and antioxidant protection. The cardiac MPS correctly predicted clinical arrhythmias associated with QT prolongation and rhythm instabilities. This high content system can help clinicians design their trials, rapidly project cardiac outcomes, and define new monitoring biomarkers to accelerate access of patients to safe coronavirus disease 2019 (COVID-19) therapeutics.
Collapse
Affiliation(s)
- Bérénice Charrez
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | - Brian Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | | | - Evan W. Miller
- Department of ChemistryUniversity of CaliforniaBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Department of Molecular and Cell BiologyUniversity of California at BerkeleyBerkeleyCaliforniaUSA
| | - Andrew G. Edwards
- Department of PharmacologySchool of MedicineUniversity of California at DavisDavisCaliforniaUSA
| | - Kevin E. Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
- Department of Materials Science and EngineeringUniversity of California at BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
46
|
Aghlmandi A, Nikshad A, Safaralizadeh R, Warkiani ME, Aghebati-Maleki L, Yousefi M. Microfluidics as efficient technology for the isolation and characterization of stem cells. EXCLI JOURNAL 2021; 20:426-443. [PMID: 33746671 PMCID: PMC7975637 DOI: 10.17179/excli2020-3028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/15/2021] [Indexed: 01/09/2023]
Abstract
The recent years have been passed with significant progressions in the utilization of microfluidic technologies for cellular investigations. The aim of microfluidics is to mimic small-scale body environment with features like optical transparency. Microfluidics can screen and monitor different cell types during culture and study cell function in response to stimuli in a fully controlled environment. No matter how the microfluidic environment is similar to in vivo environment, it is not possible to fully investigate stem cells behavior in response to stimuli during cell proliferation and differentiation. Researchers have used stem cells in different fields from fundamental researches to clinical applications. Many cells in the body possess particular functions, but stem cells do not have a specific task and can turn into almost any type of cells. Stem cells are undifferentiated cells with the ability of changing into specific cells that can be essential for the body. Researchers and physicians are interested in stem cells to use them in testing the function of the body's systems and solving their complications. This review discusses the recent advances in utilizing microfluidic techniques for the analysis of stem cells, and mentions the advantages and disadvantages of using microfluidic technology for stem cell research.
Collapse
Affiliation(s)
- Afsoon Aghlmandi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Aylin Nikshad
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Majid Ebrahimi Warkiani
- The School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
47
|
Automated feature extraction from large cardiac electrophysiological data sets. J Electrocardiol 2021; 65:157-162. [PMID: 33640635 DOI: 10.1016/j.jelectrocard.2021.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/19/2021] [Accepted: 02/11/2021] [Indexed: 12/28/2022]
Abstract
RATIONALE A new multi-electrode array-based application for the long-term recording of action potentials from electrogenic cells makes possible exciting cardiac electrophysiology studies in health and disease. With hundreds of simultaneous electrode recordings being acquired over a period of days, the main challenge becomes achieving reliable signal identification and quantification. OBJECTIVE We set out to develop an algorithm capable of automatically extracting regions of high-quality action potentials from terabyte size experimental results and to map the trains of action potentials into a low-dimensional feature space for analysis. METHODS AND RESULTS Our automatic segmentation algorithm finds regions of acceptable action potentials in large data sets of electrophysiological readings. We use spectral methods and support vector machines to classify our readings and to extract relevant features. We are able to show that action potentials from the same cell site can be recorded over days without detrimental effects to the cell membrane. The variability between measurements 24 h apart is comparable to the natural variability of the features at a single time point. CONCLUSIONS Our work contributes towards a non-invasive approach for cardiomyocyte functional maturation, as well as developmental, pathological and pharmacological studies. As the human-derived cardiac model tissue has the genetic makeup of its donor, a powerful tool for individual drug toxicity screening emerges.
Collapse
|
48
|
Jæger KH, Charwat V, Wall S, Healy KE, Tveito A. Identifying Drug Response by Combining Measurements of the Membrane Potential, the Cytosolic Calcium Concentration, and the Extracellular Potential in Microphysiological Systems. Front Pharmacol 2021; 11:569489. [PMID: 33628168 PMCID: PMC7898238 DOI: 10.3389/fphar.2020.569489] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/16/2020] [Indexed: 01/01/2023] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) offer a new means to study and understand the human cardiac action potential, and can give key insight into how compounds may interact with important molecular pathways to destabilize the electrical function of the heart. Important features of the action potential can be readily measured using standard experimental techniques, such as the use of voltage sensitive dyes and fluorescent genetic reporters to estimate transmembrane potentials and cytosolic calcium concentrations. Using previously introduced computational procedures, such measurements can be used to estimate the current density of major ion channels present in hiPSC-CMs, and how compounds may alter their behavior. However, due to the limitations of optical recordings, resolving the sodium current remains difficult from these data. Here we show that if these optical measurements are complemented with observations of the extracellular potential using multi electrode arrays (MEAs), we can accurately estimate the current density of the sodium channels. This inversion of the sodium current relies on observation of the conduction velocity which turns out to be straightforwardly computed using measurements of extracellular waves across the electrodes. The combined data including the membrane potential, the cytosolic calcium concentration and the extracellular potential further opens up for the possibility of accurately estimating the effect of novel drugs applied to hiPSC-CMs.
Collapse
Affiliation(s)
| | - Verena Charwat
- Department of Bioengineering, University of California, Berkeley, CA, United States
| | | | - Kevin E. Healy
- Department of Bioengineering, University of California, Berkeley, CA, United States
- Department of Material Science and Engineering, University of California, Berkeley, CA, United States
| | - Aslak Tveito
- Simula Research Laboratory, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
49
|
Doherty EL, Aw WY, Hickey AJ, Polacheck WJ. Microfluidic and Organ-on-a-Chip Approaches to Investigate Cellular and Microenvironmental Contributions to Cardiovascular Function and Pathology. Front Bioeng Biotechnol 2021; 9:624435. [PMID: 33614613 PMCID: PMC7890362 DOI: 10.3389/fbioe.2021.624435] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/08/2021] [Indexed: 01/06/2023] Open
Abstract
Over the past decade, advances in microfabrication and biomaterials have facilitated the development of microfluidic tissue and organ models to address challenges with conventional animal and cell culture systems. These systems have largely been developed for human disease modeling and preclinical drug development and have been increasingly used to understand cellular and molecular mechanisms, particularly in the cardiovascular system where the characteristic mechanics and architecture are difficult to recapitulate in traditional systems. Here, we review recent microfluidic approaches to model the cardiovascular system and novel insights provided by these systems. Key features of microfluidic approaches include the ability to pattern cells and extracellular matrix (ECM) at cellular length scales and the ability to use patient-derived cells. We focus the review on approaches that have leveraged these features to explore the relationship between genetic mutations and the microenvironment in cardiovascular disease progression. Additionally, we discuss limitations and benefits of the various approaches, and conclude by considering the role further advances in microfabrication technology and biochemistry techniques play in establishing microfluidic cardiovascular disease models as central tools for understanding biological mechanisms and for developing interventional strategies.
Collapse
Affiliation(s)
- Elizabeth L. Doherty
- Joint Department of Biomedical Engineering, University of Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
- University of North Carolina Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wen Yih Aw
- University of North Carolina Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Anthony J. Hickey
- Joint Department of Biomedical Engineering, University of Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
- University of North Carolina Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- RTI International, Durham, NC, United States
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
- Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
- McAllister Heart Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
50
|
Li J, Liu L, Minami I, Miyagawa S, Sawa Y. Fabrication of Thick and Anisotropic Cardiac Tissue on Nanofibrous Substrate for Repairing Infarcted Myocardium. Methods Mol Biol 2021; 2320:65-73. [PMID: 34302648 DOI: 10.1007/978-1-0716-1484-6_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this chapter, we introduce the method for fabricating thick and anisotropic cardiac tissue for heart regeneration. Aligned and biodegradable nanofiber can be prepared by electrospinning Food and Drug Administration-approved poly (lactic-co-glycolic acid) on a rotating drum. After the nanofibers are transferred on to a polydimethylsiloxane frame, the cardiomyocytes could be plated on the nanofiber to form thick and anisotropic cardiac tissue rapidly. Cardiac tissue-like construct could be easily created by one-step method, and transplanted onto the hearts of myocardium infarction models and lead to their functional recovery.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Itsunari Minami
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cell Design for Tissue Construction Faculty of Medicine, Osaka University, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Frontier Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|