1
|
Zhu A, Reid E, Jain T, Mir A, Siddiqi U, Dunne O, Hibino N. Advancing Tissue Engineering Through a Portable Perfusion and Incubation System. Bioengineering (Basel) 2025; 12:554. [PMID: 40428173 PMCID: PMC12109021 DOI: 10.3390/bioengineering12050554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Perfusion offers unique benefits to tissue-engineered systems, enhancing oxygen and nutrient transport, which improves tissue formation and growth. In this study, we present a novel and integrated portable perfusion system. Weighing < 10 lbs, the system can maintain continuous flow in a standard incubation environment (37 °C, 5% CO2), effectively functioning as a portable perfusion and tissue culturing system. To characterize the perfusion system's flow parameters, we measured the volumetric flow rate across a range of pressures and found that the system could achieve flow velocities between 1.69 to 4.6 μm/s, which is similar to in vivo interstitial flow. Computational fluid dynamics revealed fully developed laminar flow within the sample-containing region of the perfusion system, helping ensure even fluid and nutrient distribution. To study the system's compatibility with live tissues, bioengineered tissue patches were created and perfused. After 24 h of perfusion, no significant difference in cell viability was observed between the perfused samples and static controls, indicating no adverse effects on cell health. Perfusion also facilitated enhanced spatial organization within tissue patches, reducing the inter-spheroids distance. Furthermore, perfusion strengthened the tissue matrix and reduced the degradation rate of the hydrogel scaffold. Complemented by its ability to provide mobile perfusion and incubation, this novel integrated portable perfusion system holds promise for promoting tissue maturation and advancing tissue bioengineering studies.
Collapse
Affiliation(s)
- Angie Zhu
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
| | - Emmett Reid
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
| | - Tilak Jain
- 37degrees, 111 North Wabash Ave. Ste. 100, Chicago, IL 60602, USA;
| | - Amatullah Mir
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
| | - Usmaan Siddiqi
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
| | - Olivia Dunne
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
| | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (E.R.); (A.M.); (U.S.); (O.D.); (N.H.)
- Pediatric Cardiac Surgery, Advocate Children’s Hospital, 4440 W 95th St., Oak Lawn, IL 60453, USA
| |
Collapse
|
2
|
Ke M, Xu W, Hao Y, Zheng F, Yang G, Fan Y, Wang F, Nie Z, Zhu C. Construction of millimeter-scale vascularized engineered myocardial tissue using a mixed gel. Regen Biomater 2023; 11:rbad117. [PMID: 38223293 PMCID: PMC10786677 DOI: 10.1093/rb/rbad117] [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: 10/17/2023] [Revised: 12/10/2023] [Accepted: 12/17/2023] [Indexed: 01/16/2024] Open
Abstract
Engineering myocardium has shown great clinal potential for repairing permanent myocardial injury. However, the lack of perfusing blood vessels and difficulties in preparing a thick-engineered myocardium result in its limited clinical use. We prepared a mixed gel containing fibrin (5 mg/ml) and collagen I (0.2 mg/ml) and verified that human umbilical vein endothelial cells (HUVECs) and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could form microvascular lumens and myocardial cell clusters by harnessing the low-hardness and hyperelastic characteristics of fibrin. hiPSC-CMs and HUVECs in the mixed gel formed self-organized cell clusters, which were then cultured in different media using a three-phase approach. The successfully constructed vascularized engineered myocardial tissue had a spherical structure and final diameter of 1-2 mm. The tissue exhibited autonomous beats that occurred at a frequency similar to a normal human heart rate. The internal microvascular lumen could be maintained for 6 weeks and showed good results during preliminary surface re-vascularization in vitro and vascular remodeling in vivo. In summary, we propose a simple method for constructing vascularized engineered myocardial tissue, through phased cultivation that does not rely on high-end manufacturing equipment and cutting-edge preparation techniques. The constructed tissue has potential value for clinical use after preliminary evaluation.
Collapse
Affiliation(s)
- Ming Ke
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Wenhui Xu
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Yansha Hao
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Feiyang Zheng
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Guanyuan Yang
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Yonghong Fan
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Fangfang Wang
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Zhiqiang Nie
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
| | - Chuhong Zhu
- Department of Anatomy, Third Military Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing 400038, China
- Department of Plastic and Aesthetic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing 400038, China
| |
Collapse
|
3
|
Tenchurin TK, Rodina AV, Saprykin VP, Gorshkova LV, Mikhutkin AA, Kamyshinsky RA, Yakovlev DS, Vasiliev AL, Chvalun SN, Grigoriev TE. The Performance of Nonwoven PLLA Scaffolds of Different Thickness for Stem Cells Seeding and Implantation. Polymers (Basel) 2022; 14:polym14204352. [PMID: 36297930 PMCID: PMC9610477 DOI: 10.3390/polym14204352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/24/2022] [Accepted: 10/13/2022] [Indexed: 11/22/2022] Open
Abstract
The 3D reconstruction of 100 μm- and 600 μm-thick fibrous poly-L/L-lactide scaffolds was performed by confocal laser scanning microscopy and supported by scanning electron microscopy and showed that the density of the fibers on the side adjacent to the electrode is higher, which can affect cell diffusion, while the pore size is generally the same. Bone marrow mesenchymal stem cells cultured in a 600 μm-thick scaffold formed colonies and produced conditions for cell differentiation. An in vitro study of stem cells after 7 days revealed that cell proliferation and hepatocyte growth factor release in the 600 μm-thick scaffold were higher than in the 100 μm-thick scaffold. An in vivo study of scaffolds with and without stem cells implanted subcutaneously onto the backs of recipient mice was carried out to test their biodegradation and biocompatibility over a 0-3-week period. The cells seeded onto the 600 μm-thick scaffold promoted significant neovascularization in vivo. After 3 weeks, a significant number of donor cells persisted only on the inside of the 600 μm-thick scaffold. Thus, the use of bulkier matrices allows to prolong the effect of secretion of growth factors by stem cells during implantation. These 600 μm-thick scaffolds could potentially be utilized to repair and regenerate injuries with stem cell co-culture for vascularization of implant.
Collapse
Affiliation(s)
| | - Alla V. Rodina
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
| | - Vladimir P. Saprykin
- Natural Sciences Department, Moscow Region State University, 105005 Moscow, Russia
| | - Lada V. Gorshkova
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
| | | | - Roman A. Kamyshinsky
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
- Shubnikov Institute of Crystallography of FSRC “Crystallography and Photonics” RAS, 119333 Moscow, Russia
| | - Dmitry S. Yakovlev
- Russian Quantum Center, Skolkovo, 121205 Moscow, Russia
- Institute of Nano-, Bio-, Information, Cognitive and Socio-Humanistic Sciences and Technologies, Moscow Institute of Physics and Technology, State University, 141707 Dolgoprudny, Russia
| | - Alexander L. Vasiliev
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
- Shubnikov Institute of Crystallography of FSRC “Crystallography and Photonics” RAS, 119333 Moscow, Russia
- Institute of Nano-, Bio-, Information, Cognitive and Socio-Humanistic Sciences and Technologies, Moscow Institute of Physics and Technology, State University, 141707 Dolgoprudny, Russia
- Correspondence: ; Tel.: +7-910-4130115
| | - Sergey N. Chvalun
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
| | - Timofey E. Grigoriev
- National Research Centre “Kurchatov Institute”, 123098 Moscow, Russia
- Institute of Nano-, Bio-, Information, Cognitive and Socio-Humanistic Sciences and Technologies, Moscow Institute of Physics and Technology, State University, 141707 Dolgoprudny, Russia
| |
Collapse
|
4
|
Chu X, Wang M, Qiu X, Huang Y, Li T, Otieno E, Li N, Luo L, Xiao X. Strategies for constructing pluripotent stem cell- and progenitor cell-derived three-dimensional cardiac micro-tissues. J Biomed Mater Res A 2021; 110:488-503. [PMID: 34397148 DOI: 10.1002/jbm.a.37298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cardiac micro-tissue is a promising model for simulating the structural and functional features of heart in vitro. This scientific achievement provides a platform for exploration about the mechanisms on the development, damage, and regeneration of tissue, hence, paving a way toward development of novel therapies for heart diseases. However, 3D micro-tissue technology is still in its infant stages faced with many challenges such as incompleteness of the tissue microarchitecture, loss of the resident immune cells, poor reproducibility, and deficiencies in continuously feeding the nutrients and removing wastes during micro-tissue culturing. There is an urgent need to optimize the construction of 3D cardiac micro-tissue and improve functions of the involved cells. Therefore, scaffolds and cell resources for building 3D cardiac micro-tissues, strategies for inducing the maturation and functionalization of pluripotent stem cell- or cardiac progenitor cell-derived cardiomyocytes, and the major challenges were reviewed in this writing to enable future fabrication of 3D cardiac micro-tissues or organoids for drug screening, disease modeling, regeneration treatment, and so on.
Collapse
Affiliation(s)
- Xinyue Chu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Laboratory Animals Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Husbandry Engineering, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Tong Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Na Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
5
|
Tavakol DN, Fleischer S, Vunjak-Novakovic G. Harnessing organs-on-a-chip to model tissue regeneration. Cell Stem Cell 2021; 28:993-1015. [PMID: 34087161 DOI: 10.1016/j.stem.2021.05.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue engineering has markedly matured since its early beginnings in the 1980s. In addition to the original goal to regenerate damaged organs, the field has started to explore modeling of human physiology "in a dish." Induced pluripotent stem cell (iPSC) technologies now enable studies of organ regeneration and disease modeling in a patient-specific context. We discuss the potential of "organ-on-a-chip" systems to study regenerative therapies with focus on three distinct organ systems: cardiac, respiratory, and hematopoietic. We propose that the combinatorial studies of human tissues at these two scales would help realize the translational potential of tissue engineering.
Collapse
Affiliation(s)
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Medicine, Columbia University, New York, NY.
| |
Collapse
|
6
|
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
|
7
|
Zhao Y, Rafatian N, Wang EY, Wu Q, Lai BFL, Lu RX, Savoji H, Radisic M. Towards chamber specific heart-on-a-chip for drug testing applications. Adv Drug Deliv Rev 2020; 165-166:60-76. [PMID: 31917972 PMCID: PMC7338250 DOI: 10.1016/j.addr.2019.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Modeling of human organs has long been a task for scientists in order to lower the costs of therapeutic development and understand the pathological onset of human disease. For decades, despite marked differences in genetics and etiology, animal models remained the norm for drug discovery and disease modeling. Innovative biofabrication techniques have facilitated the development of organ-on-a-chip technology that has great potential to complement conventional animal models. However, human organ as a whole, more specifically the human heart, is difficult to regenerate in vitro, in terms of its chamber specific orientation and its electrical functional complexity. Recent progress with the development of induced pluripotent stem cell differentiation protocols, made recapitulating the complexity of the human heart possible through the generation of cells representative of atrial & ventricular tissue, the sinoatrial node, atrioventricular node and Purkinje fibers. Current heart-on-a-chip approaches incorporate biological, electrical, mechanical, and topographical cues to facilitate tissue maturation, therefore improving the predictive power for the chamber-specific therapeutic effects targeting adult human. In this review, we will give a summary of current advances in heart-on-a-chip technology and provide a comprehensive outlook on the challenges involved in the development of human physiologically relevant heart-on-a-chip.
Collapse
Affiliation(s)
- Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University of Health Network, Toronto, Ontario M5G 2N2, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Benjamin F L Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
8
|
Abstract
Some of the most significant leaps in the history of modern civilization-the development of article in China, the steam engine, which led to the European industrial revolution, and the era of computers-have occurred when science converged with engineering. Recently, the convergence of human pluripotent stem cell technology with biomaterials and bioengineering have launched a new medical innovation: functional human engineered tissue, which promises to revolutionize the treatment of failing organs including most critically, the heart. This compendium covers recent, state-of-the-art developments in the fields of cardiovascular tissue engineering, as well as the needs and challenges associated with the clinical use of these technologies. We have not attempted to provide an exhaustive review in stem cell biology and cardiac cell therapy; many other important and influential reports are certainly merit but already been discussed in several recent reviews. Our scope is limited to the engineered tissues that have been fabricated to repair or replace components of the heart (eg, valves, vessels, contractile tissue) that have been functionally compromised by diseases or developmental abnormalities. In particular, we have focused on using an engineered myocardial tissue to mitigate deficiencies in contractile function.
Collapse
Affiliation(s)
- Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Wuqiang Zhu
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada (M.R.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering and Department of Medicine, Columbia University, New York, NY (G.V.-N.)
| |
Collapse
|
9
|
Hogan M, Chen YT, Kolhatkar AG, Candelari CJ, Madala S, Lee TR, Birla R. Conditioning of Cardiovascular Tissue Using a Noncontact Magnetic Stretch Bioreactor with Embedded Magnetic Nanoparticles. ACS Biomater Sci Eng 2016; 2:1619-1629. [DOI: 10.1021/acsbiomaterials.6b00375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Matthew Hogan
- Department of Biomedical
Engineering, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Yi-Ting Chen
- Department
of Chemistry, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 5004, Houston, Texas 77204-5060, United States
| | - Arati G. Kolhatkar
- Department
of Chemistry, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 5004, Houston, Texas 77204-5060, United States
| | - Christopher J. Candelari
- Department of Biomedical
Engineering, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Sridhar Madala
- Indus Instruments, 721 Tristar Drive, Webster, Texas 77598, United States
| | - T. Randall Lee
- Department
of Chemistry, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 5004, Houston, Texas 77204-5060, United States
| | - Ravi Birla
- Department of Biomedical
Engineering, Science and Engineering Research Center (SERC-Building
545), University of Houston, 3605 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| |
Collapse
|
10
|
Kharaziha M, Memic A, Akbari M, Brafman DA, Nikkhah M. Nano-Enabled Approaches for Stem Cell-Based Cardiac Tissue Engineering. Adv Healthc Mater 2016; 5:1533-53. [PMID: 27199266 DOI: 10.1002/adhm.201600088] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/01/2016] [Indexed: 12/20/2022]
Abstract
Cardiac diseases are the most prevalent causes of mortality in the world, putting a major economic burden on global healthcare system. Tissue engineering strategies aim at developing efficient therapeutic approaches to overcome the current challenges in prolonging patients survival upon cardiac diseases. The integration of advanced biomaterials and stem cells has offered enormous promises for regeneration of damaged myocardium. Natural or synthetic biomaterials have been extensively used to deliver cells or bioactive molecules to the site of injury in heart. Additionally, nano-enabled approaches (e.g., nanomaterials, nanofeatured surfaces) have been instrumental in developing suitable scaffolding biomaterials and regulating stem cells microenvironment to achieve functional therapeutic outcomes. This review article explores tissue engineering strategies, which have emphasized on the use of nano-enabled approaches in combination with stem cells for regeneration and repair of injured myocardium upon myocardial infarction (MI). Primarily a wide range of biomaterials, along with different types of stem cells, which have utilized in cardiac tissue engineering will be presented. Then integration of nanomaterials and surface nanotopographies with biomaterials and stem cells for myocardial regeneration will be presented. The advantages and challenges of these approaches will be reviewed and future perspective will be discussed.
Collapse
Affiliation(s)
- Mahshid Kharaziha
- Biomaterials Research Group; Department of Materials Engineering; Isfahan University of Technology; Isfahan 8415683111 Iran
| | - Adnan Memic
- Center of Nanotechnology; King Abdulaziz University; Jeddah 21589 Saudi Arabia
| | - Mohsen Akbari
- Department of Mechanical Engineering; University of Victoria; Victoria BC Canada
| | - David A. Brafman
- School of Biological and Health Systems Engineering (SBHSE) Harington; Bioengineering Program; Arizona State University; Tempe Arizona 85287 USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE) Harington; Bioengineering Program; Arizona State University; Tempe Arizona 85287 USA
| |
Collapse
|
11
|
Qiu L, Lim YM, Chen AK, Reuveny S, Oh SKW, Tan EK, Zeng L. Microcarrier-Expanded Neural Progenitor Cells Can Survive, Differentiate, and Innervate Host Neurons Better When Transplanted as Aggregates. Cell Transplant 2016; 25:1343-57. [DOI: 10.3727/096368915x690378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neuronal progenitor cells (NPCs) derived from human embryonic stem cells (hESCs) are an excellent cell source for transplantation therapy due to their availability and ethical acceptability. However, the traditional method of expansion and differentiation of hESCs into NPCs in monolayer cultures requires a long time, and the cell yield is low. A microcarrier (MC) platform can improve the expansion of hESCs and increase the yield of NPCs. In this study, for the first time, we transplanted microcarrier-expanded hESC-derived NPCs into the striatum of adult NOD-SCID IL2Rgc null mice, either as single cells or as cell aggregates. The recipient mice were perfused, and the in vivo survival, differentiation, and targeted innervation of the transplanted cells were assessed by immunostaining. We found that both the transplanted single NPCs and aggregate NPCs were able to survive 1 month posttransplantation, as revealed by human-specific neural cell adhesion molecule (NCAM) and human nuclear antigen staining. Compared to the single cells, the transplanted cell aggregates showed better survival over a 3-month period. In addition, both the transplanted single NPCs and the aggregate NPCs were able to differentiate into DCX-positive immature neurons and Tuj1-positive neurons in vivo by 1 month posttransplantation. However, only the transplantation of aggregate NPCs was shown to result in mature neurons at 3 months posttransplantation. Furthermore, we found that the cell aggregates were able to send long axons to innervate their targets. Our study provides preclinical evidence that the use of MCs to expand and differentiate hESC-derived NPCs and transplantation of these cells as aggregates produce longer survival in vivo.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Yu Ming Lim
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Allen K. Chen
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Steve K. W. Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| |
Collapse
|
12
|
Massai D, Isu G, Madeddu D, Cerino G, Falco A, Frati C, Gallo D, Deriu MA, Falvo D'Urso Labate G, Quaini F, Audenino A, Morbiducci U. A Versatile Bioreactor for Dynamic Suspension Cell Culture. Application to the Culture of Cancer Cell Spheroids. PLoS One 2016; 11:e0154610. [PMID: 27144306 PMCID: PMC4856383 DOI: 10.1371/journal.pone.0154610] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/15/2016] [Indexed: 11/27/2022] Open
Abstract
A versatile bioreactor suitable for dynamic suspension cell culture under tunable shear stress conditions has been developed and preliminarily tested culturing cancer cell spheroids. By adopting simple technological solutions and avoiding rotating components, the bioreactor exploits the laminar hydrodynamics establishing within the culture chamber enabling dynamic cell suspension in an environment favourable to mass transport, under a wide range of tunable shear stress conditions. The design phase of the device has been supported by multiphysics modelling and has provided a comprehensive analysis of the operating principles of the bioreactor. Moreover, an explanatory example is herein presented with multiphysics simulations used to set the proper bioreactor operating conditions for preliminary in vitro biological tests on a human lung carcinoma cell line. The biological results demonstrate that the ultralow shear dynamic suspension provided by the device is beneficial for culturing cancer cell spheroids. In comparison to the static suspension control, dynamic cell suspension preserves morphological features, promotes intercellular connection, increases spheroid size (2.4-fold increase) and number of cycling cells (1.58-fold increase), and reduces double strand DNA damage (1.5-fold reduction). It is envisioned that the versatility of this bioreactor could allow investigation and expansion of different cell types in the future.
Collapse
Affiliation(s)
- Diana Massai
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Giuseppe Isu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Denise Madeddu
- Department of Clinical and Experimental Medicine, Università degli Studi di Parma, Parma, Italy
| | - Giulia Cerino
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Angela Falco
- Department of Clinical and Experimental Medicine, Università degli Studi di Parma, Parma, Italy
| | - Caterina Frati
- Department of Clinical and Experimental Medicine, Università degli Studi di Parma, Parma, Italy
| | - Diego Gallo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Marco A Deriu
- Istituto Dalle Molle di studi sull'Intelligenza Artificiale, Scuola universitaria professionale della Svizzera italiana, Università della Svizzera italiana, Manno, Switzerland
| | | | - Federico Quaini
- Department of Clinical and Experimental Medicine, Università degli Studi di Parma, Parma, Italy
| | - Alberto Audenino
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
13
|
Salazar BH, Cashion AT, Dennis RG, Birla RK. Development of a Cyclic Strain Bioreactor for Mechanical Enhancement and Assessment of Bioengineered Myocardial Constructs. Cardiovasc Eng Technol 2015; 6:533-45. [PMID: 26577484 DOI: 10.1007/s13239-015-0236-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 07/14/2015] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to develop enabling bioreactor technologies using a novel voice coil actuator system for investigating the effects of periodic strain on cardiac patches fabricated with rat cardiomyocytes. The bioengineered muscle constructs used in this study were formed by culturing rat neonatal primary cardiac cells on a fibrin gel. The physical design of the bioreactor was initially conceived using Solidworks to test clearances and perform structural strain analysis. Once the software design phase was completed the bioreactor was assembled using a combination of commercially available, custom machined, and 3-D printed parts. We utilized the bioreactor to evaluate the effect of a 4-h stretch protocol on the contractile properties of the tissue after which immunohistological assessment of the tissue was also performed. An increase in contractile force was observed after the strain protocol of 10% stretch at 1 Hz, with no significant increase observed in the control group. Additionally, an increase in cardiac myofibril alignment, connexin 43 expression, and collagen type I distribution were noted. In this study we demonstrated the effectiveness of a new bioreactor design to improve contractility of engineered cardiac muscle tissue.
Collapse
Affiliation(s)
- Betsy H Salazar
- Department of Biomedical Engineering, Science and Engineering Research Center (SERC), Cullen College of Engineering, University of Houston, 3605 Cullen Blvd, Rm. 2021, Houston, TX, 77204, USA.
| | - Avery T Cashion
- Joint Department of Biomedical Engineering, University of North Carolina/North Carolina State University, Chapel Hill, NC, USA
| | - Robert G Dennis
- Joint Department of Biomedical Engineering, University of North Carolina/North Carolina State University, Chapel Hill, NC, USA
| | - Ravi K Birla
- Department of Biomedical Engineering, Science and Engineering Research Center (SERC), Cullen College of Engineering, University of Houston, 3605 Cullen Blvd, Rm. 2021, Houston, TX, 77204, USA.
| |
Collapse
|
14
|
32-Channel System to Measure the Electrophysiological Properties of Bioengineered Cardiac Muscle. IEEE Trans Biomed Eng 2015; 62:1614-22. [DOI: 10.1109/tbme.2015.2399437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
15
|
Efficacy of supermacroporous poly(ethylene glycol)–gelatin cryogel matrix for soft tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 47:298-312. [DOI: 10.1016/j.msec.2014.11.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/11/2014] [Accepted: 11/08/2014] [Indexed: 02/07/2023]
|
16
|
Nam KH, Smith AST, Lone S, Kwon S, Kim DH. Biomimetic 3D Tissue Models for Advanced High-Throughput Drug Screening. ACTA ACUST UNITED AC 2014; 20:201-15. [PMID: 25385716 DOI: 10.1177/2211068214557813] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Indexed: 12/13/2022]
Abstract
Most current drug screening assays used to identify new drug candidates are 2D cell-based systems, even though such in vitro assays do not adequately re-create the in vivo complexity of 3D tissues. Inadequate representation of the human tissue environment during a preclinical test can result in inaccurate predictions of compound effects on overall tissue functionality. Screening for compound efficacy by focusing on a single pathway or protein target, coupled with difficulties in maintaining long-term 2D monolayers, can serve to exacerbate these issues when using such simplistic model systems for physiological drug screening applications. Numerous studies have shown that cell responses to drugs in 3D culture are improved from those in 2D, with respect to modeling in vivo tissue functionality, which highlights the advantages of using 3D-based models for preclinical drug screens. In this review, we discuss the development of microengineered 3D tissue models that accurately mimic the physiological properties of native tissue samples and highlight the advantages of using such 3D microtissue models over conventional cell-based assays for future drug screening applications. We also discuss biomimetic 3D environments, based on engineered tissues as potential preclinical models for the development of more predictive drug screening assays for specific disease models.
Collapse
Affiliation(s)
- Ki-Hwan Nam
- Department of Bioengineering, University of Washington, Seattle, WA, USA Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea Center for Analytical Instrumentation Development, The Korea Basic Science Institute, Deajeon, Republic of Korea
| | - Alec S T Smith
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Saifullah Lone
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Regenerative implants for cardiovascular tissue engineering. Transl Res 2014; 163:321-41. [PMID: 24589506 DOI: 10.1016/j.trsl.2014.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/27/2014] [Accepted: 01/27/2014] [Indexed: 01/22/2023]
Abstract
A fundamental problem that affects the field of cardiovascular surgery is the paucity of autologous tissue available for surgical reconstructive procedures. Although the best results are obtained when an individual's own tissues are used for surgical repair, this is often not possible as a result of pathology of autologous tissues or lack of a compatible replacement source from the body. The use of prosthetics is a popular solution to overcome shortage of autologous tissue, but implantation of these devices comes with an array of additional problems and complications related to biocompatibility. Transplantation offers another option that is widely used but complicated by problems related to rejection and donor organ scarcity. The field of tissue engineering represents a promising new option for replacement surgical procedures. Throughout the years, intensive interdisciplinary, translational research into cardiovascular regenerative implants has been undertaken in an effort to improve surgical outcome and better quality of life for patients with cardiovascular defects. Vascular, valvular, and heart tissue repair are the focus of these efforts. Implants for these neotissues can be divided into 2 groups: biologic and synthetic. These materials are used to facilitate the delivery of cells or drugs to diseased, damaged, or absent tissue. Furthermore, they can function as a tissue-forming device used to enhance the body's own repair mechanisms. Various preclinical studies and clinical trials using these advances have shown that tissue-engineered materials are a viable option for surgical repair, but require refinement if they are going to reach their clinical potential. With the growth and accomplishments this field has already achieved, meeting those goals in the future should be attainable.
Collapse
|
18
|
Emmert MY, Hitchcock RW, Hoerstrup SP. Cell therapy, 3D culture systems and tissue engineering for cardiac regeneration. Adv Drug Deliv Rev 2014; 69-70:254-69. [PMID: 24378579 DOI: 10.1016/j.addr.2013.12.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/06/2013] [Accepted: 12/17/2013] [Indexed: 01/02/2023]
Abstract
Ischemic Heart Disease (IHD) still represents the "Number One Killer" worldwide accounting for the death of numerous patients. However the capacity for self-regeneration of the adult heart is very limited and the loss of cardiomyocytes in the infarcted heart leads to continuous adverse cardiac-remodeling which often leads to heart-failure (HF). The concept of regenerative medicine comprising cell-based therapies, bio-engineering technologies and hybrid solutions has been proposed as a promising next-generation approach to address IHD and HF. Numerous strategies are under investigation evaluating the potential of regenerative medicine on the failing myocardium including classical cell-therapy concepts, three-dimensional culture techniques and tissue-engineering approaches. While most of these regenerative strategies have shown great potential in experimental studies, the translation into a clinical setting has either been limited or too rapid leaving many key questions unanswered. This review summarizes the current state-of-the-art, important challenges and future research directions as to regenerative approaches addressing IHD and resulting HF.
Collapse
|
19
|
Abstract
Tissue engineering aims to create, repair and/or replace tissues and organs by using cells, scaffolds, biologically active molecules and physiologic signals. It is an interdisciplinary field that integrates aspects of engineering, chemistry, biology and medicine. One of the most challenging goals in the field of cardiovascular tissue engineering is the creation of a heart muscle patch. This review describes the principles, achievements and challenges of achieving this ambitious goal of creating contractile heart muscle. In addition, the new strategy of in situ and injectable tissue engineering for myocardial repair and regeneration is presented.
Collapse
Affiliation(s)
- Jonathan Leor
- Sheba-Medical Center, Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Hashomer 52621, Israel.
| | | | | |
Collapse
|
20
|
Serpooshan V, Ruiz-Lozano P. Ultra-rapid manufacturing of engineered epicardial substitute to regenerate cardiac tissue following acute ischemic injury. Methods Mol Biol 2014; 1210:239-248. [PMID: 25173173 DOI: 10.1007/978-1-4939-1435-7_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Considering the impaired regenerative capacity of adult mammalian heart tissue, cardiovascular tissue engineering aims to create functional substitutes that can restore the structure and function of the damaged cardiac tissue. The success of cardiac regenerative therapies has been limited mainly due to poor control on the structure and properties of the tissue substitute, lack of vascularization, and immunogenicity. In this study we introduce a new approach to rapidly engineer dense biomimetic scaffolds consisting of type I collagen, to protect the heart against severe ischemic injury. Scaffold biomechanical properties are adjusted to mimic embryonic epicardium which is shown to be optimal to support cardiomyocyte contractile work. Moreover, the designed patch can serve as a delivery device for targeted, controlled release of cells or therapeutic macromolecules into the lesion area.
Collapse
Affiliation(s)
- Vahid Serpooshan
- Department of Pediatrics, Stanford Cardiovascular Institute, 300 Pasteur Dr., Stanford, CA, 94305, USA
| | | |
Collapse
|
21
|
Buikema JW, Zwetsloot PPM, Doevendans PA, Sluijter JP, Domian IJ. Expanding mouse ventricular cardiomyocytes through GSK-3 inhibition. CURRENT PROTOCOLS IN CELL BIOLOGY 2013; 61:23.9.1-23.9.10. [PMID: 24505027 PMCID: PMC3934298 DOI: 10.1002/0471143030.cb2309s61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Controlled proliferation of cardiomyocytes remains a major limitation in cell biology and one of the main underlying hurdles for true modern regenerative medicine. Here, a technique is described for robust expansion of early fetal-derived mouse ventricular cardiomyocytes on a platform usable for high-throughput molecular screening, tissue engineering and, potentially, in vivo translational experiments. This method provides a small-molecule approach to control proliferation or differentiation of early beating cardiomyocytes through modulation of the Wnt/β-catenin signaling pathway. Moreover, isolation and expansion of fetal cardiomyocytes takes less than 3 weeks, yields a relatively pure (∼70%) functional myogenic population, and is highly reproducible.
Collapse
Affiliation(s)
- Jan Willem Buikema
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA,Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA,Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands,To whom correspondence should be addressed, ,
| | - Peter-Paul M. Zwetsloot
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA,Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Joost P.G. Sluijter
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ibrahim J. Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA,Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA,Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA,To whom correspondence should be addressed, ,
| |
Collapse
|
22
|
Massai D, Cerino G, Gallo D, Pennella F, Deriu M, Rodriguez A, Montevecchi F, Bignardi C, Audenino A, Morbiducci U. Bioreactors as Engineering Support to Treat Cardiac Muscle and Vascular Disease. JOURNAL OF HEALTHCARE ENGINEERING 2013; 4:329-70. [DOI: 10.1260/2040-2295.4.3.329] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Brenner C, David R, Franz WM. Cardiovascular Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
24
|
Lieu DK, Turnbull IC, Costa KD, Li RA. Engineered human pluripotent stem cell-derived cardiac cells and tissues for electrophysiological studies. ACTA ACUST UNITED AC 2012; 9:e209-e217. [PMID: 29422934 DOI: 10.1016/j.ddmod.2012.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Human cardiomyocytes (CMs) do not proliferate in culture and are difficult to obtain for practical reasons. As such, our understanding of the mechanisms that underlie the physiological and pathophysiological development of the human heart is mostly extrapolated from studies of the mouse and other animal models or heterologus expression of defective gene product(s) in non-human cells. Although these studies provided numerous important insights, much of the exact behavior in human cells remains unexplored given that significant species differences exist. With the derivation of human embryonic stem cells (hESC) and induced pluripotent stem cells (iPSCs) from patients with underlying heart disease, a source of human CMs for disease modeling, cardiotoxicity screening and drug discovery is now available. In this review, we focus our discussion on the use of hESC/ iPSC-derived cardiac cells and tissues for studying various heart rhythm disorders and the associated pro-arrhythmogenic properties in relation to advancements in electrophysiology and tissue engineering.
Collapse
Affiliation(s)
- Deborah K Lieu
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY, United States.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, United States
| | - Irene C Turnbull
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY, United States
| | - Kevin D Costa
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY, United States
| | - Ronald A Li
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY, United States.,Stem Cell & Regenerative Medicine Consortium, University of Hong Kong, Pokfulam, Hong Kong.,Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Department of Physiology, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
25
|
Bauer M, Kang L, Qiu Y, Wu J, Peng M, Chen HH, Camci-Unal G, Bayomy AF, Sosnovik DE, Khademhosseini A, Liao R. Adult cardiac progenitor cell aggregates exhibit survival benefit both in vitro and in vivo. PLoS One 2012; 7:e50491. [PMID: 23226295 PMCID: PMC3511575 DOI: 10.1371/journal.pone.0050491] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 10/22/2012] [Indexed: 01/08/2023] Open
Abstract
Background A major hurdle in the use of exogenous stems cells for therapeutic regeneration of injured myocardium remains the poor survival of implanted cells. To date, the delivery of stem cells into myocardium has largely focused on implantation of cell suspensions. Methodology and Principal Findings We hypothesize that delivering progenitor cells in an aggregate form would serve to mimic the endogenous state with proper cell-cell contact, and may aid the survival of implanted cells. Microwell methodologies allow for the culture of homogenous 3D cell aggregates, thereby allowing cell-cell contact. In this study, we find that the culture of cardiac progenitor cells in a 3D cell aggregate augments cell survival and protects against cellular toxins and stressors, including hydrogen peroxide and anoxia/reoxygenation induced cell death. Moreover, using a murine model of cardiac ischemia-reperfusion injury, we find that delivery of cardiac progenitor cells in the form of 3D aggregates improved in vivo survival of implanted cells. Conclusion Collectively, our data support the notion that growth in 3D cellular systems and maintenance of cell-cell contact improves exogenous cell survival following delivery into myocardium. These approaches may serve as a strategy to improve cardiovascular cell-based therapies.
Collapse
Affiliation(s)
- Michael Bauer
- Division of Cardiology and Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lifeng Kang
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Yiling Qiu
- Division of Cardiology and Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jinhui Wu
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- School of Life Science, Nanjing University, Nanjing, China
| | - Michelle Peng
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Howard H. Chen
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gulden Camci-Unal
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ahmad F. Bayomy
- Division of Cardiology and Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - David E. Sosnovik
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
- Harvard-MIT, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
- * E-mail: (RL); (AK)
| | - Ronglih Liao
- Division of Cardiology and Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail: (RL); (AK)
| |
Collapse
|
26
|
Bhat S, Kumar A. Cell proliferation on three-dimensional chitosan-agarose-gelatin cryogel scaffolds for tissue engineering applications. J Biosci Bioeng 2012; 114:663-70. [PMID: 22884715 DOI: 10.1016/j.jbiosc.2012.07.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 10/28/2022]
Abstract
Tissue engineering is a potential approach for the repair of damaged tissues or organs like skin, cartilage, bone etc. Approach utilizes the scaffolds constructed from natural or synthetic polymers fabricated by the available fabrication technologies. This study focuses on the fabrication of the scaffolds using a novel technology called cryogelation, which synthesizes the scaffolds at sub-zero temperature. We have synthesized a novel scaffold from natural polymers like chitosan, agarose and gelatin in optimized ratio using the cryogelation technology. The elasticity of the scaffold was confirmed by rheological studies which supports the utility of the scaffolds for skin and cardiac tissue engineering. Proliferation of different cell types like fibroblast and cardiac cells was analysed by scanning electron microscopy (SEM) and fluorescent microscopy. Biocompatibility of the scaffolds was tested by MTT assay with specific cell type, which showed higher proliferation of the cells on the scaffolds when compared to the two dimensional culture system. Cell proliferation of C(2)C(12) and Cos 7 cells on these scaffolds was further analysed biochemically by alamar blue test and Hoechst test. Biochemical and microscopic analysis of the different cell types on these scaffolds gives an initial insight of these scaffolds towards their utility in skin and cardiac tissue engineering.
Collapse
Affiliation(s)
- Sumrita Bhat
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | | |
Collapse
|
27
|
Guo Z, Iku S, Zheng X, Sammons RL, Kuboki Y. Three-dimensional geometry of honeycomb collagen promotes higher beating rate of myocardial cells in culture. Artif Organs 2012; 36:816-9. [PMID: 22497536 DOI: 10.1111/j.1525-1594.2012.01446.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Myocardial cells were isolated from newborn rats, cultured on a novel three-dimensional (3-D) honeycomb collagen scaffold (HC) and their morphology and beating rates compared with ones on conventional plastic dishes. On the first day, the cells attached to HC had already started beating. As time went on, the rate of beating increased as the pores of HC gradually filled with the cells, which integrated to form the cell-matrix complex. At day 8, beating reached the highest frequency of 162 beats per minute, which was twice that of the control cells on plastic dishes. It was concluded that 3-D geometry of the HC is conducive to functional growth of the myocardial tissues, and will potentially be useful for tissue engineering of myocardial regeneration.
Collapse
Affiliation(s)
- Zhikun Guo
- Key Open Laboratory for Tissue Regeneration, Department of Basic Medicine, Xinxiang Medical University, Xinxiang City, Henan, China
| | | | | | | | | |
Collapse
|
28
|
Desroches BR, Zhang P, Choi BR, King ME, Maldonado AE, Li W, Rago A, Liu G, Nath N, Hartmann KM, Yang B, Koren G, Morgan JR, Mende U. Functional scaffold-free 3-D cardiac microtissues: a novel model for the investigation of heart cells. Am J Physiol Heart Circ Physiol 2012; 302:H2031-42. [PMID: 22427522 DOI: 10.1152/ajpheart.00743.2011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To bridge the gap between two-dimensional cell culture and tissue, various three-dimensional (3-D) cell culture approaches have been developed for the investigation of cardiac myocytes (CMs) and cardiac fibroblasts (CFs). However, several limitations still exist. This study was designed to develop a cardiac 3-D culture model with a scaffold-free technology that can easily and inexpensively generate large numbers of microtissues with cellular distribution and functional behavior similar to cardiac tissue. Using micromolded nonadhesive agarose hydrogels containing 822 concave recesses (800 μm deep × 400 μm wide), we demonstrated that neonatal rat ventricular CMs and CFs alone or in combination self-assembled into viable (Live/Dead stain) spherical-shaped microtissues. Importantly, when seeded simultaneously or sequentially, CMs and CFs self-sorted to be interspersed, reminiscent of their myocardial distribution, as shown by cell type-specific CellTracker or antibody labeling. Microelectrode recordings and optical mapping revealed characteristic triangular action potentials (APs) with a resting membrane potential of -66 ± 7 mV (n = 4) in spontaneously contracting CM microtissues. Under pacing, optically mapped AP duration at 90% repolarization and conduction velocity were 100 ± 30 ms and 18.0 ± 1.9 cm/s, respectively (n = 5 each). The presence of CFs led to a twofold AP prolongation in heterogenous microtissues (CM-to-CF ratio of 1:1). Importantly, Ba(2+)-sensitive inward rectifier K(+) currents and Ca(2+)-handling proteins, including sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, were detected in CM-containing microtissues. Furthermore, cell type-specific adenoviral gene transfer was achieved, with no impact on microtissue formation or cell viability. In conclusion, we developed a novel scaffold-free cardiac 3-D culture model with several advancements for the investigation of CM and CF function and cross-regulation.
Collapse
Affiliation(s)
- B R Desroches
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kossivas F, Angeli S, Kafouris D, Patrickios CS, Tzagarakis V, Constantinides C. MRI-based morphological modeling, synthesis and characterization of cardiac tissue-mimicking materials. Biomed Mater 2012; 7:035006. [PMID: 22406555 DOI: 10.1088/1748-6041/7/3/035006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study uses standard synthetic methodologies to produce tissue-mimicking materials that match the morphology and emulate the in vivo murine and human cardiac mechanical and imaging characteristics, with dynamic mechanical analysis, atomic force microscopy (AFM), scanning electron microscopy (SEM) and magnetic resonance imaging. In accordance with such aims, poly(glycerol sebacate) (PGS) elastomeric materials were synthesized (at two different glycerol (G)-sebacic (S) acid molar ratios; the first was synthesized using a G:S molar ratio of 2:2, while the second from a 2:5 G:S molar ratio, resulting in PGS2:2 and PGS2:5 elastomers, respectively). Unlike the synthesized PGS2:2 elastomers, the PGS2:5 materials were characterized by an overall mechanical instability in their loading behavior under the three successive loading conditions tested. An oscillatory response in the mechanical properties of the synthesized elastomers was observed throughout the loading cycles, with measured increased storage modulus values at the first loading cycle, stabilizing to lower values at subsequent cycles. These elastomers were characterized at 4 °C and were found to have storage modulus values of 850 and 1430 kPa at the third loading cycle, respectively, in agreement with previously reported values of the rat and human myocardium. SEM of surface topology indicated minor degradation of synthesized materials at 10 and 20 d post-immersion in the PBS buffer solution, with a noted cluster formation on the PGS2:5 elastomers. AFM nanoindentation experiments were also conducted for the measurement of the Young modulus of the sample surface (no bulk contribution). Correspondingly, the PGS2:2 elastomer indicated significantly decreased surface Young's modulus values 20 d post-PBS immersion, compared to dry conditions (Young's modulus = 1160 ± 290 kPa (dry) and 200 ± 120 kPa (20 d)). In addition to the two-dimensional (2D) elastomers, an integrative platform for accurate construction of three-dimensional tissue-mimicking models of cardiac anatomy from 2D MR images using rapid prototyping manufacturing processes was developed. For synthesized elastomers, doping strategies with two different concentrations of the MRI contrast agent Dotarem allowed independent and concurrent control of the imaging characteristics (contrast and relaxivity) during the synthetic process for increased contrast agent absorption, with tremendous potential for non-destructive in vivo use and applications to cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Fotis Kossivas
- Laboratory of Physiology and Biomedical Imaging, Department of Mechanical and Manufacturing Engineering, School of Engineering, University of Cyprus, Nicosia, Cyprus
| | | | | | | | | | | |
Collapse
|
30
|
Mathews G, Sondergaard C, Jeffreys A, Childs W, Le BL, Sahota A, Najibi S, Nolta J, Si MS. Computational analysis of contractility in engineered heart tissue. IEEE Trans Biomed Eng 2012; 59:1429-35. [PMID: 22361653 DOI: 10.1109/tbme.2012.2187899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Engineered heart tissue (EHT) is a potential therapy for heart failure and the basis of functional in vitro assays of novel cardiovascular treatments. Self-organizing EHT can be generated in fiber form, which makes the assessment of contractile function convenient with a force transducer. Contractile function is a key parameter of EHT performance. Analysis of EHT force data is often performed manually; however, this approach is time consuming, incomplete and subjective. Therefore, the purpose of this study was to develop a computer algorithm to efficiently and objectively analyze EHT force data. This algorithm incorporates data filtering, individual contraction detection and validation, inter/intracontractile analysis and intersample analysis. We found the algorithm to be accurate in contraction detection, validation and magnitude measurement as compared to human operators. The algorithm was efficient in processing hundreds of data acquisitions and was able to determine force-length curves, force-frequency relationships and compare various contractile parameters such as peak systolic force generation. We conclude that this computer algorithm is a key adjunct to the objective and efficient assessment of EHT contractile function.
Collapse
Affiliation(s)
- Grant Mathews
- University of California Davis School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Venugopal JR, Prabhakaran MP, Mukherjee S, Ravichandran R, Dan K, Ramakrishna S. Biomaterial strategies for alleviation of myocardial infarction. J R Soc Interface 2011; 9:1-19. [PMID: 21900319 PMCID: PMC3223634 DOI: 10.1098/rsif.2011.0301] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
World Health Organization estimated that heart failure initiated by coronary artery disease and myocardial infarction (MI) leads to 29 per cent of deaths worldwide. Heart failure is one of the leading causes of death in industrialized countries and is expected to become a global epidemic within the twenty-first century. MI, the main cause of heart failure, leads to a loss of cardiac tissue impairment of left ventricular function. The damaged left ventricle undergoes progressive ‘remodelling’ and chamber dilation, with myocyte slippage and fibroblast proliferation. Repair of diseased myocardium with in vitro-engineered cardiac muscle patch/injectable biopolymers with cells may become a viable option for heart failure patients. These events reflect an apparent lack of effective intrinsic mechanism for myocardial repair and regeneration. Motivated by the desire to develop minimally invasive procedures, the last 10 years observed growing efforts to develop injectable biomaterials with and without cells to treat cardiac failure. Biomaterials evaluated include alginate, fibrin, collagen, chitosan, self-assembling peptides, biopolymers and a range of synthetic hydrogels. The ultimate goal in therapeutic cardiac tissue engineering is to generate biocompatible, non-immunogenic heart muscle with morphological and functional properties similar to natural myocardium to repair MI. This review summarizes the properties of biomaterial substrates having sufficient mechanical stability, which stimulates the native collagen fibril structure for differentiating pluripotent stem cells and mesenchymal stem cells into cardiomyocytes for cardiac tissue engineering.
Collapse
Affiliation(s)
- Jayarama Reddy Venugopal
- Healthcare and Energy Materials Laboratory, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
32
|
Orlova Y, Magome N, Liu L, Chen Y, Agladze K. Electrospun nanofibers as a tool for architecture control in engineered cardiac tissue. Biomaterials 2011; 32:5615-24. [DOI: 10.1016/j.biomaterials.2011.04.042] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
|
33
|
Hollweck T, Akra B, Häussler S, Uberfuhr P, Schmitz C, Pfeifer S, Eblenkamp M, Wintermantel E, Eissner G. A novel pulsatile bioreactor for mechanical stimulation of tissue engineered cardiac constructs. J Funct Biomater 2011; 2:107-18. [PMID: 24956300 PMCID: PMC4030939 DOI: 10.3390/jfb2030107] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 07/18/2011] [Indexed: 11/17/2022] Open
Abstract
After myocardial infarction, the implantation of stem cell seeded scaffolds on the ischemic zone represents a promising strategy for restoration of heart function. However, mechanical integrity and functionality of tissue engineered constructs need to be determined prior to implantation. Therefore, in this study a novel pulsatile bioreactor mimicking the myocardial contraction was developed to analyze the behavior of mesenchymal stem cells derived from umbilical cord tissue (UCMSC) colonized on titanium-coated polytetrafluorethylene scaffolds to friction stress. The design of the bioreactor enables a simple handling and defined mechanical forces on three seeded scaffolds at physiological conditions. The compact system made of acrylic glass, Teflon®, silicone, and stainless steel allows the comparison of different media, cells and scaffolds. The bioreactor can be gas sterilized and actuated in a standard incubator. Macroscopic observations and pressure-measurements showed a uniformly sinusoidal pulsation, indicating that the bioreactor performed well. Preliminary experiments to determine the adherence rate and morphology of UCMSC after mechanical loadings showed an almost confluent cellular coating without damage on the cell surface. In summary, the bioreactor is an adequate tool for the mechanical stress of seeded scaffolds and offers dynamic stimuli for pre-conditioning of cardiac tissue engineered constructs in vitro.
Collapse
Affiliation(s)
- Trixi Hollweck
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | - Bassil Akra
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | - Simon Häussler
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | - Peter Uberfuhr
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | - Christoph Schmitz
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | - Stefan Pfeifer
- Chair of Medical Engineering, Technische Universität München, Boltzmannstrasse 15, 85748 Garching, Germany.
| | - Markus Eblenkamp
- Chair of Medical Engineering, Technische Universität München, Boltzmannstrasse 15, 85748 Garching, Germany.
| | - Erich Wintermantel
- Chair of Medical Engineering, Technische Universität München, Boltzmannstrasse 15, 85748 Garching, Germany.
| | - Günther Eissner
- Department of Cardiac Surgery, University of Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| |
Collapse
|
34
|
Video Evaluation of the Kinematics and Dynamics of the Beating Cardiac Syncytium: An Alternative to the Langendorff Method. Int J Artif Organs 2011; 34:546-58. [DOI: 10.5301/ijao.2011.8510] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2011] [Indexed: 01/06/2023]
Abstract
Many important observations and discoveries in heart physiology have been made possible using the isolated heart method of Langendorff. Nevertheless, the Langendorff method has some limitations and disadvantages such as the vulnerability of the excised heart to contusions and injuries, the probability of preconditioning during instrumentation, the possibility of inducing tissue edema, and high oxidative stress, leading to the deterioration of the contractile function. To avoid these drawbacks associated with the use of a whole heart, we alternatively used beating mouse cardiac syncytia cultured in vitro in order to assess possible ergotropic, chronotropic, and inotropic effects of drugs. To achieve this aim, we developed a method based on image processing analysis to evaluate the kinematics and the dynamics of the drug-stimulated beating syncytia starting from the video recording of their contraction movement. In this manner, in comparison with the physiological no-drug condition, we observed progressive positive ergotropic, positive chronotropic, and positive inotropic effects of 10 μM isoproterenol (β-adrenergic agonist) and early positive ergotropic, negative chronotropic, and positive inotropic effects of 10 μM phenylephrine (α-adrenergic agonist), followed by a late phase with negative ergotropic, positive chronotropic, and negative inotropic trends. Our method permitted a systematic study of in vitro beating syncytia, producing results consistent with previous works. Consequently, it could be used in in vitro studies of beating cardiac patches, as an alternative to Langendorff's heart in biochemical and pharmacological studies, and especially when the Langendorff technique is inapplicable (e.g., in studies about human cardiac syncytium in physiological and pathological conditions, patient-tailored therapeutics, and syncytium models derived from induced pluripotent/embryonic stem cells with genetic mutations). Furthermore, the method could be helpful in heart tissue engineering and bioartificial heart research to “engineer the heart piece by piece.” In particular, the proposed method could be useful in the identification of a suitable cell source, in the development and testing of “smart” biomaterials, and in the design and use of novel bioreactors and microperfusion systems.
Collapse
|
35
|
Bai XP, Zheng HX, Fang R, Wang TR, Hou XL, Li Y, Chen XB, Tian WM. Fabrication of engineered heart tissue grafts from alginate/collagen barium composite microbeads. Biomed Mater 2011; 6:045002. [DOI: 10.1088/1748-6041/6/4/045002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
36
|
Organ engineering based on decellularized matrix scaffolds. Trends Mol Med 2011; 17:424-32. [PMID: 21514224 DOI: 10.1016/j.molmed.2011.03.005] [Citation(s) in RCA: 344] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/20/2011] [Accepted: 03/22/2011] [Indexed: 12/16/2022]
Abstract
End-organ failure is one of the major healthcare challenges in the Western world. Yet, donor organ shortage and the need for immunosuppression limit the impact of transplantation. The regeneration of whole organs could theoretically overcome these hurdles. Early milestones have been met by combining stem and progenitor cells with increasingly complex scaffold materials and culture conditions. Because the native extracellular matrix (ECM) guides organ development, repair and physiologic regeneration, it provides a promising alternative to synthetic scaffolds and a foundation for regenerative efforts. Perfusion decellularization is a novel technology that generates native ECM scaffolds with intact 3D anatomical architecture and vasculature. This review summarizes achievements to date and discusses the role of native ECM scaffolds in organ regeneration.
Collapse
|
37
|
Miyagawa S, Roth M, Saito A, Sawa Y, Kostin S. Tissue-engineered cardiac constructs for cardiac repair. Ann Thorac Surg 2011; 91:320-9. [PMID: 21172551 DOI: 10.1016/j.athoracsur.2010.09.080] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 09/22/2010] [Accepted: 09/27/2010] [Indexed: 11/16/2022]
Abstract
Several recent basic research studies have described surgical methods for cardiac repair using tissue cardiomyoplasty. This review summarizes recent advances in cardiac repair using bioengineered tissue from the viewpoint of the cardiac surgeon. We conclude that the results of many basic and preclinical studies indicate that bioengineered tissue can be adapted to conventional surgical techniques. However, no clinical studies have yet proved bioengineered tissue is effective as a treatment for human heart failure. Today's cardiac surgeons can look forward to the advent of new techniques to benefit patients who respond poorly to existing treatment for heart failure.
Collapse
Affiliation(s)
- Shigeru Miyagawa
- Department of Cardiac Surgery, Kerckhoff Clinic, Bad Nauheim, Germany.
| | | | | | | | | |
Collapse
|
38
|
Marsano A, Maidhof R, Wan LQ, Wang Y, Gao J, Tandon N, Vunjak-Novakovic G. Scaffold stiffness affects the contractile function of three-dimensional engineered cardiac constructs. Biotechnol Prog 2011; 26:1382-90. [PMID: 20945492 DOI: 10.1002/btpr.435] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We investigated the effects of the initial stiffness of a three-dimensional elastomer scaffold--highly porous poly(glycerol sebacate)--on functional assembly of cardiomyocytes cultured with perfusion for 8 days. The polymer elasticity varied with the extent of polymer cross-links, resulting in three different stiffness groups, with compressive modulus of 2.35 ± 0.03 (low), 5.28 ± 0.36 (medium), and 5.99 ± 0.40 (high) kPa. Laminin coating improved the efficiency of cell seeding (from 59 ± 15 to 90 ± 21%), resulting in markedly increased final cell density, construct contractility, and matrix deposition, likely because of enhanced cell interaction and spreading on scaffold surfaces. Compact tissue was formed in the low and medium stiffness groups, but not in the high stiffness group. In particular, the low stiffness group exhibited the greatest contraction amplitude in response to electric field pacing, and had the highest compressive modulus at the end of culture. A mathematical model was developed to establish a correlation between the contractile amplitude and the cell distribution within the scaffold. Taken together, our findings suggest that the contractile function of engineered cardiac constructs positively correlates with low compressive stiffness of the scaffold.
Collapse
Affiliation(s)
- Anna Marsano
- Dept. of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Cardiovascular Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
40
|
Dengler J, Radisic M. Tissue engineering approaches for the development of a contractile cardiac patch. Future Cardiol 2010; 3:425-34. [PMID: 19804233 DOI: 10.2217/14796678.3.4.425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cardiac tissue engineering is an emerging field that aims to enhance cardiac function in patients with heart disease or congenital malformations, by providing suitable implantable replacement tissues. Various groups have developed methods of cultivating contractile 3D cardiac grafts that display functional and morphological properties of native myocardium, integrate into native tissue and provide improvement in cardiac function upon grafting onto infarcted hearts. However, several critical goals must be met before this therapy can be implemented clinically: the identification and harvesting of an appropriate human cell source, the cultivation of relatively thick ( approximately 1 cm) constructs, and the in vitro and/or in vivo vascularization of these constructs. Advances in enabling bioreactor technologies and the identification of key parameters governing the differentiation of cardiac cells will be crucial to advancement in this field.
Collapse
Affiliation(s)
- Jana Dengler
- Institute of Biomaterials & Biomedical Engineering, Department of Chemical Engineering & Applied Chemistry, Heart & Stroke Richard/Lewar Centre of Excellence, University of Toronto, 164 College Street, Room 407, Toronto, Ontario, CA M5S3G9, Canada
| | | |
Collapse
|
41
|
Akins RE, Rockwood D, Robinson KG, Sandusky D, Rabolt J, Pizarro C. Three-dimensional culture alters primary cardiac cell phenotype. Tissue Eng Part A 2010; 16:629-41. [PMID: 20001738 PMCID: PMC2813151 DOI: 10.1089/ten.tea.2009.0458] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/10/2009] [Indexed: 01/21/2023] Open
Abstract
The directed formation of complex three-dimensional (3D) tissue architecture is a fundamental goal in tissue engineering and regenerative medicine. The growth of cells in 3D structures is expected to influence cellular phenotype and function, especially relative cell distribution, expression profiles, and responsiveness to exogenous signals; however, relatively few studies have been carried out to examine the effects of 3D reaggregation on cells from critical target organs, like the heart. Accordingly, we cultured primary cardiac ventricular cells in a 3D model system using a serum-free medium to test the hypothesis that expression profiles, multicellular organizational pathways, tissue maturation markers, and responsiveness to hormone stimulation were significantly altered in stable cell populations grown in 3D versus 2D culture. We found that distinct multi-cellular structures formed in 3D in conjunction with changes in mRNA expression profile, up-regulation of endothelial cell migratory pathways, decreases in the expression of fetal genes (Nppa and Ankrd1), and increased sensitivity to tri-iodothyronine stimulation when compared to parallel 2D cultures comprising the same cell populations. These results indicate that the culture of primary cardiac cells in 3D aggregates leads to physiologically relevant alterations in component cell phenotype consistent with cardiac ventricular tissue formation and maturation.
Collapse
Affiliation(s)
- Robert E Akins
- Nemours Biomedical Research Center, AI duPont Hospital for Children, Wilmington, Delaware 19803, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
An overview for the use of stem cells for myocardial repair and regeneration is provided. The overview provides the rationale for use of stem cells in myocardial repair. Potential stem cell types and technological challenges are highlighted.
Collapse
|
43
|
Altmann B, Welle A, Giselbrecht S, Truckenmüller R, Gottwald E. The famous versus the inconvenient - or the dawn and the rise of 3D-culture systems. World J Stem Cells 2009; 1:43-8. [PMID: 21607106 PMCID: PMC3097909 DOI: 10.4252/wjsc.v1.i1.43] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Revised: 12/09/2009] [Accepted: 12/16/2009] [Indexed: 02/06/2023] Open
Abstract
One of the greatest impacts on in vitro cell biology was the introduction of three-dimensional (3D) culture systems more than six decades ago and this era may be called the dawn of 3D-tissue culture. Although the advantages were obvious, this field of research was a "sleeping beauty" until the 1970s when multicellular spheroids were discovered as ideal tumor models. With this rebirth, organotypical culture systems became valuable tools and this trend continues to increase. While in the beginning, simple approaches, such as aggregation culture techniques, were favored due to their simplicity and convenience, now more sophisticated systems are used and are still being developed. One of the boosts in the development of new culture techniques arises from elaborate manufacturing and surface modification techniques, especially micro and nano system technologies that have either improved dramatically or have evolved very recently. With the help of these tools, it will soon be possible to generate even more sophisticated and more organotypic-like culture systems. Since 3D perfused or superfused systems are much more complex to set up and maintain compared to use of petri dishes and culture flasks, the added value of 3D approaches still needs to be demonstrated.
Collapse
Affiliation(s)
- Brigitte Altmann
- Brigitte Altmann, Alexander Welle, Stefan Giselbrecht, Eric Gottwald, Institute for Biological Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | | | | | | | | |
Collapse
|
44
|
Altmann B, Welle A, Giselbrecht S, Truckenmüller R, Gottwald E. The famous versus the inconvenient - or the dawn and the rise of 3D-culture systems. World J Stem Cells 2009. [PMID: 21607106 DOI: 10.4252/wjsc.v1.i143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the greatest impacts on in vitro cell biology was the introduction of three-dimensional (3D) culture systems more than six decades ago and this era may be called the dawn of 3D-tissue culture. Although the advantages were obvious, this field of research was a "sleeping beauty" until the 1970s when multicellular spheroids were discovered as ideal tumor models. With this rebirth, organotypical culture systems became valuable tools and this trend continues to increase. While in the beginning, simple approaches, such as aggregation culture techniques, were favored due to their simplicity and convenience, now more sophisticated systems are used and are still being developed. One of the boosts in the development of new culture techniques arises from elaborate manufacturing and surface modification techniques, especially micro and nano system technologies that have either improved dramatically or have evolved very recently. With the help of these tools, it will soon be possible to generate even more sophisticated and more organotypic-like culture systems. Since 3D perfused or superfused systems are much more complex to set up and maintain compared to use of petri dishes and culture flasks, the added value of 3D approaches still needs to be demonstrated.
Collapse
Affiliation(s)
- Brigitte Altmann
- Brigitte Altmann, Alexander Welle, Stefan Giselbrecht, Eric Gottwald, Institute for Biological Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | | | | | | | | |
Collapse
|
45
|
A 3D hybrid model for tissue growth: the interplay between cell population and mass transport dynamics. Biophys J 2009; 97:401-14. [PMID: 19619455 DOI: 10.1016/j.bpj.2009.03.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 03/08/2009] [Accepted: 03/27/2009] [Indexed: 11/21/2022] Open
Abstract
To provide theoretical guidance for the design and in vitro cultivation of bioartificial tissues, we have developed a multiscale computational model that can describe the complex interplay between cell population and mass transport dynamics that governs the growth of tissues in three-dimensional scaffolds. The model has three components: a transient partial differential equation for the simultaneous diffusion and consumption of a limiting nutrient; a cellular automaton describing cell migration, proliferation, and collision; and equations that quantify how the varying nutrient concentration modulates cell division and migration. The hybrid discrete-continuous model was parallelized and solved on a distributed-memory multicomputer to study how transport limitations affect tissue regeneration rates under conditions encountered in typical bioreactors. Simulation results show that the severity of transport limitations can be estimated by the magnitude of two dimensionless groups: the Thiele modulus and the Biot number. Key parameters including the initial seeding mode, cell migration speed, and the hydrodynamic conditions in the bioreactor are shown to affect not only the overall rate, but also the pattern of tissue growth. This study lays the groundwork for more comprehensive models that can handle mixed cell cultures, multiple nutrients and growth factors, and other cellular processes, such as cell death.
Collapse
|
46
|
Electrophysiological characterization of embryonic hippocampal neurons cultured in a 3D collagen hydrogel. Biomaterials 2009; 30:4377-83. [DOI: 10.1016/j.biomaterials.2009.04.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 04/29/2009] [Indexed: 11/24/2022]
|
47
|
Abstract
Supporting or even replacing diseased myocardium with in vitro engineered heart muscle may become a viable option for patients with heart failure. The key to success will be to (1) generate human heart muscle equivalents in vitro, (2) integrate the latter into a failing heart, (3) ensure long-term functional competence of the grafts, and (4) prevent unwanted effects including arrhythmias, inflammation/rejection, and tumor formation. Several promising tissue engineering technologies have already been developed and are presently being tested in animal models. The rapidly evolving field of human stem cell biology has in parallel identified unique cell sources of potential clinical relevance. Somatic cell reprogramming and nontransduced, nonembryonic pluripotent stem cells may be of particular interest to eventually provide patient-specific cells and tissues. Yet, limited cardiac differentiation and cell immaturity still restrict a broad application of any stem cell type in cardiac muscle engineering. Bioreactor technologies, transgenic "optimization," and growth factor, as well as physical conditioning, have been used to address these caveats. This review summarizes different tissue engineering modalities, speculates on potential clinical uses, provides an overview on cell sources that may ultimately facilitate a patient-specific application, and discusses limitations of tissue engineering-based myocardial repair.
Collapse
|
48
|
Abstract
Children with severe congenital malformations, such as single-ventricle anomalies, have a daunting prognosis. Heart transplantation would be a therapeutic option but is restricted due to a lack of suitable donor organs and, even in case of successful heart transplantation, lifelong immune suppression would frequently be associated with a number of serious side effects. As an alternative to heart transplantation and classical cardiac reconstructive surgery, tissue-engineered myocardium might become available to augment hypomorphic hearts and/or provide new muscle material for complex myocardial reconstruction. These potential applications of tissue engineered myocardium will, however, impose major challenges to cardiac tissue engineers as well as heart surgeons. This review will provide an overview of available cardiac tissue-engineering technologies, discuss limitations, and speculate on a potential application of tissue-engineered heart muscle in pediatric heart surgery.
Collapse
|
49
|
Marsano A, Maidhof R, Tandon N, Gao J, Wang Y, Vunjak-Novakovic G. Engineering of functional contractile cardiac tissues cultured in a perfusion system. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2009; 2008:3590-3. [PMID: 19163485 DOI: 10.1109/iembs.2008.4649982] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Overcoming the limitations of diffusional transport in conventional culture systems remains an open issue for successfully generating thick, compact and functional cardiac tissues. Previously, it was shown that perfusion systems enhance the yield and uniformity of cell seeding and cell survival in thick cardiac constructs. The aim of our study was to form highly functional cardiac constructs starting from spatially uniform, high density cell seeded constructs. Disk-shaped elastomeric poly(glycerol sebacate) scaffolds were seeded with neonatal rat cardiomyocytes and cultured for eight days with direct perfusion of culture medium or statically in a six-well plate. In the perfusion experimental group, the integrity of some disks was well maintained, whereas in others a central hole was formed, resulting in ring-shaped constructs. This allowed us to also study the effects of construct geometry and of interstitial flow versus channel perfusion. The ring-shaped constructs appeared to have a denser and more uniform deposition of extracellular matrix. In response to electrical stimulation, the fractional area change of the ring-shaped constructs was 7.3 and 2.7 times higher than for disk-shaped tissues cultured in perfusion or statically, respectively. These findings suggest that a combination of many factors, including scaffold elasticity and geometry and the type of perfusion system applied, need to be considered in order to engineer a cardiac construct with high contractile activity.
Collapse
Affiliation(s)
- A Marsano
- Department of Biomedical Engineering, at the Columbia University, New York, USA
| | | | | | | | | | | |
Collapse
|
50
|
Placzek MR, Chung IM, Macedo HM, Ismail S, Mortera Blanco T, Lim M, Cha JM, Fauzi I, Kang Y, Yeo DCL, Ma CYJ, Polak JM, Panoskaltsis N, Mantalaris A. Stem cell bioprocessing: fundamentals and principles. J R Soc Interface 2009; 6:209-32. [PMID: 19033137 PMCID: PMC2659585 DOI: 10.1098/rsif.2008.0442] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In recent years, the potential of stem cell research for tissue engineering-based therapies and regenerative medicine clinical applications has become well established. In 2006, Chung pioneered the first entire organ transplant using adult stem cells and a scaffold for clinical evaluation. With this a new milestone was achieved, with seven patients with myelomeningocele receiving stem cell-derived bladder transplants resulting in substantial improvements in their quality of life. While a bladder is a relatively simple organ, the breakthrough highlights the incredible benefits that can be gained from the cross-disciplinary nature of tissue engineering and regenerative medicine (TERM) that encompasses stem cell research and stem cell bioprocessing. Unquestionably, the development of bioprocess technologies for the transfer of the current laboratory-based practice of stem cell tissue culture to the clinic as therapeutics necessitates the application of engineering principles and practices to achieve control, reproducibility, automation, validation and safety of the process and the product. The successful translation will require contributions from fundamental research (from developmental biology to the 'omics' technologies and advances in immunology) and from existing industrial practice (biologics), especially on automation, quality assurance and regulation. The timely development, integration and execution of various components will be critical-failures of the past (such as in the commercialization of skin equivalents) on marketing, pricing, production and advertising should not be repeated. This review aims to address the principles required for successful stem cell bioprocessing so that they can be applied deftly to clinical applications.
Collapse
Affiliation(s)
- Mark R Placzek
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|