1
|
Generali M, Kehl D, Meier D, Zorndt D, Atrott K, Saito H, Emmert MY, Hoerstrup SP. Generation and purification of iPSC-derived cardiomyocytes for clinical applications. Stem Cell Res Ther 2025; 16:189. [PMID: 40251664 PMCID: PMC12008852 DOI: 10.1186/s13287-025-04319-0] [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: 12/06/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Over the past decade, the field of cell therapy has rapidly expanded with the aim to replace and repair damaged cells and/or tissue. Depending on the disease many different cell types can be used as part of such a therapy. Here we focused on the potential treatment of myocardial infarction, where currently available treatment options are not able to regenerate the loss of healthy heart tissue. METHOD We generated good manufacturing practice (GMP)-compatible cardiomyocytes (iCMs) from transgene- and xenofree induced pluripotent stem cells (iPSCs) that can be seamless adapted for clinical applications. Further protocols were established for replating and freezing/thawing iCMs under xenofree conditions. RESULTS iCMs showed a cardiac phenotype, with the expression of specific cardiac markers and absence of pluripotency markers at RNA and protein level. To ensure a pure iCMs population for in vivo applications, we minimized risks of iPSC contamination using RNA-switch technology to ensure safety. CONCLUSION We describe the generation and further processing of xeno- and transgene-free iCMs. The use of GMP-compliant differentiation protocols ab initio facilitates the clinical translation of this project in later stages.
Collapse
Affiliation(s)
- M Generali
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
| | - D Kehl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Meier
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Zorndt
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Atrott
- Center for Surgical Research, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - H Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - M Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - S P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Carlson WD, Bosukonda D, Keck PC, Bey P, Tessier SN, Carlson FR. Cardiac preservation using ex vivo organ perfusion: new therapies for the treatment of heart failure by harnessing the power of growth factors using BMP mimetics like THR-184. Front Cardiovasc Med 2025; 12:1535778. [PMID: 40171539 PMCID: PMC11960666 DOI: 10.3389/fcvm.2025.1535778] [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: 11/27/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
As heart transplantation continues to be the gold standard therapy for end-stage heart failure, the imbalance between the supply of hearts, and the demand for them, continues to get worse. In the US alone, with less than 4,000 hearts suitable for transplant and over 100,000 potential recipients, this therapy is only available to a very few. The use of hearts Donated after Circulatory Death (DCD) and Donation after Brain Death (DBD) using ex vivo machine perfusion (EVMP) is a promising approach that has already increased the availability of suitable organs for heart transplantation. EVMP offers the promise of enabling the expansion of the overall number of heart transplants and lower rates of early graft dysfunction. These are realized through (1) safe extension of the time between procurement and transplantation and (2) ex vivo assessment of preserved hearts. Notably, ex vivo perfusion has facilitated the donation of DCD hearts and improved the success of transplantation. Nevertheless, DCD hearts suffer from serious preharvest ischemia/reperfusion injury (IRI). Despite these developments, only 40% of hearts offered for transplantation can be utilized. These devices do offer an opportunity to evaluate donor hearts for transplantation, resuscitate organs previously deemed unsuitable for transplantation, and provide a platform for the development of novel therapeutics to limit cardiac injury. Bone Morphogenetic Protein (BMP) signaling is a new target which holds the potential for ameliorating myocardial IRI. Recent studies have demonstrated that BMP signaling has a significant role in blocking the deleterious effects of injury to the heart. We have designed novel small peptide BMP mimetics that act via activin receptor-like kinase (ALK3), a type I BMP receptor. They are capable of (1) inhibiting inflammation and apoptosis, (2) blocking/reversing the epithelial-mesenchymal transition (EMT) and fibrosis, and (3) promoting tissue regeneration. In this review, we explore the promise that novel therapeutics, including these BMP mimetics, offer for the protection of hearts against myocardial injury during ex vivo transportation for cardiac transplantation. This protection represents a significant advance and a promising ex vivo therapeutic approach to expanding the donor pool by increasing the number of transplantable hearts.
Collapse
Affiliation(s)
- William D. Carlson
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Therapeutics by Design, Weston, MA, United States
| | - Dattatreyamurty Bosukonda
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Therapeutics by Design, Weston, MA, United States
| | | | - Philippe Bey
- Therapeutics by Design, Weston, MA, United States
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children’s Hospital, Boston, MA, United States
| | | |
Collapse
|
3
|
Wu Y, Wang Y, Xiao M, Zhang G, Zhang F, Tang M, Lei W, Jiang Z, Li X, Zhang H, Ren X, Xu Y, Zhao X, Guo C, Lan H, Shen Z, Zhang J, Hu S. 3D-Printed Myocardium-Specific Structure Enhances Maturation and Therapeutic Efficacy of Engineered Heart Tissue in Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409871. [PMID: 39840547 PMCID: PMC11905000 DOI: 10.1002/advs.202409871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Despite advancements in engineered heart tissue (EHT), challenges persist in achieving accurate dimensional accuracy of scaffolds and maturing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), a primary source of functional cardiac cells. Drawing inspiration from cardiac muscle fiber arrangement, a three-dimensional (3D)-printed multi-layered microporous polycaprolactone (PCL) scaffold is created with interlayer angles set at 45° to replicate the precise structure of native cardiac tissue. Compared with the control group and 90° PCL scaffolds, the 45° PCL scaffolds exhibited superior biocompatibility for cell culture and improved hiPSC-CM maturation in calcium handling. RNA sequencing demonstrated that the 45° PCL scaffold promotes the mature phenotype in hiPSC-CMs by upregulating ion channel genes. Using the 45° PCL scaffold, a multi-cellular EHT is successfully constructed, incorporating human cardiomyocytes, endothelial cells, and mesenchymal stem cells. These complex EHTs significantly enhanced hiPSC-CM engraftment in vivo, attenuated ventricular remodeling, and improved cardiac function in mouse myocardial infarction. In summary, the myocardium-specific structured EHT developed in this study represents a promising advancement in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Yong Wu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Yaning Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Miao Xiao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Guangming Zhang
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdaoShandong266520China
| | - Feixiang Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Mingliang Tang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsu226001China
| | - Wei Lei
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Ziyun Jiang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Xiaoyun Li
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Huiqi Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Xiaoyi Ren
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Yue Xu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Xiaotong Zhao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Chenxu Guo
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdaoShandong266520China
| | - Hongbo Lan
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdaoShandong266520China
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| | - Jianyi Zhang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringThe University of Alabama at BirminghamBirminghamAL35233USA
- Department of MedicineDivision of Cardiovascular DiseaseSchool of MedicineThe University of Alabama at BirminghamBirminghamAL35233USA
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalState Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215000China
| |
Collapse
|
4
|
Wang X, Cao L, Chang R, Shen J, Ma L, Li Y. Elucidating cardiomyocyte heterogeneity and maturation dynamics through integrated single-cell and spatial transcriptomics. iScience 2025; 28:111596. [PMID: 39811652 PMCID: PMC11732507 DOI: 10.1016/j.isci.2024.111596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/27/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
The intricate development and functionality of the mammalian heart are influenced by the heterogeneous nature of cardiomyocytes (CMs). In this study, single-cell and spatial transcriptomics were utilized to analyze cells from neonatal mouse hearts, resulting in a comprehensive atlas delineating the transcriptional profiles of distinct CM subsets. A continuum of maturation states was elucidated, emphasizing a progressive developmental trajectory rather than discrete stages. This approach enabled the mapping of these states across various cardiac regions, illuminating the spatial organization of CM development and the influence of the cellular microenvironment. Notably, a subset of transitional CMs was identified, characterized by a transcriptional signature marking a pivotal maturation phase, presenting a promising target for therapeutic strategies aimed at enhancing cardiac regeneration. This atlas not only elucidates fundamental aspects of cardiac development but also serves as a valuable resource for advancing research into cardiac physiology and pathology, with significant implications for regenerative medicine.
Collapse
Affiliation(s)
- Xiaoying Wang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Life Sciences and Technology, Tongji University, Shanghai, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lizhi Cao
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Rui Chang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Junwei Shen
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Linlin Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yanfei Li
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
5
|
Cui H, Yu ZX, Huang Y, Hann SY, Esworthy T, Shen YL, Zhang LG. 3D printing of thick myocardial tissue constructs with anisotropic myofibers and perfusable vascular channels. BIOMATERIALS ADVANCES 2023; 153:213579. [PMID: 37566935 DOI: 10.1016/j.bioadv.2023.213579] [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: 05/31/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Engineering of myocardial tissues has become a promising therapeutic strategy for treating myocardial infarction (MI). However, a significant challenge remains in generating clinically relevant myocardial tissues that possess native microstructural characteristics and fulfill the requirements for implantation within the human body. In this study, a thick 3D myocardial construct with anisotropic myofibers and perfusable branched vascular channels is created with clinically relevant dimensions using a customized beam-scanning stereolithography printing technique. To obtain tissue-specific matrix niches, a decellularized extracellular matrix microfiber-reinforced gelatin-based bioink is developed. The bioink plays a crucial role in facilitating the precise manufacturing of a hierarchical microstructure, enabling us to better replicate the physiological characteristics of the native myocardial tissue matrix in terms of structure, biomechanics, and bioactivity. Through the integration of the tailored bioink with our printing method, we demonstrate a biomimetic architecture, appropriate biomechanical properties, vascularization, and improved functionality of induced pluripotent stem cell-derived cardiomyocytes in the thick tissue construct in vitro. This work not only offers a novel and effective means to generate biomimetic heart tissue in vitro for the treatment of MI, but also introduces a potential methodology for creating clinically relevant tissue products to aid in other complex tissue/organ regeneration and disease model applications.
Collapse
Affiliation(s)
- Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Yimin Huang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Yin-Lin Shen
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America; Departments of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, United States of America; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, United States of America; Department of Medicine, The George Washington University, Washington, DC 20052, United States of America.
| |
Collapse
|
6
|
English EJ, Samolyk BL, Gaudette GR, Pins GD. Micropatterned fibrin scaffolds increase cardiomyocyte alignment and contractility for the fabrication of engineered myocardial tissue. J Biomed Mater Res A 2023; 111:1309-1321. [PMID: 36932841 PMCID: PMC11128133 DOI: 10.1002/jbm.a.37530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023]
Abstract
Cardiovascular disease is the leading cause of death in the United States, which can result in blockage of a coronary artery, triggering a myocardial infarction (MI), scar tissue formation in the myocardium, and ultimately heart failure. Currently, the gold-standard solution for total heart failure is a heart transplantation. An alternative to total-organ transplantation is surgically remodeling the ventricle with the implantation of a cardiac patch. Acellular cardiac patches have previously been investigated using synthetic or decellularized native materials to improve cardiac function. However, a limitation of this strategy is that acellular cardiac patches only reshape the ventricle and do not increase cardiac contractile function. Toward the development of a cardiac patch, our laboratory previously developed a cell-populated composite fibrin scaffold and aligned microthreads to recapitulate the mechanical properties of native myocardium. In this study, we explore micropatterning the surfaces of fibrin gels to mimic anisotropic native tissue architecture and promote cellular alignment of human induced pluripotent stem cell cardiomyocytes (hiPS-CM), which is crucial for increasing scaffold contractile properties. hiPS-CMs seeded on micropatterned surfaces exhibit cellular elongation, distinct sarcomere alignment, and circumferential connexin-43 staining at 14 days of culture, which are necessary for mature contractile properties. Constructs were also subject to electrical stimulation during culture to promote increased contractile properties. After 7 days of stimulation, contractile strains of micropatterned constructs were significantly higher than unpatterned controls. These results suggest that the use of micropatterned topographic cues on fibrin scaffolds may be a promising strategy for creating engineered cardiac tissue.
Collapse
Affiliation(s)
- Elizabeth J. English
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Tessera Therapeutics, Somerville, Massachusetts, USA
| | - Bryanna L. Samolyk
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Glenn R. Gaudette
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Department of Engineering, Boston College, Newton, Massachusetts, USA
| | - George D. Pins
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
7
|
Wang J, Wu Y, Wang Y, Shuai Y, Xu Z, Wan Q, Chen Y, Yang M. Graphene Oxide-Coated Patterned Silk Fibroin Films Promote Cell Adhesion and Induce Cardiomyogenic Differentiation of Human Mesenchymal Stem Cells. Biomolecules 2023; 13:990. [PMID: 37371570 DOI: 10.3390/biom13060990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/03/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiac tissue engineering is a promising strategy for the treatment of myocardial damage. Mesenchymal stem cells (MSCs) are extensively used in tissue engineering. However, transformation of MSCs into cardiac myocytes is still a challenge. Furthermore, weak adhesion of MSCs to substrates often results in poor cell viability. Here, we designed a composite matrix based on silk fibroin (SF) and graphene oxide (GO) for improving the cell adhesion and directing the differentiation of MSCs into cardiac myocytes. Specifically, patterned SF films were first produced by soft lithographic. After being treated by air plasma, GO nanosheets could be successfully coated on the patterned SF films to construct the desired matrix (P-GSF). The resultant P-GSF films presented a nano-topographic surface characterized by linear grooves interlaced with GO ridges. The P-GSF films exhibited high protein absorption and suitable mechanical strength. Furthermore, the P-GSF films accelerated the early cell adhesion and directed the growth orientation of MSCs. RT-PCR results and immunofluorescence imaging demonstrated that the P-GSF films significantly improved the cardiomyogenic differentiation of MSCs. This work indicates that patterned SF films coated with GO are promising matrix in the field of myocardial repair tissue engineering.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yi Wu
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yecheng Wang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zongpu Xu
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Quan Wan
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yuyin Chen
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
8
|
Patel L, Worch JC, Dove AP, Gehmlich K. The Utilisation of Hydrogels for iPSC-Cardiomyocyte Research. Int J Mol Sci 2023; 24:9995. [PMID: 37373141 PMCID: PMC10298477 DOI: 10.3390/ijms24129995] [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: 05/22/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiac fibroblasts' (FBs) and cardiomyocytes' (CMs) behaviour and morphology are influenced by their environment such as remodelling of the myocardium, thus highlighting the importance of biomaterial substrates in cell culture. Biomaterials have emerged as important tools for the development of physiological models, due to the range of adaptable properties of these materials, such as degradability and biocompatibility. Biomaterial hydrogels can act as alternative substrates for cellular studies, which have been particularly key to the progression of the cardiovascular field. This review will focus on the role of hydrogels in cardiac research, specifically the use of natural and synthetic biomaterials such as hyaluronic acid, polydimethylsiloxane and polyethylene glycol for culturing induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). The ability to fine-tune mechanical properties such as stiffness and the versatility of biomaterials is assessed, alongside applications of hydrogels with iPSC-CMs. Natural hydrogels often display higher biocompatibility with iPSC-CMs but often degrade quicker, whereas synthetic hydrogels can be modified to facilitate cell attachment and decrease degradation rates. iPSC-CM structure and electrophysiology can be assessed on natural and synthetic hydrogels, often resolving issues such as immaturity of iPSC-CMs. Biomaterial hydrogels can thus provide a more physiological model of the cardiac extracellular matrix compared to traditional 2D models, with the cardiac field expansively utilising hydrogels to recapitulate disease conditions such as stiffness, encourage alignment of iPSC-CMs and facilitate further model development such as engineered heart tissues (EHTs).
Collapse
Affiliation(s)
- Leena Patel
- Institute of Cardiovascular Science, University of Birmingham, Birmingham B15 2TT, UK;
| | - Joshua C. Worch
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, UK; (J.C.W.); (A.P.D.)
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, UK; (J.C.W.); (A.P.D.)
| | - Katja Gehmlich
- Institute of Cardiovascular Science, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
9
|
Esparza A, Jimenez N, Joddar B, Natividad-Diaz S. Development of in vitro cardiovascular tissue models within capillary circuit microfluidic devices fabricated with 3D Stereolithography printing. RESEARCH SQUARE 2023:rs.3.rs-2667200. [PMID: 36993455 PMCID: PMC10055652 DOI: 10.21203/rs.3.rs-2667200/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human cardiovascular tissue and diseases are difficult to study for novel drug discovery and fundamental cellular/molecular processes due to limited availability of physiologically-relevant models in vitro.[1-3] Animal models may resemble human heart structure, however there are significant differences from human cardiovascular physiology including biochemical signaling, and gene expression.[4-6] In vitro microfluidic tissue models provide a less expensive, more controlled, and reproducible platform for better quantification of isolated cellular processes in response to biochemical or biophysical stimulus.[6-12] The capillary driven-flow microfluidic device in this study was manufactured with a 3D stereolithography (SLA) printed mold and is a closed circuit system operating on principles of capillary action allowing continuous fluid movement without external power supply. Human umbilical vein endothelial cells (HUVECs) and human cardiomyocytes (AC16) were encapsulated into a fibrin hydrogel to form vascular (VTM) and cardiac (CTM) tissue models respectively. To determine response to biophysical stimulus, the 3D cardiovascular tissue was directly loaded into the device tissue culture chambers that either had no microposts (DWoP) or microposts (DWPG) for 1, 3 and 5 days. The tissues were analyzed with fluorescent microscopy for morphological differences, average tube length, and cell orientation between tissues cultured in both conditions. In DWPG VTMs displayed capillary-like tube formation with visible cell alignment and orientation, while AC16s continued to elongate around microposts by day 5. VTM and CTM models in devices with posts (DWPG) displayed cell alignment and orientation after 5 days, indicated the microposts induced biophysical cues to guide cell structure and specific organization.
Collapse
|
10
|
Joddar B, Natividad-Diaz SL, Padilla AE, Esparza AA, Ramirez SP, Chambers DR, Ibaroudene H. Engineering approaches for cardiac organoid formation and their characterization. Transl Res 2022; 250:46-67. [PMID: 35995380 PMCID: PMC10370285 DOI: 10.1016/j.trsl.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
Cardiac organoids are 3-dimensional (3D) structures composed of tissue or niche-specific cells, obtained from diverse sources, encapsulated in either a naturally derived or synthetic, extracellular matrix scaffold, and include exogenous biochemical signals such as essential growth factors. The overarching goal of developing cardiac organoid models is to establish a functional integration of cardiomyocytes with physiologically relevant cells, tissues, and structures like capillary-like networks composed of endothelial cells. These organoids used to model human heart anatomy, physiology, and disease pathologies in vitro have the potential to solve many issues related to cardiovascular drug discovery and fundamental research. The advent of patient-specific human-induced pluripotent stem cell-derived cardiovascular cells provide a unique, single-source approach to study the complex process of cardiovascular disease progression through organoid formation and incorporation into relevant, controlled microenvironments such as microfluidic devices. Strategies that aim to accomplish such a feat include microfluidic technology-based approaches, microphysiological systems, microwells, microarray-based platforms, 3D bioprinted models, and electrospun fiber mat-based scaffolds. This article discusses the engineering or technology-driven practices for making cardiac organoid models in comparison with self-assembled or scaffold-free methods to generate organoids. We further discuss emerging strategies for characterization of the bio-assembled cardiac organoids including electrophysiology and machine-learning and conclude with prospective points of interest for engineering cardiac tissues in vitro.
Collapse
Affiliation(s)
- Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Andie E Padilla
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Aibhlin A Esparza
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | | | | |
Collapse
|
11
|
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
|
12
|
Badr-Eldin SM, Aldawsari HM, Kotta S, Deb PK, Venugopala KN. Three-Dimensional In Vitro Cell Culture Models for Efficient Drug Discovery: Progress So Far and Future Prospects. Pharmaceuticals (Basel) 2022; 15:926. [PMID: 36015074 PMCID: PMC9412659 DOI: 10.3390/ph15080926] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Despite tremendous advancements in technologies and resources, drug discovery still remains a tedious and expensive process. Though most cells are cultured using 2D monolayer cultures, due to lack of specificity, biochemical incompatibility, and cell-to-cell/matrix communications, they often lag behind in the race of modern drug discovery. There exists compelling evidence that 3D cell culture models are quite promising and advantageous in mimicking in vivo conditions. It is anticipated that these 3D cell culture methods will bridge the translation of data from 2D cell culture to animal models. Although 3D technologies have been adopted widely these days, they still have certain challenges associated with them, such as the maintenance of a micro-tissue environment similar to in vivo models and a lack of reproducibility. However, newer 3D cell culture models are able to bypass these issues to a maximum extent. This review summarizes the basic principles of 3D cell culture approaches and emphasizes different 3D techniques such as hydrogels, spheroids, microfluidic devices, organoids, and 3D bioprinting methods. Besides the progress made so far in 3D cell culture systems, the article emphasizes the various challenges associated with these models and their potential role in drug repositioning, including perspectives from the COVID-19 pandemic.
Collapse
Affiliation(s)
- Shaimaa M. Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hibah M. Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sabna Kotta
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa
| |
Collapse
|
13
|
Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, Azami M. Regenerative strategies for the consequences of myocardial infarction: Chronological indication and upcoming visions. Biomed Pharmacother 2021; 146:112584. [PMID: 34968921 DOI: 10.1016/j.biopha.2021.112584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Heart muscle injury and an elevated troponin level signify myocardial infarction (MI), which may result in defective and uncoordinated segments, reduced cardiac output, and ultimately, death. Physicians apply thrombolytic therapy, coronary artery bypass graft (CABG) surgery, or percutaneous coronary intervention (PCI) to recanalize and restore blood flow to the coronary arteries, albeit they were not convincingly able to solve the heart problems. Thus, researchers aim to introduce novel substitutional therapies for regenerating and functionalizing damaged cardiac tissue based on engineering concepts. Cell-based engineering approaches, utilizing biomaterials, gene, drug, growth factor delivery systems, and tissue engineering are the most leading studies in the field of heart regeneration. Also, understanding the primary cause of MI and thus selecting the most efficient treatment method can be enhanced by preparing microdevices so-called heart-on-a-chip. In this regard, microfluidic approaches can be used as diagnostic platforms or drug screening in cardiac disease treatment. Additionally, bioprinting technique with whole organ 3D printing of human heart with major vessels, cardiomyocytes and endothelial cells can be an ideal goal for cardiac tissue engineering and remarkable achievement in near future. Consequently, this review discusses the different aspects, advancements, and challenges of the mentioned methods with presenting the advantages and disadvantages, chronological indications, and application prospects of various novel therapeutic approaches.
Collapse
Affiliation(s)
- Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran
| | - Hanif Goran Orimi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Roya Ramzgouyan
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Nemati
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
14
|
Cho J, Kim S, Lee H, Rah W, Cho HC, Kim NK, Bae S, Shin DH, Lee MG, Park IH, Tanaka Y, Shin E, Yi H, Han JW, Hwang PTJ, Jun HW, Park HJ, Cho K, Lee SW, Jung JK, Levit RD, Sussman MA, Harvey RP, Yoon YS. Regeneration of infarcted mouse hearts by cardiovascular tissue formed via the direct reprogramming of mouse fibroblasts. Nat Biomed Eng 2021; 5:880-896. [PMID: 34426676 PMCID: PMC8809198 DOI: 10.1038/s41551-021-00783-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts can be directly reprogrammed into cardiomyocytes, endothelial cells or smooth muscle cells. Here we report the reprogramming of mouse tail-tip fibroblasts simultaneously into cells resembling these three cell types using the microRNA mimic miR-208b-3p, ascorbic acid and bone morphogenetic protein 4, as well as the formation of tissue-like structures formed by the directly reprogrammed cells. Implantation of the formed cardiovascular tissue into the infarcted hearts of mice led to the migration of reprogrammed cells to the injured tissue, reducing regional cardiac strain and improving cardiac function. The migrated endothelial cells and smooth muscle cells contributed to vessel formation, and the migrated cardiomyocytes, which initially displayed immature characteristics, became mature over time and formed gap junctions with host cardiomyocytes. Direct reprogramming of somatic cells to make cardiac tissue may aid the development of applications in cell therapy, disease modelling and drug discovery for cardiovascular diseases.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Nam Kyun Kim
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Seongho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dong Hoon Shin
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Yoshiaki Tanaka
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Shin
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hong Yi
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, GA, USA
| | - Ji Woong Han
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick Tae Joon Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyuwon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sang Wook Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jae Kyung Jung
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark A Sussman
- San Diego State University Heart Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
A continuum model and simulations for large deformation of anisotropic fiber-matrix composites for cardiac tissue engineering. J Mech Behav Biomed Mater 2021; 121:104627. [PMID: 34130078 DOI: 10.1016/j.jmbbm.2021.104627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
Cardiac patch therapies promise to restore heart function and lower the risk of heart failure after heart attack. Fiber-matrix engineered tissue scaffolds have gained significant attention due to their tunable micro-structures, providing nonlinear mechanical properties similar to native anisotropic heart tissues. Mechanical properties of engineered scaffolds directly affect the stress fields generated inside and around the tissue scaffolds and have significant impact on the tissue functionality. Currently, biomedical cardiac patches are designed through experimentation and there exists a need for an accurate model that will allow micro-structural design optimization and analysis of effectiveness of the implanted patches. We have developed a three-dimensional large strain continuum model that can predict nonlinear, anisotropic mechanical response of engineered tissue scaffolds that have two orientation families of fibers inside a bulk hydrogel matrix. We have validated the predictive capability of our continuum model for the fiber-matrix composite using selected experiments and a suite of detailed finite element analysis that incorporated the micro-structural details of the composites. Comparing the continuum model predictions (1 element) against the representative volume micro-structural geometry finite element simulations (with greater than 4,00,000 elements), we show that the proposed model can accurately predict nonlinear mechanical behavior of highly anisotropic tissue scaffolds in both the longitudinal and transverse directions, as a function of the critical design parameters inter-fiber angle and fiber spacing. We show that the model can also capture native heart tissue's anisotropic large strain mechanical response. We implemented our model in the finite element software Abaqus by writing a user material subroutine UANISOHYPER and demonstrated its predictive abilities by conducting a full three-dimensional analysis of engineered tissue patch application on an infarcted heart.
Collapse
|
16
|
Cho S, Lee C, Skylar-Scott MA, Heilshorn SC, Wu JC. Reconstructing the heart using iPSCs: Engineering strategies and applications. J Mol Cell Cardiol 2021; 157:56-65. [PMID: 33895197 DOI: 10.1016/j.yjmcc.2021.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have emerged as a key component of cardiac tissue engineering, enabling studies of cardiovascular disease mechanisms, drug responses, and developmental processes in human 3D tissue models assembled from isogenic cells. Since the very first engineered heart tissues were introduced more than two decades ago, a wide array of iPSC-derived cardiac spheroids, organoids, and heart-on-a-chip models have been developed incorporating the latest available technologies and materials. In this review, we will first outline the fundamental biological building blocks required to form a functional unit of cardiac muscle, including iPSC-derived cells differentiated by soluble factors (e.g., small molecules), extracellular matrix scaffolds, and exogenous biophysical maturation cues. We will then summarize the different fabrication approaches and strategies employed to reconstruct the heart in vitro at varying scales and geometries. Finally, we will discuss how these platforms, with continued improvements in scalability and tissue maturity, can contribute to both basic cardiovascular research and clinical applications in the future.
Collapse
Affiliation(s)
- Sangkyun Cho
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Chelsea Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Mark A Skylar-Scott
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Betty Irene Moore Children's Heart Center, Stanford Children's Health, Stanford, CA 94025, USA
| | - Sarah C Heilshorn
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA.
| |
Collapse
|
17
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
18
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
19
|
Lowenthal J, Gerecht S. If You Build It, They Will Come: Towards Self-Assembly of Functional Cardiovascular Tissues. Circ Res 2020; 127:225-228. [PMID: 32614715 DOI: 10.1161/circresaha.120.317111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Justin Lowenthal
- From the Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD (J.L.).,Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (J.L., S.G.).,Department of Biomedical Engineering, School of Medicine and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (J.L., S.G.)
| | - Sharon Gerecht
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (J.L., S.G.).,Department of Biomedical Engineering, School of Medicine and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (J.L., S.G.).,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (S.G.).,Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD (S.G.).,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD (S.G.)
| |
Collapse
|
20
|
Jonker SS, Giraud GD, Chang EI, Elman MR, Louey S. Coronary vascular growth matches IGF-1-stimulated cardiac growth in fetal sheep. FASEB J 2020; 34:10041-10055. [PMID: 32573852 DOI: 10.1096/fj.202000215r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
As loss of contractile function in heart disease could often be mitigated by increased cardiomyocyte number, expansion of cardiomyocyte endowment paired with increased vascular supply is a desirable therapeutic goal. Insulin-like growth factor 1 (IGF-1) administration increases fetal cardiomyocyte proliferation and heart mass, but how fetal IGF-1 treatment affects coronary growth and function is unknown. Near-term fetal sheep underwent surgical instrumentation and were studied from 127 to 134 d gestation (term = 147 d), receiving either IGF-1 LR3 or vehicle. Coronary growth and function were interrogated using pressure-flow relationships, an episode of acute hypoxia with progressive blockade of adenosine receptors and nitric oxide synthase, and by modeling the determinants of coronary flow. The main findings were that coronary conductance was preserved on a per-gram basis following IGF-1 treatment, adenosine and nitric oxide contributed to hypoxia-mediated coronary vasodilation similarly in IGF-1-treated and Control fetuses, and the relationships between coronary flow and blood oxygen contents were similar between groups. We conclude that IGF-1-stimulated fetal myocardial growth is accompanied by appropriate expansion and function of the coronary vasculature. These findings support IGF-1 as a potential strategy to increase cardiac myocyte and coronary vascular endowment at birth.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| | - George D Giraud
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA.,Division of Cardiology, VA Portland Health Care System, Portland, OR, USA
| | - Eileen I Chang
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| | - Miriam R Elman
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, USA
| | - Samantha Louey
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
21
|
Wheelwright M, Mikkila J, Bedada FB, Mandegar MA, Thompson BR, Metzger JM. Advancing physiological maturation in human induced pluripotent stem cell-derived cardiac muscle by gene editing an inducible adult troponin isoform switch. Stem Cells 2020; 38:1254-1266. [PMID: 32497296 DOI: 10.1002/stem.3235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/11/2020] [Indexed: 01/11/2023]
Abstract
Advancing maturation of stem cell-derived cardiac muscle represents a major barrier to progress in cardiac regenerative medicine. Cardiac muscle maturation involves a myriad of gene, protein, and cell-based transitions, spanning across all aspects of cardiac muscle form and function. We focused here on a key developmentally controlled transition in the cardiac sarcomere, the functional unit of the heart. Using a gene-editing platform, human induced pluripotent stem cell (hiPSCs) were engineered with a drug-inducible expression cassette driving the adult cardiac troponin I (cTnI) regulatory isoform, a transition shown to be a rate-limiting step in advancing sarcomeric maturation of hiPSC cardiac muscle (hiPSC-CM) toward the adult state. Findings show that induction of the adult cTnI isoform resulted in the physiological acquisition of adult-like cardiac contractile function in hiPSC-CMs in vitro. Specifically, cTnI induction accelerated relaxation kinetics at baseline conditions, a result independent of alterations in the kinetics of the intracellular Ca2+ transient. In comparison, isogenic unedited hiPSC-CMs had no cTnI induction and no change in relaxation function. Temporal control of adult cTnI isoform induction did not alter other developmentally regulated sarcomere transitions, including myosin heavy chain isoform expression, nor did it affect expression of SERCA2a or phospholamban. Taken together, precision genetic targeting of sarcomere maturation via inducible TnI isoform switching enables physiologically relevant adult myocardium-like contractile adaptations that are essential for beat-to-beat modulation of adult human heart performance. These findings have relevance to hiPSC-CM structure-function and drug-discovery studies in vitro, as well as for potential future clinical applications of physiologically optimized hiPSC-CM in cardiac regeneration/repair.
Collapse
Affiliation(s)
- Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jennifer Mikkila
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Mohammad A Mandegar
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
22
|
Patrick PS, Bear JC, Fitzke HE, Zaw-Thin M, Parkin IP, Lythgoe MF, Kalber TL, Stuckey DJ. Radio-metal cross-linking of alginate hydrogels for non-invasive in vivo imaging. Biomaterials 2020; 243:119930. [PMID: 32171101 PMCID: PMC7103761 DOI: 10.1016/j.biomaterials.2020.119930] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/30/2022]
Abstract
Alginate hydrogels are cross-linked polymers with high water content, tuneable chemical and material properties, and a range of biomedical applications including drug delivery, tissue engineering, and cell therapy. However, their similarity to soft tissue often renders them undetectable within the body using conventional bio-medical imaging techniques. This leaves much unknown about their behaviour in vivo, posing a challenge to therapy development and validation. To address this, we report a novel, fast, and simple method of incorporating the nuclear imaging radio-metal 111In into the structure of alginate hydrogels by utilising its previously-undescribed capacity as an ionic cross-linking agent. This enabled non-invasive in vivo nuclear imaging of hydrogel delivery and retention across the whole body, over time, and across a range of model therapies including: nasal and oral drug delivery, stem cell transplantation, and cardiac tissue engineering. This information will facilitate the development of novel therapeutic hydrogel formulations, encompassing alginate, across disease categories.
Collapse
Affiliation(s)
- P Stephen Patrick
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - Joseph C Bear
- School of Life Science, Pharmacy & Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Heather E Fitzke
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - May Zaw-Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Ivan P Parkin
- Materials Chemistry Centre, Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Daniel J Stuckey
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| |
Collapse
|
23
|
Cui H, Liu C, Esworthy T, Huang Y, Yu ZX, Zhou X, San H, Lee SJ, Hann SY, Boehm M, Mohiuddin M, Fisher JP, Zhang LG. 4D physiologically adaptable cardiac patch: A 4-month in vivo study for the treatment of myocardial infarction. SCIENCE ADVANCES 2020; 6:eabb5067. [PMID: 32637623 PMCID: PMC7314523 DOI: 10.1126/sciadv.abb5067] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/11/2020] [Indexed: 05/20/2023]
Abstract
There has been considerable progress in engineering cardiac scaffolds for the treatment of myocardial infarction (MI). However, it is still challenging to replicate the structural specificity and variability of cardiac tissues using traditional bioengineering approaches. In this study, a four-dimensional (4D) cardiac patch with physiological adaptability has been printed by beam-scanning stereolithography. By combining a unique 4D self-morphing capacity with expandable microstructure, the specific design has been shown to improve both the biomechanical properties of the patches themselves and the dynamic integration of the patch with the beating heart. Our results demonstrate improved vascularization and cardiomyocyte maturation in vitro under physiologically relevant mechanical stimulation, as well as increased cell engraftment and vascular supply in a murine chronic MI model. This work not only potentially provides an effective treatment method for MI but also contributes a cutting-edge methodology to enhance the structural design of complex tissues for organ regeneration.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Yimin Huang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Hong San
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Se-jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Manfred Boehm
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Muhammad Mohiuddin
- Cardiac Xenotransplantation Program, Department of Surgery, University of Maryland, Baltimore, MD 21201, USA
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Medicine, The George Washington University, Washington, DC 20052, USA
- Corresponding author.
| |
Collapse
|
24
|
Simon LR, Masters KS. Disease-inspired tissue engineering: Investigation of cardiovascular pathologies. ACS Biomater Sci Eng 2019; 6:2518-2532. [PMID: 32974421 DOI: 10.1021/acsbiomaterials.9b01067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Once focused exclusively on the creation of tissues to repair or replace diseased or damaged organs, the field of tissue engineering has undergone an important evolution in recent years. Namely, tissue engineering techniques are increasingly being applied to intentionally generate pathological conditions. Motivated in part by the wide gap between 2D cultures and animal models in the current disease modeling continuum, disease-inspired tissue-engineered platforms have numerous potential applications, and may serve to advance our understanding and clinical treatment of various diseases. This review will focus on recent progress toward generating tissue-engineered models of cardiovascular diseases, including cardiac hypertrophy, fibrosis, and ischemia reperfusion injury, atherosclerosis, and calcific aortic valve disease, with an emphasis on how these disease-inspired platforms can be used to decipher disease etiology. Each pathology is discussed in the context of generating both disease-specific cells as well as disease-specific extracellular environments, with an eye toward future opportunities to integrate different tools to yield more complex and physiologically relevant culture platforms. Ultimately, the development of effective disease treatments relies upon our ability to develop appropriate experimental models; as cardiovascular diseases are the leading cause of death worldwide, the insights yielded by improved in vitro disease modeling could have substantial ramifications for public health and clinical care.
Collapse
Affiliation(s)
- LaTonya R Simon
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705
| | - Kristyn S Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705.,Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
25
|
Beffagna G. Zebrafish as a Smart Model to Understand Regeneration After Heart Injury: How Fish Could Help Humans. Front Cardiovasc Med 2019; 6:107. [PMID: 31448289 PMCID: PMC6691037 DOI: 10.3389/fcvm.2019.00107] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/19/2019] [Indexed: 12/26/2022] Open
Abstract
Myocardial infarction (MI) in humans is a common cause of cardiac injury and results in irreversible loss of myocardial cells and formation of fibrotic scar tissue. This fibrotic tissue preserves the integrity of the ventricular wall but undermines pump function, leading to congestive heart failure. Unfortunately, the mammalian heart is unable to replace cardiomyocytes, so the life expectancy for patients after an episode of MI is lower than for most common types of cancers. Whereas, humans cannot efficiently regenerate their heart after injury, the teleost zebrafish have the capability to repair a “broken” heart. The zebrafish is probably one of the most important models for developmental and regenerative biology of the heart. In the last decades, the zebrafish has become increasingly important for scientific research: it has many characteristics that make it a smart model for studying human disease. Moreover, adult zebrafish efficiently regenerate their hearts following different forms of injury. Due to these characteristics, and to the availability of genetic approaches, and biosensor zebrafish lines, it has been established useful for studying molecular mechanisms of heart regeneration. Regeneration of cardiomyocytes in zebrafish is not based on stem cells or transdifferentiation of other cells but on the proliferation of preexisting cardiomyocytes. For this reason, future studies into the zebrafish cardiac regenerative mechanisms could identify specific molecules able to regulate the proliferation of preexisting cardiomyocytes; these factors may be studied in order to understand regulation of myocardial plasticity in cardiac repair processes after injury and, in particular, after MI in humans.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
26
|
De Luca M, Aiuti A, Cossu G, Parmar M, Pellegrini G, Robey PG. Advances in stem cell research and therapeutic development. Nat Cell Biol 2019; 21:801-811. [PMID: 31209293 DOI: 10.1038/s41556-019-0344-z] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 05/09/2019] [Indexed: 12/12/2022]
Abstract
Despite many reports of putative stem-cell-based treatments in genetic and degenerative disorders or severe injuries, the number of proven stem cell therapies has remained small. In this Review, we survey advances in stem cell research and describe the cell types that are currently being used in the clinic or are close to clinical trials. Finally, we analyse the scientific rationale, experimental approaches, caveats and results underpinning the clinical use of such stem cells.
Collapse
Affiliation(s)
- Michele De Luca
- Center for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Graziella Pellegrini
- Center for Regenerative Medicine "Stefano Ferrari", Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Gehron Robey
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
27
|
Han S, Sun J, He S, Tang M, Chai R. The application of graphene-based biomaterials in biomedicine. Am J Transl Res 2019; 11:3246-3260. [PMID: 31312342 PMCID: PMC6614642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/16/2019] [Indexed: 06/10/2023]
Abstract
Graphene-based nanocomposites have attracted more and more attention recently in the field of biology and biomedicine. Graphene and its derivatives have been integrated with drugs, nucleic acids, antibodies, and other molecules. And these materials could be use as nanocomposite carriers or scaffold materials taking advantages of their enormous specific surface area, good elasticity and ductility, excellent biocompatibility, and outstanding mechanical strength. In addition, these composites have strong near-infrared absorbance and can act as photothermal agents to kill target cells through physical or chemical mechanisms. Along with significant advances in cell and organ transplantation, many of these materials have been explored in recent years for use in tissue engineering and regenerative medicine. Tissue engineering includes bone, nerve, heart, and muscle tissue engineering based on two-dimensional and three-dimensional graphene-based matrices or scaffolds possessing certain mechanical strengths and electrical conductivities, and the aim is to produce bioactive tissues to replace or repair natural tissue by promoting osteogenic, neuronal, and myogenic differentiation and myocardial cell growth. In this review, the basic properties of graphene-based complexes are systematically described and the biomedical applications of graphene-based materials in vivo and in vitro are summarized. This review first discusses the safety of graphene-based materials in terms of their biocompatibility and toxicity, and then it discusses these materials' applications in biosensing, photothermal therapy, stem cell culture, and tissue engineering. This review therefore provides a comprehensive understanding of graphene and its derivatives and their present and future applications.
Collapse
Affiliation(s)
- Shanying Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast UniversityNanjing 210096, China
| | - Jie Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast UniversityNanjing 210096, China
| | - Shuangba He
- Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast UniversityNanjing 211102, China
| | - Mingliang Tang
- MOE Key Laboratory for Developmental Genes and Human Disease, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast UniversityNanjing 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong UniversityNantong 226001, China
- Joint Research Institute of Southeast University and Monash UniversitySuzhou 215123, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast UniversityNanjing 210096, China
- Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast UniversityNanjing 211102, China
- MOE Key Laboratory for Developmental Genes and Human Disease, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast UniversityNanjing 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong UniversityNantong 226001, China
- Institute for Stem Cell and Regeneration, Chinese Academy of ScienceBeijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical UniversityBeijing 100069, China
- Joint Research Institute of Southeast University and Monash UniversitySuzhou 215123, China
| |
Collapse
|
28
|
Psarras S, Beis D, Nikouli S, Tsikitis M, Capetanaki Y. Three in a Box: Understanding Cardiomyocyte, Fibroblast, and Innate Immune Cell Interactions to Orchestrate Cardiac Repair Processes. Front Cardiovasc Med 2019; 6:32. [PMID: 31001541 PMCID: PMC6454035 DOI: 10.3389/fcvm.2019.00032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Following an insult by both intrinsic and extrinsic pathways, complex cellular, and molecular interactions determine a successful recovery or inadequate repair of damaged tissue. The efficiency of this process is particularly important in the heart, an organ characterized by very limited regenerative and repair capacity in higher adult vertebrates. Cardiac insult is characteristically associated with fibrosis and heart failure, as a result of cardiomyocyte death, myocardial degeneration, and adverse remodeling. Recent evidence implies that resident non-cardiomyocytes, fibroblasts but also macrophages -pillars of the innate immunity- form part of the inflammatory response and decisively affect the repair process following a cardiac insult. Multiple studies in model organisms (mouse, zebrafish) of various developmental stages (adult and neonatal) combined with genetically engineered cell plasticity and differentiation intervention protocols -mainly targeting cardiac fibroblasts or progenitor cells-reveal particular roles of resident and recruited innate immune cells and their secretome in the coordination of cardiac repair. The interplay of innate immune cells with cardiac fibroblasts and cardiomyocytes is emerging as a crucial platform to help our understanding and, importantly, to allow the development of effective interventions sufficient to minimize cardiac damage and dysfunction after injury.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
29
|
Zong H, Xia X, Liang Y, Dai S, Alsaedi A, Hayat T, Kong F, Pan JH. Designing function-oriented artificial nanomaterials and membranes via electrospinning and electrospraying techniques. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1075-1091. [DOI: 10.1016/j.msec.2017.11.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/27/2017] [Accepted: 11/11/2017] [Indexed: 12/16/2022]
|
30
|
Taylor DA, Frazier OH, Elgalad A, Hochman-Mendez C, Sampaio LC. Building a Total Bioartificial Heart: Harnessing Nature to Overcome the Current Hurdles. Artif Organs 2018; 42:970-982. [PMID: 30044011 DOI: 10.1111/aor.13336] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022]
Abstract
Engineering a bioartificial heart has become a possibility in part because of the regenerative medicine approaches to repairing or replacing damaged organs that have evolved over the past two decades. With the advent of inducible pluripotent stem cell technology, it is now possible to generate personalized cells that make the concept of autologous tissue engineering imaginable. Scaffolds that provide form, function, and biological cues to cells likewise potentially enable the engineering of biocompatible vascularized solid organs. Decellularized organs or tissue matrices retain organ complexity and structure at the macro and micro scales, contain biologically active molecules that support cell phenotype and function, and are vascularized allowing full thickness tissue generation. There is also dynamic reciprocity between the extracellular matrix and cells, which does not occur with synthetic scaffolds and allows both to evolve as functional need changes, making it a unique scaffold. Yet, building a whole heart from decellularized scaffolds and cells requires delivering hundreds of billions of multiple types of cardiac cells appropriately and providing a milieu where they can survive and mature. We propose a novel type of in vivo organ engineering utilizing pre-clinical models where decellularized hearts are heterotopically transplanted with the intent to harness the capability of the body to at least in part repopulate the scaffold. By adding load and electrical input, possibly via temporary mechanical assistance, we posit that vascular and parenchymal cell maturation can occur. In this study, we implanted porcine decellularized hearts acutely and chronically in living recipients in a heterotopic position. We demonstrated that the surgical procedure is critical to prevent coagulation and to increase graft patency. We also demonstrated that short-term implantation promotes endothelial cell adhesion to the vessel lumens and that long-term implantation also promotes tissue formation with evidence of cardiomyocytes and endothelial cells present within the graft. Utilizing endogenous repair capabilities of the recipient in response to a naked ECM, we allowed the transplanted scaffold to direct host cells-both organizationally and functionally. Thus, the scaffold provided necessary cues for cell organization and remodeling within the transplanted organ. Future work would involve culturing partially recellularized engineered organs in bioreactors where mechanical and electrical stimulation can be controlled to promote organ development and then transplanting these after a minimal level of maturation has been achieved.
Collapse
Affiliation(s)
- Doris A Taylor
- Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - O Howard Frazier
- Cullen Cardiovascular Surgery Research, Texas Heart Institute, Houston, TX, USA
| | | | | | - Luiz C Sampaio
- Cullen Cardiovascular Surgery Research, Texas Heart Institute, Houston, TX, USA
| |
Collapse
|
31
|
Guo X, Jiang H, Chen J, Zhang BF, Hu Q, Yang S, Yang J, Zhang J. RP105 ameliorates hypoxia̸reoxygenation injury in cardiac microvascular endothelial cells by suppressing TLR4̸MAPKs̸NF-κB signaling. Int J Mol Med 2018; 42:505-513. [PMID: 29693119 DOI: 10.3892/ijmm.2018.3621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 12/05/2017] [Indexed: 11/10/2022] Open
Abstract
The radioprotective 105 kDa protein (RP105) has been implicated in the pathological process of multiple cardiovascular diseases through its functional and physical interactions with Toll‑like receptor 4 (TLR4). However, the effects of RP105 on cardiac microvascular endothelial cells (CMECs) in response to hypoxia̸reoxygenation (H̸R) injury have not been extensively investigated. The aim of the present study was to elucidate the potential roles of RP105 in the protection of CMECs against H̸R injury, and investigate the underlying mechanisms. CMECs isolated from Sprague‑Dawley rats were transduced with adenoviral vectors encoding RP105 or green fluorescent protein (GFP). At 48 h post‑transfection, CMECs were subjected to hypoxia for 4 h and reoxygenation for 2 h (H̸R) to simulate the in vivo ischemia̸reperfusion model. The mRNA and protein levels of RP105 were detected by reverse transcription‑quantitative polymerase chain reaction and western blot analysis, respectively. The effects of RP105 on CMEC proliferation, migration and apoptosis were measured by GFP‑8, Transwell chamber and flow cytometry assays, respectively. The secretion of interleukin (IL)‑6 and tumor necrosis factor (TNF)‑α in the culture medium was measured by ELISA. Moreover, the expression level of TLR4, p38 mitogen‑activated protein kinase (MAPK), extracellular-signal-regulated kinase 1̸2, c-Jun N-terminal kinase, nuclear factor (NF)‑κB̸p65, IL‑6, TNF‑α and intercellular adhesion melecule‑1 was evaluated by western blot analysis. The results demonstrated that RP105 was minimally expressed in CMECs subjected to H̸R injury. Overexpression of RP105 via adenoviral vectors was able to significantly protect CMECs against H̸R injury, as evidenced by the promotion of cell proliferation and migration, as well as the amelioration of inflammation and apoptosis. These beneficial effects were at least partly mediated through inhibition of TLR4̸MAPKs̸NF‑κB signaling. Therefore, RP105 may be a promising candidate for prevention against CMECs‑associated H̸R injury.
Collapse
Affiliation(s)
- Xin Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo-Fang Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shuo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
32
|
Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, Yu ZX, Fisher JP, Mohiuddin M, Zhang LG. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv Drug Deliv Rev 2018; 132:252-269. [PMID: 30053441 PMCID: PMC6226324 DOI: 10.1016/j.addr.2018.07.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Compared to traditional therapeutic strategies, three-dimensional (3D) bioprinting is one of the most advanced techniques for creating complicated cardiovascular implants with biomimetic features, which are capable of recapitulating both the native physiochemical and biomechanical characteristics of the cardiovascular system. The present review provides an overview of the cardiovascular system, as well as describes the principles of, and recent advances in, 3D bioprinting cardiovascular tissues and models. Moreover, this review will focus on the applications of 3D bioprinting technology in cardiovascular repair/regeneration and pharmacological modeling, further discussing current challenges and perspectives.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, USA
| | | | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
33
|
Huang S, Yang Y, Yang Q, Zhao Q, Ye X. Engineered circulatory scaffolds for building cardiac tissue. J Thorac Dis 2018; 10:S2312-S2328. [PMID: 30123572 DOI: 10.21037/jtd.2017.12.92] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) is the terminal state of cardiovascular disease (CVD), leading numerous patients to death every year. Cardiac tissue engineering is a multidisciplinary field of creating functional cardiac patches in vitro to promote cardiac function after transplantation onto damaged zone, giving the hope for patients with end-stage HF. However, the limited thickness of cardiac patches results in the graft failure of survival and function due to insufficient blood supply. To date, prevascularized cardiac tissue, with the use of circulatory scaffolds, holds the promise to be inosculated and perfused with host vasculature to eventually promote cardiac pumping function. Circulatory scaffolds play its role to provide oxygen and nutrients and take metabolic wastes away, and achieve anastomosis with host vasculature in vivo. Of worth note, heart-on-a-chip based on circulatory scaffolds now has been considered as a valuable unit to broaden the research for building cardiac tissue. In this review, we will present recent different strategies to engineer circulatory scaffolds for building cardiac tissue with microvasculature, followed by its current state and future direction.
Collapse
Affiliation(s)
- Shixing Huang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yang Yang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qi Yang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qiang Zhao
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xiaofeng Ye
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
34
|
Fakoya AOJ, Otohinoyi DA, Yusuf J. Current Trends in Biomaterial Utilization for Cardiopulmonary System Regeneration. Stem Cells Int 2018; 2018:3123961. [PMID: 29853910 PMCID: PMC5949153 DOI: 10.1155/2018/3123961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/15/2017] [Accepted: 03/01/2018] [Indexed: 12/28/2022] Open
Abstract
The cardiopulmonary system is made up of the heart and the lungs, with the core function of one complementing the other. The unimpeded and optimal cycling of blood between these two systems is pivotal to the overall function of the entire human body. Although the function of the cardiopulmonary system appears uncomplicated, the tissues that make up this system are undoubtedly complex. Hence, damage to this system is undesirable as its capacity to self-regenerate is quite limited. The surge in the incidence and prevalence of cardiopulmonary diseases has reached a critical state for a top-notch response as it currently tops the mortality table. Several therapies currently being utilized can only sustain chronically ailing patients for a short period while they are awaiting a possible transplant, which is also not devoid of complications. Regenerative therapeutic techniques now appear to be a potential approach to solve this conundrum posed by these poorly self-regenerating tissues. Stem cell therapy alone appears not to be sufficient to provide the desired tissue regeneration and hence the drive for biomaterials that can support its transplantation and translation, providing not only physical support to seeded cells but also chemical and physiological cues to the cells to facilitate tissue regeneration. The cardiac and pulmonary systems, although literarily seen as just being functionally and spatially cooperative, as shown by their diverse and dissimilar adult cellular and tissue composition has been proven to share some common embryological codevelopment. However, necessitating their consideration for separate review is the immense adult architectural difference in these systems. This review also looks at details on new biological and synthetic biomaterials, tissue engineering, nanotechnology, and organ decellularization for cardiopulmonary regenerative therapies.
Collapse
Affiliation(s)
| | | | - Joshua Yusuf
- All Saints University School of Medicine, Roseau, Dominica
- All Saints University School of Medicine, Kingstown, Saint Vincent and the Grenadines
| |
Collapse
|
35
|
Ogle BM, Bursac N, Domian I, Huang NF, Menasché P, Murry CE, Pruitt B, Radisic M, Wu JC, Wu SM, Zhang J, Zimmermann WH, Vunjak-Novakovic G. Distilling complexity to advance cardiac tissue engineering. Sci Transl Med 2017; 8:342ps13. [PMID: 27280684 DOI: 10.1126/scitranslmed.aad2304] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The promise of cardiac tissue engineering is in the ability to recapitulate in vitro the functional aspects of a healthy heart and disease pathology as well as to design replacement muscle for clinical therapy. Parts of this promise have been realized; others have not. In a meeting of scientists in this field, five central challenges or "big questions" were articulated that, if addressed, could substantially advance the current state of the art in modeling heart disease and realizing heart repair.
Collapse
Affiliation(s)
- Brenda M Ogle
- Department of Biomedical Engineering, Stem Cell Institute, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Ibrahim Domian
- Harvard Medical School and Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA. Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Philippe Menasché
- Department of Cardiovascular Surgery, INSERM U 970, Hôpital Européen Georges Pompidou and University Paris Descartes, 75006 Paris, France
| | - Charles E Murry
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, Departments of Pathology, Bioengineering, and Medicine, University of Washington, Seattle, WA 98109, USA
| | - Beth Pruitt
- Departments of Mechanical Engineering and, by courtesy, Molecular and Cellular Physiology and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Joseph C Wu
- Stanford Cardiovascular Institute and Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sean M Wu
- Departments of Medicine and Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), partner site Göttingen, 37075 Göttingen, Germany
| | - Gordana Vunjak-Novakovic
- Departments of Biomedical Engineering and Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
36
|
Sommese L, Zullo A, Schiano C, Mancini FP, Napoli C. Possible Muscle Repair in the Human Cardiovascular System. Stem Cell Rev Rep 2017; 13:170-191. [PMID: 28058671 DOI: 10.1007/s12015-016-9711-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regenerative potential of tissues and organs could promote survival, extended lifespan and healthy life in multicellular organisms. Niches of adult stemness are widely distributed and lead to the anatomical and functional regeneration of the damaged organ. Conversely, muscular regeneration in mammals, and humans in particular, is very limited and not a single piece of muscle can fully regrow after a severe injury. Therefore, muscle repair after myocardial infarction is still a chimera. Recently, it has been recognized that epigenetics could play a role in tissue regrowth since it guarantees the maintenance of cellular identity in differentiated cells and, therefore, the stability of organs and tissues. The removal of these locks can shift a specific cell identity back to the stem-like one. Given the gradual loss of tissue renewal potential in the course of evolution, in the last few years many different attempts to retrieve such potential by means of cell therapy approaches have been performed in experimental models. Here we review pathways and mechanisms involved in the in vivo repair of cardiovascular muscle tissues in humans. Moreover, we address the ongoing research on mammalian cardiac muscle repair based on adult stem cell transplantation and pro-regenerative factor delivery. This latter issue, involving genetic manipulations of adult cells, paves the way for developing possible therapeutic strategies in the field of cardiovascular muscle repair.
Collapse
Affiliation(s)
- Linda Sommese
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.
| | - Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy.,CEINGE Advanced Biotechnologies, s.c.ar.l, Naples, Italy
| | | | - Francesco P Mancini
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Claudio Napoli
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.,IRCCS Foundation SDN, Naples, Italy
| |
Collapse
|
37
|
Human Induced Pluripotent Stem Cell-Derived Cardiomyocyte Encapsulating Bioactive Hydrogels Improve Rat Heart Function Post Myocardial Infarction. Stem Cell Reports 2017; 9:1415-1422. [PMID: 28988988 PMCID: PMC5830963 DOI: 10.1016/j.stemcr.2017.09.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering offers an exciting possibility for cardiac repair post myocardial infarction. We assessed the effects of combined polyethylene glycol hydrogel (PEG), human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM), and erythropoietin (EPO) therapy in a rat model of myocardial infarction. PEG with/out iPSC-CMs and EPO; iPSC-CMs in saline; or saline alone was injected into infarcted hearts shortly after infarction. Injection of almost any combination of the therapeutics limited acute elevations in chamber volumes. After 10 weeks, attenuation of ventricular remodeling was identified in all groups that received PEG injections, while ejection fractions were significantly increased in the gel-EPO, cell, and gel-cell-EPO groups. In all treatment groups, infarct thickness was increased and regions of muscle were identified within the scar. However, no grafted cells were detected. Hence, iPSC-CM-encapsulating bioactive hydrogel therapy can improve cardiac function post myocardial infarction and increase infarct thickness and muscle content despite a lack of sustained donor-cell engraftment. Hydrogels tailored to myocardial properties do not impair normal heart function Stem cell-encapsulating bioactive hydrogels can be delivered to infarcted hearts Bioactive hydrogels increased cardiac function, infarct thickness, and muscle content Cell-related benefits were observed even though donor cells did not stably engraft
Collapse
|
38
|
Chaudhuri R, Ramachandran M, Moharil P, Harumalani M, Jaiswal AK. Biomaterials and cells for cardiac tissue engineering: Current choices. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.121] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
39
|
González-Rosa JM, Burns CE, Burns CG. Zebrafish heart regeneration: 15 years of discoveries. ACTA ACUST UNITED AC 2017; 4:105-123. [PMID: 28979788 PMCID: PMC5617908 DOI: 10.1002/reg2.83] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Compared to other organs such as the liver, the adult human heart lacks the capacity to regenerate on a macroscopic scale after injury. As a result, myocardial infarctions are responsible for approximately half of all cardiovascular related deaths. In contrast, the zebrafish heart regenerates efficiently upon injury through robust myocardial proliferation. Therefore, deciphering the mechanisms that underlie the zebrafish heart's endogenous regenerative capacity represents an exciting avenue to identify novel therapeutic strategies for inducing regeneration of the human heart. This review provides a historical overview of adult zebrafish heart regeneration. We summarize 15 years of research, with a special focus on recent developments from this fascinating field. We discuss experimental findings that address fundamental questions of regeneration research. What is the origin of regenerated muscle? How is regeneration controlled from a genetic and molecular perspective? How do different cell types interact to achieve organ regeneration? Understanding natural models of heart regeneration will bring us closer to answering the ultimate question: how can we stimulate myocardial regeneration in humans?
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center Massachusetts General Hospital Charlestown MA 02129 USA.,Harvard Medical School Boston MA 02115 USA
| | - Caroline E Burns
- Cardiovascular Research Center Massachusetts General Hospital Charlestown MA 02129 USA.,Harvard Medical School Boston MA 02115 USA.,Harvard Stem Cell Institute Cambridge MA 02138 USA
| | - C Geoffrey Burns
- Cardiovascular Research Center Massachusetts General Hospital Charlestown MA 02129 USA.,Harvard Medical School Boston MA 02115 USA
| |
Collapse
|
40
|
Choi YJ, Yi HG, Kim SW, Cho DW. 3D Cell Printed Tissue Analogues: A New Platform for Theranostics. Theranostics 2017; 7:3118-3137. [PMID: 28839468 PMCID: PMC5566110 DOI: 10.7150/thno.19396] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/29/2017] [Indexed: 01/08/2023] Open
Abstract
Stem cell theranostics has received much attention for noninvasively monitoring and tracing transplanted therapeutic stem cells through imaging agents and imaging modalities. Despite the excellent regenerative capability of stem cells, their efficacy has been limited due to low cellular retention, low survival rate, and low engraftment after implantation. Three-dimensional (3D) cell printing provides stem cells with the similar architecture and microenvironment of the native tissue and facilitates the generation of a 3D tissue-like construct that exhibits remarkable regenerative capacity and functionality as well as enhanced cell viability. Thus, 3D cell printing can overcome the current concerns of stem cell therapy by delivering the 3D construct to the damaged site. Despite the advantages of 3D cell printing, the in vivo and in vitro tracking and monitoring of the performance of 3D cell printed tissue in a noninvasive and real-time manner have not been thoroughly studied. In this review, we explore the recent progress in 3D cell technology and its applications. Finally, we investigate their potential limitations and suggest future perspectives on 3D cell printing and stem cell theranostics.
Collapse
Affiliation(s)
- Yeong-Jin Choi
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-781, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-781, Republic of Korea
| | - Seok-Won Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-781, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-781, Republic of Korea
| |
Collapse
|
41
|
Mochizuki N, Pearson JT, Kitamura S. Beyond proof of concepts for ideal cardiac regenerative therapy. J Thorac Cardiovasc Surg 2017. [PMID: 28645824 DOI: 10.1016/j.jtcvs.2017.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - James T Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Soichiro Kitamura
- Department of Cardiac Surgery, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.
| |
Collapse
|
42
|
Akintewe OO, Roberts EG, Rim NG, Ferguson MA, Wong JY. Design Approaches to Myocardial and Vascular Tissue Engineering. Annu Rev Biomed Eng 2017; 19:389-414. [DOI: 10.1146/annurev-bioeng-071516-044641] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Olukemi O. Akintewe
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215;, ,
| | - Erin G. Roberts
- Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215;,
| | - Nae-Gyune Rim
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215;, ,
| | - Michael A.H. Ferguson
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215;, ,
| | - Joyce Y. Wong
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215;, ,
- Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215;,
| |
Collapse
|
43
|
Fibroblasts as maestros orchestrating tissue regeneration. J Tissue Eng Regen Med 2017; 12:240-251. [DOI: 10.1002/term.2405] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 12/05/2016] [Accepted: 01/09/2017] [Indexed: 12/12/2022]
|
44
|
Shahabipour F, Banach M, Johnston TP, Pirro M, Sahebkar A. Novel approaches toward the generation of bioscaffolds as a potential therapy in cardiovascular tissue engineering. Int J Cardiol 2017; 228:319-326. [DOI: 10.1016/j.ijcard.2016.11.210] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/06/2016] [Indexed: 12/18/2022]
|
45
|
Cho HM, Kim PH, Chang HK, Shen YM, Bonsra K, Kang BJ, Yum SY, Kim JH, Lee SY, Choi MC, Kim HH, Jang G, Cho JY. Targeted Genome Engineering to Control VEGF Expression in Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells: Potential Implications for the Treatment of Myocardial Infarction. Stem Cells Transl Med 2017; 6:1040-1051. [PMID: 28186692 PMCID: PMC5442764 DOI: 10.1002/sctm.16-0114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/11/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022] Open
Abstract
Human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs) exhibit potency for the regeneration of infarcted hearts. Vascular endothelial growth factor (VEGF) is capable of inducing angiogenesis and can boost stem cell‐based therapeutic effects. However, high levels of VEGF can cause abnormal blood vessel growth and hemangiomas. Thus, a controllable system to induce therapeutic levels of VEGF is required for cell therapy. We generated an inducible VEGF‐secreting stem cell (VEGF/hUCB‐MSC) that controls the expression of VEGF and tested the therapeutic efficacy in rat myocardial infarction (MI) model to apply functional stem cells to MI. To introduce the inducible VEGF gene cassette into a safe harbor site of the hUCB‐MSC chromosome, the transcription activator‐like effector nucleases system was used. After confirming the integration of the cassette into the locus, VEGF secretion in physiological concentration from VEGF/hUCB‐MSCs after doxycycline (Dox) induction was proved in conditioned media. VEGF secretion was detected in mice implanted with VEGF/hUCB‐MSCs grown via a cell sheet system. Vessel formation was induced in mice transplanted with Matrigel containing VEGF/hUCB‐MSCs treated with Dox. Moreover, seeding of the VEGF/hUCB‐MSCs onto the cardiac patch significantly improved the left ventricle ejection fraction and fractional shortening in a rat MI model upon VEGF induction. Induced VEGF/hUCB‐MSC patches significantly decreased the MI size and fibrosis and increased muscle thickness, suggesting improved survival of cardiomyocytes and protection from MI damage. These results suggest that our inducible VEGF‐secreting stem cell system is an effective therapeutic approach for the treatment of MI. Stem Cells Translational Medicine2017;6:1040–1051
Collapse
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Pyung-Hwan Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Kyung Chang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yi-Ming Shen
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwaku Bonsra
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Byung-Jae Kang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - So-Yeong Lee
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Min-Cheol Choi
- Department of Veterinary Radiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
46
|
Jang J, Park HJ, Kim SW, Kim H, Park JY, Na SJ, Kim HJ, Park MN, Choi SH, Park SH, Kim SW, Kwon SM, Kim PJ, Cho DW. 3D printed complex tissue construct using stem cell-laden decellularized extracellular matrix bioinks for cardiac repair. Biomaterials 2017; 112:264-274. [DOI: 10.1016/j.biomaterials.2016.10.026] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 12/13/2022]
|
47
|
Liu Y, Lu J, Xu G, Wei J, Zhang Z, Li X. Tuning the conductivity and inner structure of electrospun fibers to promote cardiomyocyte elongation and synchronous beating. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:865-74. [DOI: 10.1016/j.msec.2016.07.069] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/16/2016] [Accepted: 07/26/2016] [Indexed: 01/25/2023]
|
48
|
Kim JH, Joo HJ, Kim M, Choi SC, Lee JI, Hong SJ, Lim DS. Transplantation of Adipose-Derived Stem Cell Sheet Attenuates Adverse Cardiac Remodeling in Acute Myocardial Infarction. Tissue Eng Part A 2016; 23:1-11. [PMID: 27676105 DOI: 10.1089/ten.tea.2016.0023] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Adipose-derived stem cell (ADSC) transplantation has been proposed to improve cardiac function and acute myocardial infarction (AMI). Recently, cell sheet technology has been investigated for its potential applicability in cardiac injury. However, a detailed comparison of the functional recovery in the injured myocardium between cell sheets and conventional cell injection has not been adequately examined. ADSCs were isolated from the inguinal fat tissue of ICR mice. Three groups of AMI induction only (sham), intramyocardial injection of ADSCs (imADSC), and ADSC sheet transplantation (shADSC) were compared by using rat AMI models. Engraftment of ADSCs was better sustained through 28 days in the shADSC group compared with the imADSC group. Ejection fraction was improved in both imADSC and shADSC groups compared with the sham group. Ventricular wall thickness in the infarct zone was higher in the shADSC group compared with both imADSC and sham groups. Growth factor and cytokine expression in the implanted heart tissue were higher in the shADSC group compared with both imADSC and sham groups. Furthermore, only the shADSC group showed donor-derived vessels at the peri-infarct zone. Taken together, these results indicate that, although shADSC resulted in a similar improvement in left ventricular systolic function, it significantly promoted cellular engraftment and upregulated growth factor and cytokine expression, and, ultimately, attenuated adverse cardiac remodeling in rat AMI models compared with imADSC.
Collapse
Affiliation(s)
- Jong-Ho Kim
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| | - Hyung Joon Joo
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| | - Mina Kim
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| | - Seung-Cheol Choi
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| | - Jeong Ik Lee
- 2 Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine and Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science & Technology (IBST), Konkuk University , Seoul, South Korea
| | - Soon Jun Hong
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| | - Do-Sun Lim
- 1 Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University , Seoul, South Korea
| |
Collapse
|
49
|
Riemenschneider SB, Mattia DJ, Wendel JS, Schaefer JA, Ye L, Guzman PA, Tranquillo RT. Inosculation and perfusion of pre-vascularized tissue patches containing aligned human microvessels after myocardial infarction. Biomaterials 2016; 97:51-61. [PMID: 27162074 PMCID: PMC4891978 DOI: 10.1016/j.biomaterials.2016.04.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/15/2016] [Accepted: 04/20/2016] [Indexed: 12/18/2022]
Abstract
A major goal of tissue engineering is the creation of pre-vascularized tissues that have a high density of organized microvessels that can be rapidly perfused following implantation. This is especially critical for highly metabolic tissues like myocardium, where a thick myocardial engineered tissue would require rapid perfusion within the first several days to survive transplantation. In the present work, tissue patches containing human microvessels that were either randomly oriented or aligned were placed acutely on rat hearts post-infarction and for each case it was determined whether rapid inosculation could occur and perfusion of the patch could be maintained for 6 days in an infarct environment. Patches containing self-assembled microvessels were formed by co-entrapment of human blood outgrowth endothelial cells and human pericytes in fibrin gel. Cell-induced gel contraction was mechanically-constrained resulting in samples with high densities of microvessels that were either randomly oriented (with 420 ± 140 lumens/mm(2)) or uniaxially aligned (with 940 ± 240 lumens/mm(2)) at the time of implantation. These patches were sutured onto the epicardial surface of the hearts of athymic rats following permanent ligation of the left anterior descending artery. In both aligned and randomly oriented microvessel patches, inosculation occurred and perfusion of the transplanted human microvessels was maintained, proving the in vivo vascularization potential of these engineered tissues. No difference was found in the number of human microvessels that were perfused in the randomly oriented (111 ± 75 perfused lumens/mm(2)) and aligned (173 ± 97 perfused lumens/mm(2)) patches. Our results demonstrate that tissue patches containing a high density of either aligned or randomly oriented human pre-formed microvessels achieve rapid perfusion in the myocardial infarct environment - a necessary first-step toward the creation of a thick, perfusable heart patch.
Collapse
Affiliation(s)
- Sonja B Riemenschneider
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Donald J Mattia
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Jacqueline S Wendel
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Jeremy A Schaefer
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Lei Ye
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Pilar A Guzman
- Integrative Biology and Physiology Phenotyping Core, University of Minnesota, Minneapolis, MN, USA
| | - Robert T Tranquillo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
50
|
Malheiro A, Wieringa P, Mota C, Baker M, Moroni L. Patterning Vasculature: The Role of Biofabrication to Achieve an Integrated Multicellular Ecosystem. ACS Biomater Sci Eng 2016; 2:1694-1709. [DOI: 10.1021/acsbiomaterials.6b00269] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Afonso Malheiro
- Department
of Complex Tissue
Regeneration, MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Paul Wieringa
- Department
of Complex Tissue
Regeneration, MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Carlos Mota
- Department
of Complex Tissue
Regeneration, MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew Baker
- Department
of Complex Tissue
Regeneration, MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department
of Complex Tissue
Regeneration, MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|