1
|
Mukasheva F, Adilova L, Dyussenbinov A, Yernaimanova B, Abilev M, Akilbekova D. Optimizing scaffold pore size for tissue engineering: insights across various tissue types. Front Bioeng Biotechnol 2024; 12:1444986. [PMID: 39600888 PMCID: PMC11588461 DOI: 10.3389/fbioe.2024.1444986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Scaffold porosity is a critical factor in replicating the complex in vivo microenvironment, directly influencing cellular interactions, migration, nutrient transfer, vascularization, and the formation of functional tissues. For optimal tissue formation, scaffold design must account for various parameters, including material composition, morphology, mechanical properties, and cellular compatibility. This review highlights the importance of interconnected porosity and pore size, emphasizing their impact on cellular behavior and tissue formation across several tissue engineering domains, such as skin, bone, cardiovascular, and lung tissues. Specific pore size ranges enhance scaffold functionality for different tissues: small pores (∼1-2 µm) aid epidermal cell attachment in skin regeneration, moderate pores (∼2-12 µm) support dermal migration, and larger pores (∼40-100 µm) facilitate vascular structures. For bone tissue engineering, multi-layered scaffolds with smaller pores (50-100 µm) foster cell attachment, while larger pores (200-400 µm) enhance nutrient diffusion and angiogenesis. Cardiovascular and lung tissues benefit from moderate pore sizes (∼25-60 µm) to balance cell integration and nutrient diffusion. By addressing critical design challenges and optimizing pore size distributions, this review provides insights into scaffold innovations, ultimately advancing tissue regeneration strategies.
Collapse
Affiliation(s)
- Fariza Mukasheva
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Laura Adilova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Aibek Dyussenbinov
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Bota Yernaimanova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Madi Abilev
- Department of Analytical, Colloid Chemistry and Technology of Rare Elements, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Dana Akilbekova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
2
|
Lai P, Sheng M, Ye JH, Tang ZX, Hu S, Wang B, Yuan JL, Yang YH, Zhong YM, Liao YL. Research trends in cardiovascular tissue engineering from 1992 to 2022: a bibliometric analysis. Front Cardiovasc Med 2023; 10:1208227. [PMID: 37593146 PMCID: PMC10427867 DOI: 10.3389/fcvm.2023.1208227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Background Cardiovascular tissue engineering (CTE) is a promising technique to treat incurable cardiovascular diseases, such as myocardial infarction and ischemic cardiomyopathy. Plenty of studies related to CTE have been published in the last 30 years. However, an analysis of the research status, trends, and potential directions in this field is still lacking. The present study applies a bibliometric analysis to reveal CTE research trends and potential directions. Methods On 5 August 2022, research articles and review papers on CTE were searched from the Web of Science Core Collection with inclusion and exclusion criteria. Publication trends, research directions, and visual maps in this field were obtained using Excel (Microsoft 2009), VOSviewer, and Citespace software. Results A total of 2,273 documents from 1992 to 2022 were included in the final analysis. Publications on CTE showed an upward trend from 1992 [number of publications (Np):1] to 2021 (Np:165). The United States (Np: 916, number of citations: 152,377, H-index: 124) contributed the most publications and citations in this field. Research on CTE has a wide distribution of disciplines, led by engineering (Np: 788, number of citations: 40,563, H-index: 105). "Functional maturation" [red cluster, average published year (APY): 2018.63, 30 times], "cell-derived cardiomyocytes" (red cluster, APY: 2018.43, 46 times), "composite scaffolds" (green cluster, APY: 2018.54, 41 times), and "maturation" (red cluster, APY: 2018.17, 84 times) are the main emerging keywords in this area. Conclusion Research on CTE is a hot research topic. The United States is a dominant player in CTE research. Interdisciplinary collaboration has played a critical role in the progress of CTE. Studies on functional maturation and the development of novel biologically relevant materials and related applications will be the potential research directions in this field.
Collapse
Affiliation(s)
- Ping Lai
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Ming Sheng
- Department of Library, Gannan Medical University, Ganzhou, China
| | - Jin-hua Ye
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Zhi-xian Tang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shuo Hu
- Department of Heart Medical Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Bei Wang
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
| | - Jing-lin Yuan
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
| | - Yi-hong Yang
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
| | - Yi-ming Zhong
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yong-ling Liao
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Brimmer S, Ji P, Birla AK, Keswani SG, Caldarone CA, Birla RK. Recent advances in biological pumps as a building block for bioartificial hearts. Front Bioeng Biotechnol 2023; 11:1061622. [PMID: 36741765 PMCID: PMC9895798 DOI: 10.3389/fbioe.2023.1061622] [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: 10/04/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
The field of biological pumps is a subset of cardiac tissue engineering and focused on the development of tubular grafts that are designed generate intraluminal pressure. In the simplest embodiment, biological pumps are tubular grafts with contractile cardiomyocytes on the external surface. The rationale for biological pumps is a transition from planar 3D cardiac patches to functional biological pumps, on the way to complete bioartificial hearts. Biological pumps also have applications as a standalone device, for example, to support the Fontan circulation in pediatric patients. In recent years, there has been a lot of progress in the field of biological pumps, with innovative fabrication technologies. Examples include the use of cell sheet engineering, self-organized heart muscle, bioprinting and in vivo bio chambers for vascularization. Several materials have been tested for biological pumps and included resected aortic segments from rodents, type I collagen, and fibrin hydrogel, to name a few. Multiple bioreactors have been tested to condition biological pumps and replicate the complex in vivo environment during controlled in vitro culture. The purpose of this article is to provide an overview of the field of the biological pumps, outlining progress in the field over the past several years. In particular, different fabrication methods, biomaterial platforms for tubular grafts and examples of bioreactors will be presented. In addition, we present an overview of some of the challenges that need to be overcome for the field of biological pumps to move forward.
Collapse
Affiliation(s)
- Sunita Brimmer
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Pengfei Ji
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Aditya K. Birla
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States
| | - Sundeep G. Keswani
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Christopher A. Caldarone
- Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Ravi K. Birla
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States,*Correspondence: Ravi K. Birla,
| |
Collapse
|
4
|
Yang C, Zhu C, Li Y, Li Z, Zhang Z, Xu J, Chen M, Li R, Liu S, Wu Y, Huang Z, Wu C. Injectable selenium-containing polymeric hydrogel formulation for effective treatment of myocardial infarction. Front Bioeng Biotechnol 2022; 10:912562. [PMID: 36032710 PMCID: PMC9403312 DOI: 10.3389/fbioe.2022.912562] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/08/2022] [Indexed: 12/04/2022] Open
Abstract
Myocardial infarction (MI) is a serious threat to people’s life and health, which is significantly hindered by effective treatment formulations. Interestingly, our recent endeavour of designing selenium-containing polymeric hydrogel has been experimentally proved to be helpful in combating inflammatory responses and treating MI. The design was inspired by selenium with anti-inflammatory and anti-fibrosis activities, and the formulation could also serve as a support of myocardial tissue upon the failure of this function. In details, an injectable selenium-containing polymeric hydrogel, namely, poly[di-(1-hydroxylyndecyl) selenide/polypropylene glycol/polyethylene glycol urethane] [poly(DH-SE/PEG/PPG urethane)], was synthesised by combining a thermosensitive PPG block, DH-Se (which has oxidation-reduction properties), and hydrophilic PEG segments. Based on the established mouse model of MI, this formulation was experimentally validated to effectively promote the recovery of cardiac function. At the same time, we confirmed by enzyme-linked immunosorbent assay, Masson staining and Western blotting that this formulation could inhibit inflammation and fibrosis, so as to significantly improve left ventricular remodelling. In summary, a selenium-containing polymeric hydrogel formulation analysed in the current study could be a promising therapeutic formulation, which can provide new strategies towards the effective treatment of myocardial infarction or even other inflammatory diseases.
Collapse
Affiliation(s)
- Cui Yang
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chunyan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yanling Li
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore.,Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore
| | - Zhenghao Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Jiajia Xu
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Runjing Li
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Shixiao Liu
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Yunlong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, School of Medicine, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
5
|
Possible Treatment of Myocardial Infarct Based on Tissue Engineering Using a Cellularized Solid Collagen Scaffold Functionalized with Arg-Glyc-Asp (RGD) Peptide. Int J Mol Sci 2021; 22:ijms222212563. [PMID: 34830447 PMCID: PMC8620820 DOI: 10.3390/ijms222212563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.
Collapse
|
6
|
Li X, Zhang W, Zhang C, Wo Y, Ma A, Li Y, Zhang X. The role of bFGF in preventing the shrinkage of cardiac progenitor cell-engineered conduction tissue by downregulating α-SMA expression. Life Sci 2021; 282:119794. [PMID: 34237312 DOI: 10.1016/j.lfs.2021.119794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 11/30/2022]
Abstract
AIMS Engineered conduction tissues (ECTs) fabricated from cardiac progenitor cells (CPCs) and collagen sponges were precisely targeted for the treatment of atrioventricular conduction block in our previous studies. However, obvious shrinkage and deformation of ECTs was observed during in vitro culture. According to the literature, it can be speculated that basic fibroblast growth factor (bFGF) may downregulate alpha-smooth muscle actin (α-SMA) produced by CPCs to prevent the shrinkage of CPC-engineered conduction tissues. MAIN METHODS In this study, culture media with or without bFGF were used for both cell culture and 3D tissue construction. The expression of α-SMA and the size change of engineered tissue were analyzed to evaluate the feasibility of adding bFGF to regulate α-SMA expression and shrinkage of constructs. In addition, cardiac-specific examinations were performed to evaluate the effect of bFGF on cardiac tissue formation. KEY FINDINGS Supplementation with bFGF efficiently relieved shrinkage of engineered tissue by downregulating the expression of α-SMA at both the cellular and 3D tissue levels. Moreover, bFGF had a positive influence on cardiac tissue formation in terms of cell viability, tissue organization and electrical conduction velocity. SIGNIFICANCE This study provides a guide for both shape control and quality improvement of CPC-engineered cardiac tissues.
Collapse
Affiliation(s)
- Xiaotong Li
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Anatomy, Naval Medical University, Shanghai, China
| | - Wenbo Zhang
- Rheumatology Department of Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chuansen Zhang
- Department of Anatomy, Naval Medical University, Shanghai, China
| | - Yan Wo
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Airong Ma
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Zhang
- Department of Anatomy, Naval Medical University, Shanghai, China.
| |
Collapse
|
7
|
Tadevosyan K, Iglesias-García O, Mazo MM, Prósper F, Raya A. Engineering and Assessing Cardiac Tissue Complexity. Int J Mol Sci 2021; 22:ijms22031479. [PMID: 33540699 PMCID: PMC7867236 DOI: 10.3390/ijms22031479] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiac tissue engineering is very much in a current focus of regenerative medicine research as it represents a promising strategy for cardiac disease modelling, cardiotoxicity testing and cardiovascular repair. Advances in this field over the last two decades have enabled the generation of human engineered cardiac tissue constructs with progressively increased functional capabilities. However, reproducing tissue-like properties is still a pending issue, as constructs generated to date remain immature relative to native adult heart. Moreover, there is a high degree of heterogeneity in the methodologies used to assess the functionality and cardiac maturation state of engineered cardiac tissue constructs, which further complicates the comparison of constructs generated in different ways. Here, we present an overview of the general approaches developed to generate functional cardiac tissues, discussing the different cell sources, biomaterials, and types of engineering strategies utilized to date. Moreover, we discuss the main functional assays used to evaluate the cardiac maturation state of the constructs, both at the cellular and the tissue levels. We trust that researchers interested in developing engineered cardiac tissue constructs will find the information reviewed here useful. Furthermore, we believe that providing a unified framework for comparison will further the development of human engineered cardiac tissue constructs displaying the specific properties best suited for each particular application.
Collapse
Affiliation(s)
- Karine Tadevosyan
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Center for Networked Biomedical Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| |
Collapse
|
8
|
Melby JA, de Lange WJ, Zhang J, Roberts DS, Mitchell SD, Tucholski T, Kim G, Kyrvasilis A, McIlwain SJ, Kamp TJ, Ralphe JC, Ge Y. Functionally Integrated Top-Down Proteomics for Standardized Assessment of Human Induced Pluripotent Stem Cell-Derived Engineered Cardiac Tissues. J Proteome Res 2021; 20:1424-1433. [PMID: 33395532 DOI: 10.1021/acs.jproteome.0c00830] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Three-dimensional (3D) human induced pluripotent stem cell-derived engineered cardiac tissues (hiPSC-ECTs) have emerged as a promising alternative to two-dimensional hiPSC-cardiomyocyte monolayer systems because hiPSC-ECTs are a closer representation of endogenous cardiac tissues and more faithfully reflect the relevant cardiac pathophysiology. The ability to perform functional and molecular assessments using the same hiPSC-ECT construct would allow for more reliable correlation between observed functional performance and underlying molecular events, and thus is critically needed. Herein, for the first time, we have established an integrated method that permits sequential assessment of functional properties and top-down proteomics from the same single hiPSC-ECT construct. We quantitatively determined the differences in isometric twitch force and the sarcomeric proteoforms between two groups of hiPSC-ECTs that differed in the duration of time of 3D-ECT culture. Importantly, by using this integrated method we discovered a new and strong correlation between the measured contractile parameters and the phosphorylation levels of alpha-tropomyosin between the two groups of hiPSC-ECTs. The integration of functional assessments together with molecular characterization by top-down proteomics in the same hiPSC-ECT construct enables a holistic analysis of hiPSC-ECTs to accelerate their applications in disease modeling, cardiotoxicity, and drug discovery. Data are available via ProteomeXchange with identifier PXD022814.
Collapse
Affiliation(s)
- Jake A Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Willem J de Lange
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianhua Zhang
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Stanford D Mitchell
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Trisha Tucholski
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Gina Kim
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Andreas Kyrvasilis
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Sean J McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Human Proteomics Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
9
|
Helfer A, Bursac N. Frame-Hydrogel Methodology for Engineering Highly Functional Cardiac Tissue Constructs. Methods Mol Biol 2021; 2158:171-186. [PMID: 32857373 PMCID: PMC7672525 DOI: 10.1007/978-1-0716-0668-1_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Engineered cardiac tissues hold tremendous promise for in vitro drug discovery, studies of heart development and disease, and therapeutic applications. Here, we describe a versatile "frame-hydrogel" methodology to generate engineered cardiac tissues with highly mature functional properties. This methodology has been successfully utilized with a variety of cell sources (neonatal rat ventricular myocytes, human and mouse pluripotent stem cell-derived cardiomyocytes) to generate tissues with diverse 3D geometries (patch, bundle, network) and levels of structural and functional anisotropy. Maturation of such engineered cardiac tissues is rapidly achieved without the need for exogenous electrical or mechanical stimulation or use of complex bioreactors, with tissues routinely reaching conduction velocities and specific forces of 25 cm/s and 20 mN/mm2, respectively, and forces per input cardiomyocyte of up to 12 nN. This method is reproducible and readily scalable to generate small tissues ideal for in vitro testing as well as tissues with large, clinically relevant dimensions.
Collapse
Affiliation(s)
- Abbigail Helfer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
10
|
Allen AC, Barone E, Momtahan N, Crosby CO, Tu C, Deng W, Polansky K, Zoldan J. Temporal Impact of Substrate Anisotropy on Differentiating Cardiomyocyte Alignment and Functionality. Tissue Eng Part A 2019; 25:1426-1437. [PMID: 30727863 PMCID: PMC6939589 DOI: 10.1089/ten.tea.2018.0258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/29/2019] [Indexed: 01/14/2023] Open
Abstract
Anisotropic biomaterials can affect cell function by driving cell alignment, which is critical for cardiac engineered tissues. Recent work, however, has shown that pluripotent stem cell-derived cardiomyocytes may self-align over long periods of time. To determine how the degree of biomaterial substrate anisotropy impacts differentiating cardiomyocyte structure and function, we differentiated mouse embryonic stem cells to cardiomyocytes on nonaligned, semialigned, and aligned fibrous substrates and evaluated cell alignment, contractile displacement, and calcium transient synchronicity over time. Although cardiomyocyte gene expression was not affected by fiber alignment, we observed gradient- and threshold-based differences in cardiomyocyte alignment and function. Cardiomyocyte alignment increased with the degree of fiber alignment in a gradient-based manner at early time points and in a threshold-based manner at later time points. Calcium transient synchronization tightly followed cardiomyocyte alignment behavior, allowing highly anisotropic biomaterials to drive calcium transient synchronization within 8 days, while such synchronized cardiomyocyte behavior required 20 days of culture on nonaligned biomaterials. In contrast, cardiomyocyte contractile displacement had no directional preference on day 8 yet became anisotropic in the direction of fiber alignment on aligned fibers by day 20. Biomaterial anisotropy impact on differentiating cardiomyocyte structure and function is temporally dependent. Impact Statement This work demonstrates that biomaterial anisotropy can quickly drive desired pluripotent stem cell-derived cardiomyocyte structure and function. Such an understanding of matrix anisotropy's time-dependent influence on stem cell-derived cardiomyocyte function will have future applications in the development of cardiac cell therapies and in vitro cardiac tissues for drug testing. Furthermore, our work has broader implications concerning biomaterial anisotropy effects on other cell types in which function relies on alignment, such as myocytes and neurons.
Collapse
Affiliation(s)
- Alicia C.B. Allen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Elissa Barone
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Nima Momtahan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Cody O. Crosby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Chengyi Tu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Wei Deng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Krista Polansky
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
11
|
Meyers CA, Xu J, Zhang L, Chang L, Wang Y, Asatrian G, Ding C, Yan N, Zou E, Broderick K, Lee M, Peault B, James AW. Skeletogenic Capacity of Human Perivascular Stem Cells Obtained Via Magnetic-Activated Cell Sorting. Tissue Eng Part A 2019; 25:1658-1666. [PMID: 31020920 DOI: 10.1089/ten.tea.2019.0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesenchymal progenitor cell population defined by their perivascular residence. PSC are increasingly studied for their application in skeletal regenerative medicine. PSC from subcutaneous white adipose tissue are most commonly isolated via fluorescence-activated cell sorting (FACS), and defined as a bipartite population of CD146+CD34-CD31-CD45- pericytes and CD34+CD146-CD31-CD45- adventitial cells. FACS poses several challenges for clinical translation, including requirements for facilities, equipment, and personnel. The purpose of this study is to identify if magnetic-activated cell sorting (MACS) is a feasible method to derive PSC, and to determine if MACS-derived PSC are comparable to our previous experience with FACS-derived PSC. In brief, CD146+ pericytes and CD34+ adventitial cells were enriched from human lipoaspirate using a multistep column approach. Next, cell identity and purity were analyzed by flow cytometry. In vitro multilineage differentiation studies were performed with MACS-defined PSC subsets. Finally, in vivo application was performed in nonhealing calvarial bone defects in Scid mice. Results showed that human CD146+ pericytes and CD34+ adventitial cells may be enriched by MACS, with defined purity, anticipated cell surface marker expression, and capacity for multilineage differentiation. In vivo, MACS-derived PSC induce ossification of bone defects. These data document the feasibility of a MACS approach for the enrichment and application of PSC in the field of tissue engineering and regenerative medicine. Impact Statement Our findings suggest that perivascular stem/stromal cells, and in particular adventitial cells, may be isolated by magnetic-activated cell sorting and applied as an uncultured autologous stem cell therapy in a same-day setting for bone defect repair.
Collapse
Affiliation(s)
- Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Leititia Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning Province, P.R. China
| | - Leslie Chang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Greg Asatrian
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Catherine Ding
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Noah Yan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Erin Zou
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Broderick
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Min Lee
- School of Dentistry, University of California, Los Angeles, California
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California.,Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
12
|
Wragg NM, Burke L, Wilson SL. A critical review of current progress in 3D kidney biomanufacturing: advances, challenges, and recommendations. RENAL REPLACEMENT THERAPY 2019. [DOI: 10.1186/s41100-019-0218-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
13
|
Biomaterializing the promise of cardiac tissue engineering. Biotechnol Adv 2019; 42:107353. [PMID: 30794878 DOI: 10.1016/j.biotechadv.2019.02.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
Abstract
During an average individual's lifespan, the human heart pumps nearly 200 million liters of blood delivered by approximately 3 billion heartbeats. Therefore, it is not surprising that native myocardium under this incredible demand is extraordinarily complex, both structurally and functionally. As a result, successful engineering of adult-mimetic functional cardiac tissues is likely to require utilization of highly specialized biomaterials representative of the native extracellular microenvironment. There is currently no single biomaterial that fully recapitulates the architecture or the biochemical and biomechanical properties of adult myocardium. However, significant effort has gone toward designing highly functional materials and tissue constructs that may one day provide a ready source of cardiac tissue grafts to address the overwhelming burden of cardiomyopathic disease. In the near term, biomaterial-based scaffolds are helping to generate in vitro systems for querying the mechanisms underlying human heart homeostasis and disease and discovering new, patient-specific therapeutics. When combined with advances in minimally-invasive cardiac delivery, ongoing efforts will likely lead to scalable cell and biomaterial technologies for use in clinical practice. In this review, we describe recent progress in the field of cardiac tissue engineering with particular emphasis on use of biomaterials for therapeutic tissue design and delivery.
Collapse
|
14
|
Current Challenges and Emergent Technologies for Manufacturing Artificial Right Ventricle to Pulmonary Artery (RV-PA) Cardiac Conduits. Cardiovasc Eng Technol 2019; 10:205-215. [DOI: 10.1007/s13239-019-00406-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 02/05/2019] [Indexed: 01/12/2023]
|
15
|
Sharma D, Ferguson M, Kamp TJ, Zhao F. Constructing Biomimetic Cardiac Tissues: A Review of Scaffold Materials for Engineering Cardiac Patches. EMERGENT MATERIALS 2019; 2:181-191. [PMID: 33225220 PMCID: PMC7678685 DOI: 10.1007/s42247-019-00046-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/13/2019] [Indexed: 05/18/2023]
Abstract
Engineered cardiac patches (ECPs) hold great promise to repair ischemia-induced damages to the myocardium. Recent studies have provided robust technological advances in obtaining pure cardiac cell populations as well as various novel scaffold materials to generate engineered cardiac tissues that can significantly improve electrical and contractile functions of damaged myocardium. Given the significance in understanding the cellular and extracellular structural as well as compositional details of native human heart wall, in order to fabricate most suitable scaffold material for cardiac patches, herein, we have reviewed the structure of the human pericardium and heart wall as well as the compositional details of cardiac extracellular matrix (ECM). Moreover, several strategies to obtain cardiac-specific scaffold materials have been reviewed, including natural, synthetic and hybrid hydrogels, electrospun fibers, decellularized native tissues or whole organs, and scaffolds derived from engineered cell sheets. This review provides a comprehensive analysis of different scaffold materials for engineering cardiac tissues.
Collapse
Affiliation(s)
- Dhavan Sharma
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Morgan Ferguson
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Timothy J Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
16
|
Han Y, Yang W, Cui W, Yang K, Wang X, Chen Y, Deng L, Zhao Y, Jin W. Retracted Article: Development of functional hydrogels for heart failure. J Mater Chem B 2019; 7:1563-1580. [DOI: 10.1039/c8tb02591f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hydrogel-based approaches were reviewed for cardiac tissue engineering and myocardial regeneration in ischemia-induced heart failure, with an emphasis on functional studies, translational status, and clinical advancements.
Collapse
Affiliation(s)
- Yanxin Han
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Wenbo Yang
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases
- Shanghai Institute of Traumatology and Orthopaedics
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Ke Yang
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Xiaoqun Wang
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Yanjia Chen
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases
- Shanghai Institute of Traumatology and Orthopaedics
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Yuanjin Zhao
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases
- Shanghai Institute of Traumatology and Orthopaedics
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| | - Wei Jin
- Department of Cardiology
- Institute of Cardiovascular Diseases
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
| |
Collapse
|
17
|
Kim S, Cho AN, Min S, Kim S, Cho SW. Organoids for Advanced Therapeutics and Disease Models. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Suran Kim
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Ann-Na Cho
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Sungjin Min
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Sooyeon Kim
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| |
Collapse
|
18
|
Jackman C, Li H, Bursac N. Long-term contractile activity and thyroid hormone supplementation produce engineered rat myocardium with adult-like structure and function. Acta Biomater 2018; 78:98-110. [PMID: 30086384 DOI: 10.1016/j.actbio.2018.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
The field of cardiac tissue engineering has developed rapidly, but structural and functional immaturity of engineered heart tissues hinder their widespread use. Here, we show that a combination of low-rate (0.2 Hz) contractile activity and thyroid hormone (T3) supplementation significantly promote structural and functional maturation of engineered rat cardiac tissues ("cardiobundles"). The progressive maturation of cardiobundles during first 2 weeks of culture resulted in cell cycle exit and loss of spontaneous activity, which in longer culture yielded decreased contractile function. Maintaining a low level of contractile activity by 0.2 Hz pacing between culture weeks 3 and 5, combined with T3 treatment, yielded significant growth of cardiobundle and myocyte cross-sectional areas (by 68% and 32%, respectively), increased nuclei numbers (by 22%), improved twitch force (by 39%), shortened action potential duration (by 32%), polarized N-cadherin distribution, and switch from immature (slow skeletal) to mature (fast) cardiac troponin I isoform expression. Along with advanced functional output (conduction velocity 53.7 ± 0.8 cm/s, specific force 70.1 ± 5.8 mN/mm2), quantitative ultrastructural analyses revealed similar metrics and abundance of sarcomeres, T-tubules, M-bands, and intercalated disks compared to native age-matched (5-week) and adult (3-month) ventricular myocytes. Unlike 0.2 Hz regime, chronic 1 Hz pacing resulted in significant cardiomyocyte loss and formation of necrotic core despite the use of dynamic culture. Overall, our results demonstrate remarkable ultrastructural and functional maturation of neonatal rat cardiomyocytes in 3D culture and reveal importance of combined biophysical and hormonal inputs for in vitro engineering of adult-like myocardium. STATEMENT OF SIGNIFICANCE Compared to human stem cell-derived cardiomyocytes, neonatal rat ventricular myocytes show advanced maturation state which makes them suitable for in vitro studies of postnatal cardiac development. Still, maturation process from a neonatal to an adult cardiomyocyte has not been recapitulated in rodent cell cultures. Here, we show that low-frequency pacing and thyroid hormone supplementation of 3D engineered neonatal rat cardiac tissues synergistically yield significant increase in cell and tissue volume, robust formation of T-tubules and M-lines, improved sarcomere organization, and faster and more forceful contractions. To the best of our knowledge, 5-week old engineered cardiac tissues described in this study are the first that exhibit both ultrastructural and functional characteristics approaching or matching those of adult ventricular myocardium.
Collapse
|
19
|
Peña B, Laughter M, Jett S, Rowland TJ, Taylor MRG, Mestroni L, Park D. Injectable Hydrogels for Cardiac Tissue Engineering. Macromol Biosci 2018; 18:e1800079. [PMID: 29733514 PMCID: PMC6166441 DOI: 10.1002/mabi.201800079] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/27/2018] [Indexed: 12/21/2022]
Abstract
In light of the limited efficacy of current treatments for cardiac regeneration, tissue engineering approaches have been explored for their potential to provide mechanical support to injured cardiac tissues, deliver cardio-protective molecules, and improve cell-based therapeutic techniques. Injectable hydrogels are a particularly appealing system as they hold promise as a minimally invasive therapeutic approach. Moreover, injectable acellular alginate-based hydrogels have been tested clinically in patients with myocardial infarction (MI) and show preservation of the left ventricular (LV) indices and left ventricular ejection fraction (LVEF). This review provides an overview of recent developments that have occurred in the design and engineering of various injectable hydrogel systems for cardiac tissue engineering efforts, including a comparison of natural versus synthetic systems with emphasis on the ideal characteristics for biomimetic cardiac materials.
Collapse
Affiliation(s)
- Brisa Peña
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, 12700 E.19th Avenue, Bldg. P15, Aurora, CO, 80045, USA
| | - Melissa Laughter
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, CO, 80045, USA
| | - Susan Jett
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, 12700 E.19th Avenue, Bldg. P15, Aurora, CO, 80045, USA
| | - Teisha J Rowland
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, 12700 E.19th Avenue, Bldg. P15, Aurora, CO, 80045, USA
| | - Matthew R G Taylor
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, 12700 E.19th Avenue, Bldg. P15, Aurora, CO, 80045, USA
| | - Luisa Mestroni
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, 12700 E.19th Avenue, Bldg. P15, Aurora, CO, 80045, USA
| | - Daewon Park
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, CO, 80045, USA
| |
Collapse
|
20
|
Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 2018; 13:27. [PMID: 29788997 PMCID: PMC5964712 DOI: 10.1186/s13024-018-0258-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS), affect millions of people every year and so far, there are no therapeutic cures available. Even though animal and histological models have been of great aid in understanding disease mechanisms and identifying possible therapeutic strategies, in order to find disease-modifying solutions there is still a critical need for systems that can provide more predictive and physiologically relevant results. One possible avenue is the development of patient-derived models, e.g. by reprogramming patient somatic cells into human induced pluripotent stem cells (hiPSCs), which can then be differentiated into any cell type for modelling. These systems contain key genetic information from the donors, and therefore have enormous potential as tools in the investigation of pathological mechanisms underlying disease phenotype, and progression, as well as in drug testing platforms. hiPSCs have been widely cultured in 2D systems, but in order to mimic human brain complexity, 3D models have been proposed as a more advanced alternative. This review will focus on the use of patient-derived hiPSCs to model AD, PD, HD and ALS. In brief, we will cover the available stem cells, types of 2D and 3D culture systems, existing models for neurodegenerative diseases, obstacles to model these diseases in vitro, and current perspectives in the field.
Collapse
Affiliation(s)
- Eduarda G Z Centeno
- Department of Biotechnology, Federal University of Pelotas, Campus Capão do Leão, Pelotas, RS, 96160-000, Brazil.,Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Helena Cimarosti
- Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Angela Bithell
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, RG6 6UB, UK.
| |
Collapse
|
21
|
Gao L, Gregorich ZR, Zhu W, Mattapally S, Oduk Y, Lou X, Kannappan R, Borovjagin AV, Walcott GP, Pollard AE, Fast VG, Hu X, Lloyd SG, Ge Y, Zhang J. Large Cardiac Muscle Patches Engineered From Human Induced-Pluripotent Stem Cell-Derived Cardiac Cells Improve Recovery From Myocardial Infarction in Swine. Circulation 2018; 137:1712-1730. [PMID: 29233823 PMCID: PMC5903991 DOI: 10.1161/circulationaha.117.030785] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Here, we generated human cardiac muscle patches (hCMPs) of clinically relevant dimensions (4 cm × 2 cm × 1.25 mm) by suspending cardiomyocytes, smooth muscle cells, and endothelial cells that had been differentiated from human induced-pluripotent stem cells in a fibrin scaffold and then culturing the construct on a dynamic (rocking) platform. METHODS In vitro assessments of hCMPs suggest maturation in response to dynamic culture stimulation. In vivo assessments were conducted in a porcine model of myocardial infarction (MI). Animal groups included: MI hearts treated with 2 hCMPs (MI+hCMP, n=13), MI hearts treated with 2 cell-free open fibrin patches (n=14), or MI hearts with neither experimental patch (n=15); a fourth group of animals underwent sham surgery (Sham, n=8). Cardiac function and infarct size were evaluated by MRI, arrhythmia incidence by implanted loop recorders, and the engraftment rate by calculation of quantitative polymerase chain reaction measurements of expression of the human Y chromosome. Additional studies examined the myocardial protein expression profile changes and potential mechanisms of action that related to exosomes from the cell patch. RESULTS The hCMPs began to beat synchronously within 1 day of fabrication, and after 7 days of dynamic culture stimulation, in vitro assessments indicated the mechanisms related to the improvements in electronic mechanical coupling, calcium-handling, and force generation, suggesting a maturation process during the dynamic culture. The engraftment rate was 10.9±1.8% at 4 weeks after the transplantation. The hCMP transplantation was associated with significant improvements in left ventricular function, infarct size, myocardial wall stress, myocardial hypertrophy, and reduced apoptosis in the periscar boarder zone myocardium. hCMP transplantation also reversed some MI-associated changes in sarcomeric regulatory protein phosphorylation. The exosomes released from the hCMP appeared to have cytoprotective properties that improved cardiomyocyte survival. CONCLUSIONS We have fabricated a clinically relevant size of hCMP with trilineage cardiac cells derived from human induced-pluripotent stem cells. The hCMP matures in vitro during 7 days of dynamic culture. Transplantation of this type of hCMP results in significantly reduced infarct size and improvements in cardiac function that are associated with reduction in left ventricular wall stress. The hCMP treatment is not associated with significant changes in arrhythmogenicity.
Collapse
Affiliation(s)
- Ling Gao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Zachery R Gregorich
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison (Z.R.G., Y.G.)
| | - Wuqiang Zhu
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Saidulu Mattapally
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Yasin Oduk
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Xi Lou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Ramaswamy Kannappan
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Anton V Borovjagin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Gregory P Walcott
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Andrew E Pollard
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Vladimir G Fast
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Xinyang Hu
- Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H.)
| | - Steven G Lloyd
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.)
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison (Z.R.G., Y.G.)
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (L.G., W.Z., S.M., Y.O., X.LO., R.K., A.V.B., G.P.W., A.E.P., V.G.F., S.G.L., J.Z.).
| |
Collapse
|
22
|
Applications of genetically engineered human pluripotent stem cell reporters in cardiac stem cell biology. Curr Opin Biotechnol 2018; 52:66-73. [PMID: 29579626 DOI: 10.1016/j.copbio.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Abstract
The advent of human pluripotent stem cells (hPSCs) has benefited many fields, from regenerative medicine to disease modeling, with an especially profound effect in cardiac research. Coupled with other novel technologies in genome engineering, hPSCs offer a great opportunity to delineate human cardiac lineages, investigate inherited cardiovascular diseases, and assess the safety and efficacy of cell-based therapies. In this review, we provide an overview of methods for generating genetically engineered hPSC reporters and a succinct synopsis of a variety of hPSC reporters, with a particular focus on their applications in cardiac stem cell biology.
Collapse
|
23
|
Slamecka J, McClellan S, Wilk A, Laurini J, Manci E, Hoerstrup SP, Weber B, Owen L. Induced pluripotent stem cells derived from human amnion in chemically defined conditions. Cell Cycle 2018; 17:330-347. [PMID: 29143560 DOI: 10.1080/15384101.2017.1403690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fetal stem cells are a unique type of adult stem cells that have been suggested to be broadly multipotent with some features of pluripotency. Their clinical potential has been documented but their upgrade to full pluripotency could open up a wide range of cell-based therapies particularly suited for pediatric tissue engineering, longitudinal studies or disease modeling. Here we describe episomal reprogramming of mesenchymal stem cells from the human amnion to pluripotency (AM-iPSC) in chemically defined conditions. The AM-iPSC expressed markers of embryonic stem cells, readily formed teratomas with tissues of all three germ layers present and had a normal karyotype after around 40 passages in culture. We employed novel computational methods to determine the degree of pluripotency from microarray and RNA sequencing data in these novel lines alongside an iPSC and ESC control and found that all lines were deemed pluripotent, however, with variable scores. Differential expression analysis then identified several groups of genes that potentially regulate this variability in lines within the boundaries of pluripotency, including metallothionein proteins. By further studying this variability, characteristics relevant to cell-based therapies, like differentiation propensity, could be uncovered and predicted in the pluripotent stage.
Collapse
Affiliation(s)
| | | | - Anna Wilk
- a Mitchell Cancer Institute, University of South Alabama , USA
| | - Javier Laurini
- c College of Medicine, University of South Alabama , Mobile , AL , USA
| | - Elizabeth Manci
- d College of Medicine, University of South Alabama , Mobile , AL , USA
| | - Simon P Hoerstrup
- b Institute for Regenerative Medicine, University of Zurich , Switzerland.,f Center for Applied Biotechnology and Molecular Medicine (CABMM) , University of Zurich - Irchel Campus , Zurich , Switzerland
| | - Benedikt Weber
- b Institute for Regenerative Medicine, University of Zurich , Switzerland.,e Department of Dermatology , University Hospital Zurich , Switzerland.,f Center for Applied Biotechnology and Molecular Medicine (CABMM) , University of Zurich - Irchel Campus , Zurich , Switzerland
| | - Laurie Owen
- g University of California San Diego , La Jolla , CA , USA
| |
Collapse
|
24
|
Mills RJ, Voges HK, Porrello ER, Hudson JE. Disease modeling and functional screening using engineered heart tissue. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Jackman CP, Ganapathi AM, Asfour H, Qian Y, Allen BW, Li Y, Bursac N. Engineered cardiac tissue patch maintains structural and electrical properties after epicardial implantation. Biomaterials 2018; 159:48-58. [PMID: 29309993 DOI: 10.1016/j.biomaterials.2018.01.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/17/2017] [Accepted: 01/01/2018] [Indexed: 12/22/2022]
Abstract
Functional cardiac tissue engineering holds promise as a candidate therapy for myocardial infarction and heart failure. Generation of "strong-contracting and fast-conducting" cardiac tissue patches capable of electromechanical coupling with host myocardium could allow efficient improvement of heart function without increased arrhythmogenic risks. Towards that goal, we engineered highly functional 1 cm × 1 cm cardiac tissue patches made of neonatal rat ventricular cells which after 2 weeks of culture exhibited force of contraction of 18.0 ± 1.4 mN, conduction velocity (CV) of 32.3 ± 1.8 cm/s, and sustained chronic activation when paced at rates as high as 8.7 ± 0.8 Hz. Patches transduced with genetically-encoded calcium indicator (GCaMP6) were implanted onto adult rat ventricles and after 4-6 weeks assessed for action potential conduction and electrical integration by two-camera optical mapping of GCaMP6-reported Ca2+ transients in the patch and RH237-reported action potentials in the recipient heart. Of the 13 implanted patches, 11 (85%) engrafted, maintained structural integrity, and conducted action potentials with average CVs and Ca2+ transient durations comparable to those before implantation. Despite preserved graft electrical properties, no anterograde or retrograde conduction could be induced between the patch and host cardiomyocytes, indicating lack of electrical integration. Electrical properties of the underlying myocardium were not changed by the engrafted patch. From immunostaining analyses, implanted patches were highly vascularized and expressed abundant electromechanical junctions, but remained separated from the epicardium by a non-myocyte layer. In summary, our studies demonstrate generation of highly functional cardiac tissue patches that can robustly engraft on the epicardial surface, vascularize, and maintain electrical function, but do not couple with host tissue. The lack of graft-host electrical integration is therefore a critical obstacle to development of efficient tissue engineering therapies for heart repair.
Collapse
Affiliation(s)
| | - Asvin M Ganapathi
- Duke University Medical Center, Department of General Surgery, Durham, NC, USA
| | - Huda Asfour
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Ying Qian
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Brian W Allen
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Yanzhen Li
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Nenad Bursac
- Duke University, Department of Biomedical Engineering, Durham, NC, USA.
| |
Collapse
|
26
|
Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat Commun 2017; 8:1825. [PMID: 29184059 PMCID: PMC5705709 DOI: 10.1038/s41467-017-01946-x] [Citation(s) in RCA: 297] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 10/27/2017] [Indexed: 12/25/2022] Open
Abstract
Despite increased use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for drug development and disease modeling studies, methods to generate large, functional heart tissues for human therapy are lacking. Here we present a “Cardiopatch” platform for 3D culture and maturation of hiPSC-CMs that after 5 weeks of differentiation show robust electromechanical coupling, consistent H-zones, I-bands, and evidence for T-tubules and M-bands. Cardiopatch maturation markers and functional output increase during culture, approaching values of adult myocardium. Cardiopatches can be scaled up to clinically relevant dimensions, while preserving spatially uniform properties with high conduction velocities and contractile stresses. Within window chambers in nude mice, cardiopatches undergo vascularization by host vessels and continue to fire Ca2+ transients. When implanted onto rat hearts, cardiopatches robustly engraft, maintain pre-implantation electrical function, and do not increase the incidence of arrhythmias. These studies provide enabling technology for future use of hiPSC-CM tissues in human heart repair. Cardiomyocytes derived from human induced pluripotent stem cells could be used to generate cardiac tissues for regenerative purposes. Here the authors describe a method to obtain large bioengineered heart tissues showing advanced maturation, functional features and engraftment capacity.
Collapse
|
27
|
Yanamandala M, Zhu W, Garry DJ, Kamp TJ, Hare JM, Jun HW, Yoon YS, Bursac N, Prabhu SD, Dorn GW, Bolli R, Kitsis RN, Zhang J. Overcoming the Roadblocks to Cardiac Cell Therapy Using Tissue Engineering. J Am Coll Cardiol 2017; 70:766-775. [PMID: 28774384 PMCID: PMC5553556 DOI: 10.1016/j.jacc.2017.06.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 12/20/2022]
Abstract
Transplantations of various stem cells or their progeny have repeatedly improved cardiac performance in animal models of myocardial injury; however, the benefits observed in clinical trials have been generally less consistent. Some of the recognized challenges are poor engraftment of implanted cells and, in the case of human cardiomyocytes, functional immaturity and lack of electrical integration, leading to limited contribution to the heart's contractile activity and increased arrhythmogenic risks. Advances in tissue and genetic engineering techniques are expected to improve the survival and integration of transplanted cells, and to support structural, functional, and bioenergetic recovery of the recipient hearts. Specifically, application of a prefabricated cardiac tissue patch to prevent dilation and to improve pumping efficiency of the infarcted heart offers a promising strategy for making stem cell therapy a clinical reality.
Collapse
Affiliation(s)
- Mounica Yanamandala
- Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Wuqiang Zhu
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joshua M Hare
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Young-Sup Yoon
- Department of Medicine, Emory University, and Severance Biomedical Science Institute, Yonsei University College of Medicine, Atlanta, Georgia
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Sumanth D Prabhu
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gerald W Dorn
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky
| | - Richard N Kitsis
- Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Jianyi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
28
|
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
|
29
|
Morrissette-McAlmon J, Blazeski A, Somers S, Kostecki G, Tung L, Grayson WL. Adipose-derived perivascular mesenchymal stromal/stem cells promote functional vascular tissue engineering for cardiac regenerative purposes. J Tissue Eng Regen Med 2017; 12:e962-e972. [PMID: 28103423 DOI: 10.1002/term.2418] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 12/18/2016] [Accepted: 01/16/2017] [Indexed: 11/07/2022]
Abstract
Cardiac tissue engineering approaches have the potential to regenerate functional myocardium with intrinsic vascular networks. This study compared the relative effects of human adipose-derived stem/stromal cells (hASCs) and human dermal fibroblasts (hDFs) in cocultures with neonatal rat ventricular cardiomyocytes (NRVCMs) and human umbilical vein endothelial cells (HUVECs). At the same ratios of NRVCM:hASC and NRVCM:hDF, the hASC cocultures displayed shorter action potentials and maintained capture at faster pacing rates. Similarly, in coculture with HUVECs, hASC:HUVEC exhibited superior ability to support vascular capillary network formation relative to hDF:HUVEC. Based on these studies, a range of suitable cell ratios were determined to develop a triculture system. Six seeding ratios of NRVCM:hASC:HUVEC were tested and it was found that a ratio of 500:50:25 cells (i.e. 250,000:25,000:12,500 cells/cm2 ) resulted in the formation of robust vascular networks while retaining action potential durations and propagation similar to pure NRVCM cultures. Tricultures in this ratio exhibited an average conduction velocity of 20 ± 2 cm/s, action potential durations at 80% repolarization (APD80 ) and APD30 of 122 ± 5 ms and 59 ± 4 ms, respectively, and maximum capture rate of 7.4 ± 0.6 Hz. The NRVCM control groups had APD80 and APD30 of 120 ± 9 ms and 51 ± 5 ms, with a maximum capture rate of 7.3 ± 0.2 Hz. In summary, the combination of hASCs in the appropriate ratios with NRVCMs and HUVECs can facilitate the formation of densely vascularized cardiac tissues that appear not to impact the electrophysiological function of cardiomyocytes negatively. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Justin Morrissette-McAlmon
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adriana Blazeski
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Somers
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Geran Kostecki
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Warren L Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD, USA.,Institute for Nanobiotechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD, USA
| |
Collapse
|
30
|
Vilquin JT, Etienne J. [Cell therapies for cardiopathies: the shift of paradigms]. Med Sci (Paris) 2016; 32 Hors série n°2:30-39. [PMID: 27869075 DOI: 10.1051/medsci/201632s209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Heart failure is a major concern for public health systems, and several approaches of cellular therapy are being investigated with the goal of improving the function of these failing hearts. Many cell types have been used (skeletal myoblasts, hematopoietic, endothelial or mesenchymal progenitors, cardiac cells…), most often in the indication of post-ischemic heart failure rather than in the indication of genetic dilated cardiomyopathy. It is easier, indeed, to target a restricted area than the whole myocardium. Several clinical trials have reported slight but encouraging functional benefits, but their interpretations were frequently limited by the small sizes of cohorts, and by the biological variabilities inherent to the patients status and to the biology of the cells. These trials also shed light on unexpected mechanisms of action of the cells, which are changing the concepts and methodologies of the studies. The functional benefits observed would be due, indeed, to the secretion of trophic factors by the cells, instead of their true structural and mechanical integration within the myocardial tissue. Accordingly, the new generations of clinical trials aim at improving the size and homogeneity of the patient cohorts to increase the statistical power. On the other hand, several studies are associating or conditionning cells with biomaterials or cocktails of cytokines to improve their survival and their biological efficacy. In parallel, bio-engineering investigates several ways to support cells in vitro and in vivo, to sustain the architectural structure of the failing myocardium, to produce ex vivo some true substitutive cardiac tissue, or to purely replace the cells by their active secreted products. Several therapeutic devices should emerge from these researches, and the choice of their respective use will be ultimately guided by the medical indication.
Collapse
Affiliation(s)
- Jean-Thomas Vilquin
- Centre de Recherche en Myologie, Sorbonne Universités, UPMC-Inserm UMRS 974, CNRS FRE 3617, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jessy Etienne
- Centre de Recherche en Myologie, Sorbonne Universités, UPMC-Inserm UMRS 974, CNRS FRE 3617, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| |
Collapse
|
31
|
Cogels of Hyaluronic Acid and Acellular Matrix for Cultivation of Adipose-Derived Stem Cells: Potential Application for Vocal Fold Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6584054. [PMID: 27981051 PMCID: PMC5131240 DOI: 10.1155/2016/6584054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/10/2016] [Accepted: 09/29/2016] [Indexed: 11/28/2022]
Abstract
Stem cells based tissue engineering has been one of the potential promising therapies in the research on the repair of tissue diseases including the vocal fold. Decellularized extracellular matrix (DCM) as a promising scaffold has be used widely in tissue engineering; however, it remained to be an important issue in vocal fold regeneration. Here, we applied the hydrogels (hyaluronic acid [HA], HA-collagen [HA-Col], and HA-DCM) to determine the effects of hydrogel on the growth and differentiation of human adipose-derived stem cells (hADSCs) into superficial lamina propria fibroblasts. hADSCs were isolated and characterized by fluorescence-activated cell sorting. The results indicated that HA-DCM hydrogel enhanced cell proliferation and prolonged cell morphology significantly compared to HA and HA-Col hydrogel. Importantly, the differentiation of hADSCs into fibroblasts was also promoted by cogels of HA-Col and HA-DCM significantly. The differentiation of hADSCs towards superficial lamina propria fibroblasts was accelerated by the secretion of HGF, IL-8, and VEGF, the decorin and elastin expression, and the synthesis of chondroitin sulfate significantly. Therefore, the cogel of HA-DCM hydrogel was shown to be outstanding in apparent stimulation of hADSCs proliferation and differentiation to vocal fold fibroblasts through secretion of important growth factors and synthesis of extracellular matrix.
Collapse
|
32
|
Shadrin IY, Khodabukus A, Bursac N. Striated muscle function, regeneration, and repair. Cell Mol Life Sci 2016; 73:4175-4202. [PMID: 27271751 PMCID: PMC5056123 DOI: 10.1007/s00018-016-2285-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
As the only striated muscle tissues in the body, skeletal and cardiac muscle share numerous structural and functional characteristics, while exhibiting vastly different size and regenerative potential. Healthy skeletal muscle harbors a robust regenerative response that becomes inadequate after large muscle loss or in degenerative pathologies and aging. In contrast, the mammalian heart loses its regenerative capacity shortly after birth, leaving it susceptible to permanent damage by acute injury or chronic disease. In this review, we compare and contrast the physiology and regenerative potential of native skeletal and cardiac muscles, mechanisms underlying striated muscle dysfunction, and bioengineering strategies to treat muscle disorders. We focus on different sources for cellular therapy, biomaterials to augment the endogenous regenerative response, and progress in engineering and application of mature striated muscle tissues in vitro and in vivo. Finally, we discuss the challenges and perspectives in translating muscle bioengineering strategies to clinical practice.
Collapse
Affiliation(s)
- I Y Shadrin
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - A Khodabukus
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - N Bursac
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA.
| |
Collapse
|
33
|
Ruan JL, Tulloch NL, Razumova MV, Saiget M, Muskheli V, Pabon L, Reinecke H, Regnier M, Murry CE. Mechanical Stress Conditioning and Electrical Stimulation Promote Contractility and Force Maturation of Induced Pluripotent Stem Cell-Derived Human Cardiac Tissue. Circulation 2016; 134:1557-1567. [PMID: 27737958 DOI: 10.1161/circulationaha.114.014998] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tissue engineering enables the generation of functional human cardiac tissue with cells derived in vitro in combination with biocompatible materials. Human-induced pluripotent stem cell-derived cardiomyocytes provide a cell source for cardiac tissue engineering; however, their immaturity limits their potential applications. Here we sought to study the effect of mechanical conditioning and electric pacing on the maturation of human-induced pluripotent stem cell-derived cardiac tissues. METHODS Cardiomyocytes derived from human-induced pluripotent stem cells were used to generate collagen-based bioengineered human cardiac tissue. Engineered tissue constructs were subjected to different mechanical stress and electric pacing conditions. RESULTS The engineered human myocardium exhibits Frank-Starling-type force-length relationships. After 2 weeks of static stress conditioning, the engineered myocardium demonstrated increases in contractility (0.63±0.10 mN/mm2 vs 0.055±0.009 mN/mm2 for no stress), tensile stiffness, construct alignment, and cell size. Stress conditioning also increased SERCA2 (Sarco/Endoplasmic Reticulum Calcium ATPase 2) expression, which correlated with a less negative force-frequency relationship. When electric pacing was combined with static stress conditioning, the tissues showed an additional increase in force production (1.34±0.19 mN/mm2), with no change in construct alignment or cell size, suggesting maturation of excitation-contraction coupling. Supporting this notion, we found expression of RYR2 (Ryanodine Receptor 2) and SERCA2 further increased by combined static stress and electric stimulation. CONCLUSIONS These studies demonstrate that electric pacing and mechanical stimulation promote maturation of the structural, mechanical, and force generation properties of human-induced pluripotent stem cell-derived cardiac tissues.
Collapse
Affiliation(s)
- Jia-Ling Ruan
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Nathaniel L Tulloch
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Maria V Razumova
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Mark Saiget
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Veronica Muskheli
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Lil Pabon
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Hans Reinecke
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle
| | - Michael Regnier
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle.
| | - Charles E Murry
- From Department of Bioengineering (J.-L.R, M.V.R., M.R., C.E.M.), Program in Molecular and Cellular Biology (N.L.T.), Department of Pathology (N.L.T., M.R., V.M., L.P., H.R., C.E.M.), and Department of Medicine/Cardiology (C.E.M.), Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle.
| |
Collapse
|
34
|
Jackman CP, Carlson AL, Bursac N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials 2016; 111:66-79. [PMID: 27723557 DOI: 10.1016/j.biomaterials.2016.09.024] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 01/02/2023]
Abstract
Engineered cardiac tissues hold promise for cell therapy and drug development, but exhibit inadequate function and maturity. In this study, we sought to significantly improve the function and maturation of rat and human engineered cardiac tissues. We developed dynamic, free-floating culture conditions for engineering "cardiobundles", 3-dimensional cylindrical tissues made from neonatal rat cardiomyocytes or human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) embedded in fibrin-based hydrogel. Compared to static culture, 2-week dynamic culture of neonatal rat cardiobundles significantly increased expression of sarcomeric proteins, cardiomyocyte size (∼2.1-fold), contractile force (∼3.5-fold), and conduction velocity of action potentials (∼1.4-fold). The average contractile force per cross-sectional area (59.7 mN/mm2) and conduction velocity (52.5 cm/s) matched or approached those of adult rat myocardium, respectively. The inferior function of statically cultured cardiobundles was rescued by transfer to dynamic conditions, which was accompanied by an increase in mTORC1 activity and decline in AMPK phosphorylation and was blocked by rapamycin. Furthermore, dynamic culture effects did not stimulate ERK1/2 pathway and were insensitive to blockers of mechanosensitive channels, suggesting increased nutrient availability rather than mechanical stimulation as the upstream activator of mTORC1. Direct comparison with phenylephrine treatment confirmed that dynamic culture promoted physiological cardiomyocyte growth rather than pathological hypertrophy. Optimized dynamic culture conditions also augmented function of human cardiobundles made reproducibly from cardiomyocytes derived from multiple hPSC lines, resulting in significantly increased contraction force (∼2.5-fold) and conduction velocity (∼1.4-fold). The average specific force of 23.2 mN/mm2 and conduction velocity of 25.8 cm/s approached the functional metrics of adult human myocardium. In conclusion, we have developed a versatile methodology for engineering cardiac tissues with a near-adult functional output without the need for exogenous electrical or mechanical stimulation, and have identified mTOR signaling as an important mechanism for advancing tissue maturation and function in vitro.
Collapse
Affiliation(s)
| | - Aaron L Carlson
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.
| |
Collapse
|
35
|
Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the Damaged Heart: Mesenchymal Stem Cells, Cell-Based Therapy, and Engineered Heart Tissue. Physiol Rev 2016; 96:1127-68. [PMID: 27335447 PMCID: PMC6345247 DOI: 10.1152/physrev.00019.2015] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are broadly distributed cells that retain postnatal capacity for self-renewal and multilineage differentiation. MSCs evade immune detection, secrete an array of anti-inflammatory and anti-fibrotic mediators, and very importantly activate resident precursors. These properties form the basis for the strategy of clinical application of cell-based therapeutics for inflammatory and fibrotic conditions. In cardiovascular medicine, administration of autologous or allogeneic MSCs in patients with ischemic and nonischemic cardiomyopathy holds significant promise. Numerous preclinical studies of ischemic and nonischemic cardiomyopathy employing MSC-based therapy have demonstrated that the properties of reducing fibrosis, stimulating angiogenesis, and cardiomyogenesis have led to improvements in the structure and function of remodeled ventricles. Further attempts have been made to augment MSCs' effects through genetic modification and cell preconditioning. Progression of MSC therapy to early clinical trials has supported their role in improving cardiac structure and function, functional capacity, and patient quality of life. Emerging data have supported larger clinical trials that have been either completed or are currently underway. Mechanistically, MSC therapy is thought to benefit the heart by stimulating innate anti-fibrotic and regenerative responses. The mechanisms of action involve paracrine signaling, cell-cell interactions, and fusion with resident cells. Trans-differentiation of MSCs to bona fide cardiomyocytes and coronary vessels is also thought to occur, although at a nonphysiological level. Recently, MSC-based tissue engineering for cardiovascular disease has been examined with quite encouraging results. This review discusses MSCs from their basic biological characteristics to their role as a promising therapeutic strategy for clinical cardiovascular disease.
Collapse
Affiliation(s)
- Samuel Golpanian
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Ariel Wolf
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
36
|
Abstract
ABSTRACT
In February 2016, The Company of Biologists hosted an intimate gathering of leading international researchers at the forefront of experimental cardiovascular regeneration, with its emphasis on ‘Transdifferentiation and Tissue Plasticity in Cardiovascular Rejuvenation’. As I review here, participants at the workshop revealed how understanding cardiac growth and lineage decisions at their most fundamental level has transformed the strategies in hand that presently energize the prospects for human heart repair.
Collapse
Affiliation(s)
- Michael D. Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W14 8DZ, UK
| |
Collapse
|
37
|
Stem cell therapy for heart failure: Ensuring regenerative proficiency. Trends Cardiovasc Med 2016; 26:395-404. [PMID: 27020904 DOI: 10.1016/j.tcm.2016.01.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/08/2016] [Accepted: 01/20/2016] [Indexed: 02/07/2023]
Abstract
Patient-derived stem cells enable promising regenerative strategies, but display heterogenous cardiac reparative proficiency, leading to unpredictable therapeutic outcomes impeding practice adoption. Means to establish and certify the regenerative potency of emerging biotherapies are thus warranted. In this era of clinomics, deconvolution of variant cytoreparative performance in clinical trials offers an unprecedented opportunity to map pathways that segregate regenerative from non-regenerative states informing the evolution of cardio-regenerative quality systems. A maiden example of this approach is cardiopoiesis-mediated lineage specification developed to ensure regenerative performance. Successfully tested in pre-clinical and early clinical studies, the safety and efficacy of the cardiopoietic stem cell phenotype is undergoing validation in pivotal trials for chronic ischemic cardiomyopathy offering the prospect of a next-generation regenerative solution for heart failure.
Collapse
|
38
|
Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv Drug Deliv Rev 2016; 96:110-34. [PMID: 25956564 DOI: 10.1016/j.addr.2015.04.019] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/24/2015] [Accepted: 04/25/2015] [Indexed: 12/19/2022]
Abstract
Engineering functional human cardiac tissue that mimics the native adult morphological and functional phenotype has been a long held objective. In the last 5 years, the field of cardiac tissue engineering has transitioned from cardiac tissues derived from various animal species to the production of the first generation of human engineered cardiac tissues (hECTs), due to recent advances in human stem cell biology. Despite this progress, the hECTs generated to date remain immature relative to the native adult myocardium. In this review, we focus on the maturation challenge in the context of hECTs, the present state of the art, and future perspectives in terms of regenerative medicine, drug discovery, preclinical safety testing and pathophysiological studies.
Collapse
|
39
|
Pilarczyk G, Raulf A, Gunkel M, Fleischmann BK, Lemor R, Hausmann M. Tissue-Mimicking Geometrical Constraints Stimulate Tissue-Like Constitution and Activity of Mouse Neonatal and Human-Induced Pluripotent Stem Cell-Derived Cardiac Myocytes. J Funct Biomater 2016; 7:E1. [PMID: 26751484 PMCID: PMC4810060 DOI: 10.3390/jfb7010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/15/2022] Open
Abstract
The present work addresses the question of to what extent a geometrical support acts as a physiological determining template in the setup of artificial cardiac tissue. Surface patterns with alternating concave to convex transitions of cell size dimensions were used to organize and orientate human-induced pluripotent stem cell (hIPSC)-derived cardiac myocytes and mouse neonatal cardiac myocytes. The shape of the cells, as well as the organization of the contractile apparatus recapitulates the anisotropic line pattern geometry being derived from tissue geometry motives. The intracellular organization of the contractile apparatus and the cell coupling via gap junctions of cell assemblies growing in a random or organized pattern were examined. Cell spatial and temporal coordinated excitation and contraction has been compared on plain and patterned substrates. While the α-actinin cytoskeletal organization is comparable to terminally-developed native ventricular tissue, connexin-43 expression does not recapitulate gap junction distribution of heart muscle tissue. However, coordinated contractions could be observed. The results of tissue-like cell ensemble organization open new insights into geometry-dependent cell organization, the cultivation of artificial heart tissue from stem cells and the anisotropy-dependent activity of therapeutic compounds.
Collapse
Affiliation(s)
- Götz Pilarczyk
- Kirchhoff Institute für Physik, Im Neuenheimer Feld INF 270, Heidelberg D-69120, Germany.
| | - Alexandra Raulf
- Institut für Physiologie der Unversität Bonn, Life & Brain Center, Sigmund Freud Strasse 25, Bonn D-53127, Germany.
| | - Manuel Gunkel
- ViroQuant Cell Networks RNAi Screening Facility, BioQuant Center, Im Neuenheimer Feld INF 267, Heidelberg D-69120, Germany.
| | - Bernd K Fleischmann
- Institut für Physiologie der Unversität Bonn, Life & Brain Center, Sigmund Freud Strasse 25, Bonn D-53127, Germany.
| | - Robert Lemor
- Luxembourg Institute for Science and Technology, 5 avenue des Hauts-Fourneaux, Esch-Belval L-4362, Luxembourg.
| | - Michael Hausmann
- Kirchhoff Institute für Physik, Im Neuenheimer Feld INF 270, Heidelberg D-69120, Germany.
| |
Collapse
|