1
|
Behnam R, Dinari M, Behzad T. Innovative unsaturated polyurethane for cardiac regeneration: Enhancing cytocompatibility and gene expression through gelatin modification. Int J Biol Macromol 2025; 316:144659. [PMID: 40441578 DOI: 10.1016/j.ijbiomac.2025.144659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/11/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
Developing a biomimetic platform that can effectively support myocardial repair and regeneration after a heart attack remains a major challenge for researchers. Polyurethanes (PU), known for their versatile properties, are increasingly being recognized as promising materials for creating cardiac patches. This research study developed the synthesis of a novel unsaturated chain extender via the Baylis-Hillman reaction for incorporation into the backbone of PU to create the unsaturated bridge for post-modification response. Thiol-ene chemistry performed post-modification of unsaturated hard domains of PU with thiolated gelatin as another reactant. Modification of the hard segment domains of PU with gelatin can have a synergistic effect on fabricating a new platform for repairing cardiac tissue. The films were characterized by Attenuated Total Reflectance Fourier Transform Infrared Spectroscopy (ATR-FTIR), Nuclear Magnetic Resonance (NMR), mechanical tests, in vitro degradation rate, water contact angle measurement, and swelling measurement. Metabolic activity and cytocompatibility of the cells onto the films were assessed by MTT assay. An investigation of cardiac-specific marker expression was conducted using immunofluorescence staining and quantitative real-time PCR. The findings showed that none of the substances were cytotoxic to cardiomyocytes, and that the expression of cadiac-specific genes, such as Troponin T, GATA4, Connexin43, and Alpha-SMA, was higher than that of the control group.
Collapse
Affiliation(s)
- Reza Behnam
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Mohammad Dinari
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Tayebeh Behzad
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
2
|
Meng Y, Xu L, Cheng G. Bioelectronics hydrogels for implantable cardiac and brain disease medical treatment application. Int J Biol Macromol 2025; 299:139945. [PMID: 39837454 DOI: 10.1016/j.ijbiomac.2025.139945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Hydrogel-based bioelectronic systems offer significant benefits for point-of-care diagnosis, treatment of cardiac and cerebral disease, surgical procedures, and other medical applications, ushering in a new era of advancements in medical technology. Progress in hydrogel-based bioelectronics has advanced from basic instrument and sensing capabilities to sophisticated multimodal perceptions and feedback systems. Addressing challenges related to immune responses and inflammation regulation after implantation, physiological dynamic mechanism, biological toxicology as well as device size, power consumption, stability, and signal conversion is crucial for the practical implementation of hydrogel-based bioelectronics in medical implants. Therefore, further exploration of hydrogel-based bioelectronics is imperative, and a comprehensive review is necessary to steer the development of these technologies for use in implantable therapies for cardiac and brain/neural conditions. In this review, a concise overview is provided on the fundamental principles underlying ionic electronic and ionic bioelectronic mechanisms. Additionally, a comprehensive examination is conducted on various bioelectronic materials integrated within hydrogels for applications in implantable medical treatments. The analysis encompasses a detailed discussion on the representative structures and physical attributes of hydrogels. This includes an exploration of their intrinsic properties such as mechanical strength, dynamic capabilities, shape-memory features, stability, stretchability, and water retention characteristics. Moreover, the discussion extends to properties related to interactions with tissues or the environment, such as adhesiveness, responsiveness, and degradability. The intricate relationships between the structure and properties of hydrogels are thoroughly examined, along with an elucidation of how these properties influence their applications in implantable medical treatments. The review also delves into the processing techniques and characterization methods employed for hydrogels. Furthermore, recent breakthroughs in the applications of hydrogels are logically explored, covering aspects such as materials, structure, properties, functions, fabrication procedures, and hybridization with other materials. Finally, the review concludes by outlining the future prospects and challenges associated with hydrogels-based bioelectronics systems.
Collapse
Affiliation(s)
- Yanfang Meng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China
| | - Lin Xu
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| | - Guanggui Cheng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| |
Collapse
|
3
|
Li T, Wan Z, Wang Q, Qiao F, Pan G, Zhao C, Zhu Y, Zhou H, Tan Y, Zhou Z, Zhang D. Utilizing Tissues Self-Assembled in Fiber Optic-Based "Chinese Guzheng Strings" for Contractility Sensing and Drug Efficacy Evaluation: A Practical Approach. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406144. [PMID: 39822158 DOI: 10.1002/smll.202406144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/25/2024] [Indexed: 01/19/2025]
Abstract
Recent advances in drug design and compound synthesis have highlighted the increasing need for effective methods of toxicity evaluation. A specialized force sensor, known as the light wavelength-encoded "Chinese guzheng" is developed. This innovative sensor is equipped with optical fiber strings and utilizes a wavelength-encoded fiber Bragg grating (FBG) that is chemically etched to reduce its diameter. This design allows the sensor to detect minimal forces as low as l µN. This sensor is successfully applied to monitor human-induced pluripotent stem cell-derived human-engineered heart tissue (hEHT) models that can self-assemble and contact optical fiber-based strings. The sensor detects micro newton contraction forces in real-time by measuring the wavelength drift resulting from hEHT contractions. In addition, the sensor is precise and durable, exhibiting a fatigue resistance of up to 800 000 cycles, making it suitable for long-term monitoring. The device effectively measured the contractile force of the hEHTs under various physiological conditions, including natural contraction, electrical stimulation, and stretching. Moreover, multichannel detection enables the study and demonstration of short- and long-term effectiveness of multiple drugs. This breakthrough sensor addresses the critical need for high-precision real-time monitoring in drug evaluation and provides a solid foundation for screening drugs to treat cardiomyopathy.
Collapse
Affiliation(s)
- Tianliang Li
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Zhongjun Wan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Stem Cells and Tissue Engineering Manufacture Center, School of Life Science, Hubei University, Wuhan, Hubei, 430062, China
| | - Qian'ao Wang
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Feng Qiao
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Ganlin Pan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Stem Cells and Tissue Engineering Manufacture Center, School of Life Science, Hubei University, Wuhan, Hubei, 430062, China
| | - Chen Zhao
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Yongwen Zhu
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Haotian Zhou
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Yuegang Tan
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Zude Zhou
- School of Mechanical and Electronic Engineering, Wuhan University of Technology, Wuhan, Hubei, 430070, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Stem Cells and Tissue Engineering Manufacture Center, School of Life Science, Hubei University, Wuhan, Hubei, 430062, China
- Cardiovascular Institute, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| |
Collapse
|
4
|
Oh EJ, Kim HM, Kwak S, Huh C, Chung HY. The Formation of Human Arteriovenous Malformation Organoids and Their Characteristics. Cells 2024; 13:1955. [PMID: 39682704 PMCID: PMC11640006 DOI: 10.3390/cells13231955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Arteriovenous malformations (AVMs) are characterized by direct connections between arteries and veins without intervening capillaries, with the concomitant formation of abnormal vascular networks associated with angiogenesis. However, the current understanding of the diagnosis and treatment of AVMs is limited, and no in vitro disease models exist at present for studying this condition. In this study, we produced endothelial cells (ECs) in two-dimensional cultures and three-dimensional (3D) blood vessel organoids (BVOs), comparing gene expression profiles between normal and AVM organoids. The normal and AVM organoids were examined via immunofluorescence staining using CD31 and phalloidin. The AVM organoids showed significantly higher expression levels of CD31 and phalloidin than the normal organoids. Genes such as FSTL1, associated with angiogenesis, showed significantly higher expression in the AVM organoids than in the normal organoids. In contrast, the MARCKS gene exhibited no significant difference in expression between the two types of organoids. The capillaries and related CSPG4 genes exhibited the lowest expression in the 3D AVM organoids. Furthermore, hsa-mir-135b-5p, a small RNA related to AVMs, showed elevated expression in AVM tissues and significantly higher levels in 3D AVM organoids. In our study, we were able to successfully establish AVM organoids (hBVOs) containing ECs and mural cells through advancements in stem cell and tissue engineering. These organoids serve as valuable models for investigating disease mechanisms, drug development, and screening potential therapeutic interventions in drug discovery. These findings contribute essential insights for the development of treatment strategies targeting AVMs.
Collapse
Affiliation(s)
- Eun Jung Oh
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (E.J.O.); (H.M.K.); (S.K.)
| | - Hyun Mi Kim
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (E.J.O.); (H.M.K.); (S.K.)
| | - Suin Kwak
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (E.J.O.); (H.M.K.); (S.K.)
| | - Chanhoe Huh
- College of Medicine, Yonsei University, Seoul 03722, Republic of Korea;
| | - Ho Yun Chung
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (E.J.O.); (H.M.K.); (S.K.)
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
5
|
Yildirim Y, Degener L, Reuter L, Petersen J, Gabel L, Sommer A, Pahrmann C, Reichenspurner H, Pecha S. Evaluation of cell survival in different 3D-printed geometric shapes of human iPSC-derived engineered heart tissue. Artif Organs 2024; 48:1251-1263. [PMID: 39041632 DOI: 10.1111/aor.14833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVES Engineered Heart Tissue (EHT) is a promising tool to repair heart muscle defects and can additionally be used for drug testing. Due to the absence of an in vitro vascularization, EHT geometry crucially impacts nutrient and oxygen supply by diffusion capacity. We analyzed cardiomyocyte survival in different EHT geometries. METHODS Different geometries with varying surface-area-to-volume-ratios were calculated (structure A (Ring) AS/V = 58.47 mm2/440 μL3, structure B (Infinity) 25.86 mm2/440 μL3). EHTs were generated from hiPSC-derived cardiomyocytes (4 × 106) and a fibrin/thrombin hydrogel. Cell viability was evaluated by RT-PCR, cytometric studies, and Bioluminescence imaging. RESULTS Using 3D-printed casting molds, spontaneously beating EHTs can be generated in various geometric forms. At day 7, the RT-PCR analyses showed a significantly higher Troponin-T value in ring EHTs, compared to infinity EHTs. In cytometric studies, we evaluated 15% more Troponin-T positive cells in ring (73% ± 12%), compared to infinity EHTs (58% ± 11%, p = 0.04). BLI visualized significantly higher cell survival in ring EHTs (ROI = A: 1.14 × 106 p/s and B: 8.47 × 105 p/s, p < 0.001) compared to infinity EHTs during longitudinal cultivation process. CONCLUSION Use of 3D-printing allows the creation of EHTs in all desired geometric shapes. The geometry with an optimized surface-area-to-volume-ratio (ring EHT) demonstrated a significantly higher cell survival measured by RT-PCR, Bioluminescence imaging, and cytometric studies using FACS analysis.
Collapse
Affiliation(s)
- Yalin Yildirim
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Louisa Degener
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Lukas Reuter
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Johannes Petersen
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lilian Gabel
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Annika Sommer
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Christiane Pahrmann
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Simon Pecha
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
6
|
Tetsuka H, Gobbi S, Hatanaka T, Pirrami L, Shin SR. Wirelessly steerable bioelectronic neuromuscular robots adapting neurocardiac junctions. Sci Robot 2024; 9:eado0051. [PMID: 39321274 DOI: 10.1126/scirobotics.ado0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024]
Abstract
Biological motions of native muscle tissues rely on the nervous system to interface movement with the surrounding environment. The neural innervation of muscles, crucial for regulating movement, is the fundamental infrastructure for swiftly responding to changes in body tissue requirements. This study introduces a bioelectronic neuromuscular robot integrated with the motor nervous system through electrical synapses to evoke cardiac muscle activities and steer robotic motion. Serving as an artificial brain and wirelessly regulating selective neural activation to initiate robot fin motion, a wireless frequency multiplexing bioelectronic device is used to control the robot. Frequency multiplexing bioelectronics enables the control of the robot locomotion speed and direction by modulating the flapping of the robot fins through the wireless motor innervation of cardiac muscles. The robots demonstrated an average locomotion speed of ~0.52 ± 0.22 millimeters per second, fin-flapping frequency up to 2.0 hertz, and turning locomotion path curvature of ~0.11 ± 0.04 radians per millimeter. These systems will contribute to the expansion of biohybrid machines into the brain-to-motor frontier for developing autonomous biohybrid systems capable of advanced adaptive motor control and learning.
Collapse
Affiliation(s)
- Hiroyuki Tetsuka
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, MA 02139, USA
- Research Strategy Office, Toyota Research Institute of North America, Toyota Motor North America, 1555 Woodridge Avenue, Ann Arbor, MI 48105, USA
| | - Samuele Gobbi
- iPrint Institute, HEIA-FR, HES-SO University of Applied Sciences and Arts Western Switzerland, Fribourg 1700, Switzerland
| | - Takaaki Hatanaka
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, MA 02139, USA
- Research Strategy Office, Toyota Research Institute of North America, Toyota Motor North America, 1555 Woodridge Avenue, Ann Arbor, MI 48105, USA
| | - Lorenzo Pirrami
- iPrint Institute, HEIA-FR, HES-SO University of Applied Sciences and Arts Western Switzerland, Fribourg 1700, Switzerland
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
7
|
Majumder R. In silico thermal control of spiral wave dynamics in excitable cardiac tissue. BIOPHYSICAL REPORTS 2024; 4:100170. [PMID: 38960373 PMCID: PMC11304022 DOI: 10.1016/j.bpr.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Self-organizing spiral waves of excitation occur in many complex excitable systems. In the heart, for example, they are associated with the occurrence of fatal cardiac arrhythmias such as tachycardia and fibrillation, which can lead to sudden cardiac death. The control of these waves is therefore necessary for the treatment of the disease. In this letter, I present an innovative approach to control cardiac arrhythmias using low (nonfreezing) temperatures. This approach differs from all previous established techniques in that it involves no drugs, no genetic modification, no injection of foreign bodies, no application of voltage shocks (high or low, single or pulsed), and no curative damage to the heart. It relies on regional cooling of cardiac tissue to create a transient inhomogeneity in the electrophysiological properties. This inhomogeneity can then be manipulated to control the dynamics of the reentrant waves. This approach is, to my knowledge, the most sustainable theoretical proposal for the treatment of cardiac arrhythmias in the clinic.
Collapse
|
8
|
Liu Y, Kamran R, Han X, Wang M, Li Q, Lai D, Naruse K, Takahashi K. Human heart-on-a-chip microphysiological system comprising endothelial cells, fibroblasts, and iPSC-derived cardiomyocytes. Sci Rep 2024; 14:18063. [PMID: 39117679 PMCID: PMC11310341 DOI: 10.1038/s41598-024-68275-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
In recent years, research on organ-on-a-chip technology has been flourishing, particularly for drug screening and disease model development. Fibroblasts and vascular endothelial cells engage in crosstalk through paracrine signaling and direct cell-cell contact, which is essential for the normal development and function of the heart. Therefore, to faithfully recapitulate cardiac function, it is imperative to incorporate fibroblasts and vascular endothelial cells into a heart-on-a-chip model. Here, we report the development of a human heart-on-a-chip composed of induced pluripotent stem cell (iPSC)-derived cardiomyocytes, fibroblasts, and vascular endothelial cells. Vascular endothelial cells cultured on microfluidic channels responded to the flow of culture medium mimicking blood flow by orienting themselves parallel to the flow direction, akin to in vivo vascular alignment in response to blood flow. Furthermore, the flow of culture medium promoted integrity among vascular endothelial cells, as evidenced by CD31 staining and lower apparent permeability. The tri-culture condition of iPSC-derived cardiomyocytes, fibroblasts, and vascular endothelial cells resulted in higher expression of the ventricular cardiomyocyte marker IRX4 and increased contractility compared to the bi-culture condition with iPSC-derived cardiomyocytes and fibroblasts alone. Such tri-culture-derived cardiac tissues exhibited cardiac responses similar to in vivo hearts, including an increase in heart rate upon noradrenaline administration. In summary, we have achieved the development of a heart-on-a-chip composed of cardiomyocytes, fibroblasts, and vascular endothelial cells that mimics in vivo cardiac behavior.
Collapse
Affiliation(s)
- Yun Liu
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Rumaisa Kamran
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Xiaoxia Han
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Mengxue Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Qiang Li
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Daoyue Lai
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan.
| |
Collapse
|
9
|
Cyr JA, Burdett C, Pürstl JT, Thompson RP, Troughton SC, Sinha S, Best SM, Cameron RE. Characterizing collagen scaffold compliance with native myocardial strains using an ex-vivo cardiac model: The physio-mechanical influence of scaffold architecture and attachment method. Acta Biomater 2024; 184:239-253. [PMID: 38942187 DOI: 10.1016/j.actbio.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/30/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Applied to the epicardium in-vivo, regenerative cardiac patches support the ventricular wall, reduce wall stresses, encourage ventricular wall thickening, and improve ventricular function. Scaffold engraftment, however, remains a challenge. After implantation, scaffolds are subject to the complex, time-varying, biomechanical environment of the myocardium. The mechanical capacity of engineered tissue to biomimetically deform and simultaneously support the damaged native tissue is crucial for its efficacy. To date, however, the biomechanical response of engineered tissue applied directly to live myocardium has not been characterized. In this paper, we utilize optical imaging of a Langendorff ex-vivo cardiac model to characterize the native deformation of the epicardium as well as that of attached engineered scaffolds. We utilize digital image correlation, linear strain, and 2D principal strain analysis to assess the mechanical compliance of acellular ice templated collagen scaffolds. Scaffolds had either aligned or isotropic porous architecture and were adhered directly to the live epicardial surface with either sutures or cyanoacrylate glue. We demonstrate that the biomechanical characteristics of native myocardial deformation on the epicardial surface can be reproduced by an ex-vivo cardiac model. Furthermore, we identified that scaffolds with unidirectionally aligned pores adhered with suture fixation most accurately recapitulated the deformation of the native epicardium. Our study contributes a translational characterization methodology to assess the physio-mechanical performance of engineered cardiac tissue and adds to the growing body of evidence showing that anisotropic scaffold architecture improves the functional biomimetic capacity of engineered cardiac tissue. STATEMENT OF SIGNIFICANCE: Engineered cardiac tissue offers potential for myocardial repair, but engraftment remains a challenge. In-vivo, engineered scaffolds are subject to complex biomechanical stresses and the mechanical capacity of scaffolds to biomimetically deform is critical. To date, the biomechanical response of engineered scaffolds applied to live myocardium has not been characterized. In this paper, we utilize optical imaging of an ex-vivo cardiac model to characterize the deformation of the native epicardium and scaffolds attached directly to the heart. Comparing scaffold architecture and fixation method, we demonstrate that sutured scaffolds with anisotropic pores aligned with the native alignment of the superficial myocardium best recapitulate native deformation. Our study contributes a physio-mechanical characterization methodology for cardiac tissue engineering scaffolds.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Clare Burdett
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Julia T Pürstl
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Robert P Thompson
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Samuel C Troughton
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Serena M Best
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
10
|
S. AD, P. SPA, Naveen J, Khan T, Khahro SH. Advancement in biomedical implant materials-a mini review. Front Bioeng Biotechnol 2024; 12:1400918. [PMID: 39021364 PMCID: PMC11252025 DOI: 10.3389/fbioe.2024.1400918] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/31/2024] [Indexed: 07/20/2024] Open
Abstract
Metal alloys like stainless steel, titanium, and cobalt-chromium alloys are preferable for bio-implants due to their exceptional strength, tribological properties, and biocompatibility. However, long-term implantation of metal alloys can lead to inflammation, swelling, and itching because of ion leaching. To address this issue, polymers are increasingly being utilized in orthopedic applications, replacing metallic components such as bone fixation plates, screws, and scaffolds, as well as minimizing metal-on-metal contact in total hip and knee joint replacements. Ceramics, known for their hardness, thermal barrier, wear, and corrosion resistance, find extensive application in electrochemical, fuel, and biomedical industries. This review delves into a variety of biocompatible materials engineered to seamlessly integrate with the body, reducing adverse reactions like inflammation, toxicity, or immune responses. Additionally, this review examines the potential of various biomaterials including metals, polymers, and ceramics for implant applications. While metallic biomaterials remain indispensable, polymers and ceramics show promise as alternative options. However, surface-modified metallic materials offer a hybrid effect, combining the strengths of different constituents. The future of biomedical implant materials lies in advanced fabrication techniques and personalized designs, facilitating tailored solutions for complex medical needs.
Collapse
Affiliation(s)
- Ashish Daniel S.
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, India
| | - Suya Prem Anand P.
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, India
| | - Jesuarockiam Naveen
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, India
| | - Tabrej Khan
- Department of Engineering Management, Faculty of Engineering, Prince Sultan University, Riyadh, Saudi Arabia
| | - Shabir Hussain Khahro
- Department of Engineering Management, Faculty of Engineering, Prince Sultan University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Hosseini S, Kong X, Givens SE, Ogle BM, Tolkacheva EG. Use of Optical Mapping to assess the level of maturation of Engineered Heart Tissues. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40040191 DOI: 10.1109/embc53108.2024.10782946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
This research discusses the use of high-resolution optical mapping technique to quantify electrophysiological characteristics of engineered heart tissues (EHTs) that can be used to assess their maturation. Specifically, this study utilizes two electrophysiological parameters: maximum derivative of voltage over time (dv/dt max) and activation time (AT) to the end of assessing EHT functionality. We also investigated the importance of removing noise and artifacts and applying different binary masks to optimize these parameters. Finally, we compare dv/dt max and AT between EHT and murine adult cardiac tissue and demonstrated the sensitivity of these parameters in detecting the electrophysiological characteristics of both primary and engineered tissues.
Collapse
|
12
|
Katili PA, Karima AP, Azwani W, Antarianto RD, Djer MM. Application of Human Induced Pluripotent Stem Cells for Tissue Engineered Cardiomyocyte Modelling. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023; 9:431-446. [DOI: 10.1007/s40883-023-00294-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/06/2025]
Abstract
Abstract
Purpose
Cardiac
tissue engineering opens up opportunities for regenerative therapy in heart diseases. Current technologies improve engineered cardiac tissue characteristics by combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with non-cardiomyocytes, selective biomaterials, and additional growth factors. Animal models are still required to determine cardiac patches’ overall in vivo effect before initiating human trials. Here, we review the current in vivo studies of cardiac patches using hiPSC-CMs.
Methods
We performed a literature search for studies on cardiac patch in vivo application and compared outcomes based on cell engraftment, functional changes, and safety profiles.
Results
Present studies confirm the beneficial results of combining hiPSC-CMs with other cardiac cell lineages and biomaterials. They improved the functional capacity of the heart, showed a reduction in infarct size, and initiated an adaptive inflammatory process through neovascularisation.
Conclusion
The cardiac patch is currently the most effective delivery system, proving safety and improvements in animal models, which are suggested to be the role of the paracrine mechanism. Further studies should focus on honing in vitro patch characteristics to achieve ideal results.
Lay Summary
Cardiac tissue engineering answers the demand for regenerative therapy in heart diseases. Combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with biomaterials and growth factors in cardiac patches improves the heart’s structural and functional characteristics. This delivery system is safe and efficient for delivering many cells and minimising cellular loss in vivo. Rat and porcine models of ischemic and non-ischemic heart diseases demonstrated the benefits of this therapy, which include cell engraftment, reduced infarct size, and increased left ventricular (LV) systolic function, with no reported critical adverse events. These reports sufficiently provide evidence of feasible improvements to proceed towards further trials.
Collapse
|
13
|
Fan C, He J, Xu S, Yan J, Jin L, Dai J, Hu B. Advances in biomaterial-based cardiac organoids. BIOMATERIALS ADVANCES 2023; 153:213502. [PMID: 37352743 DOI: 10.1016/j.bioadv.2023.213502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular disease (CVD) is one of the important causes of death worldwide. The incidence and mortality rates are increasing annually with the intensification of social aging. The efficacy of drug therapy is limited in individuals suffering from severe heart failure due to the inability of myocardial cells to undergo regeneration and the challenging nature of cardiac tissue repair following injury. Consequently, surgical transplantation stands as the most efficient approach for treatment. Nevertheless, the shortage of donors and the considerable number of heart failure patients worldwide, estimated at 26 million, results in an alarming treatment deficit, with only around 5000 heart transplants feasible annually. The existing major alternatives, such as mechanical or xenogeneic hearts, have significant flaws, such as high cost and rejection, and are challenging to implement for large-scale, long-term use. An organoid is a three-dimensional (3D) cell tissue that mimics the characteristics of an organ. The critical application has been rated in annual biotechnology by authoritative journals, such as Science and Cell. Related industries have achieved rapid growth in recent years. Based on this technology, cardiac organoids are expected to pave the way for viable heart repair and treatment and play an essential role in pathological research, drug screening, and other areas. This review centers on the examination of biomaterials employed in cardiac repair, strategies employed for the reconstruction of cardiac structure and function, clinical investigations pertaining to cardiac repair, and the prospective applications of cardiac organoids. From basic research to clinical practice, the current status, latest progress, challenges, and prospects of biomaterial-based cardiac repair are summarized and discussed, providing a reference for future exploration and development of cardiac regeneration strategies.
Collapse
Affiliation(s)
- Caixia Fan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jiaxiong He
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Sijia Xu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China
| | - Junyan Yan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Lifang Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China.
| | - Baowei Hu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
14
|
Yadid M, Hagel M, Labro MB, Le Roi B, Flaxer C, Flaxer E, Barnea AR, Tejman‐Yarden S, Silberman E, Li X, Rauti R, Leichtmann‐Bardoogo Y, Yuan H, Maoz BM. A Platform for Assessing Cellular Contractile Function Based on Magnetic Manipulation of Magnetoresponsive Hydrogel Films. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207498. [PMID: 37485582 PMCID: PMC10520681 DOI: 10.1002/advs.202207498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 06/08/2023] [Indexed: 07/25/2023]
Abstract
Despite significant advancements in in vitro cardiac modeling approaches, researchers still lack the capacity to obtain in vitro measurements of a key indicator of cardiac function: contractility, or stroke volume under specific loading conditions-defined as the pressures to which the heart is subjected prior to and during contraction. This work puts forward a platform that creates this capability, by providing a means of dynamically controlling loading conditions in vitro. This dynamic tissue loading platform consists of a thin magnetoresponsive hydrogel cantilever on which 2D engineered myocardial tissue is cultured. Exposing the cantilever to an external magnetic field-generated by positioning magnets at a controlled distance from the cantilever-causes the hydrogel film to stretch, creating tissue load. Next, cell contraction is induced through electrical stimulation, and the force of the contraction is recorded, by measuring the cantilever's deflection. Force-length-based measurements of contractility are then derived, comparable to clinical measurements. In an illustrative application, the platform is used to measure contractility both in untreated myocardial tissue and in tissue exposed to an inotropic agent. Clear differences are observed between conditions, suggesting that the proposed platform has significant potential to provide clinically relevant measurements of contractility.
Collapse
Affiliation(s)
- Moran Yadid
- The Azrieli Faculty of MedicineBar Ilan University8 Henrietta Szold St.Safed1311502Israel
- The Shmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv69978Israel
| | - Mario Hagel
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | | | - Baptiste Le Roi
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Carina Flaxer
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Eli Flaxer
- AFEKA – Tel‐Aviv Academic College of EngineeringTel‐Aviv69107Israel
| | - A. Ronny Barnea
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Shai Tejman‐Yarden
- The Edmond J. Safra International Congenital Heart CenterSheba Medical CenterRamat Gan52621Israel
- The Engineering Medical Research LabSheba Medical CenterRamat Gan52621Israel
- The Sackler School of MedicineTel Aviv UniversityTel Aviv69978Israel
| | - Eric Silberman
- The Shmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv69978Israel
| | - Xin Li
- Shenzhen Key Laboratory of Soft Mechanics and Smart ManufacturingDepartment of Mechanics and Aerospace EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Rossana Rauti
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbino61029Italy
| | | | - Hongyan Yuan
- Shenzhen Key Laboratory of Soft Mechanics and Smart ManufacturingDepartment of Mechanics and Aerospace EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Ben M. Maoz
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
- The Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv69978Israel
| |
Collapse
|
15
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
16
|
Ainsworth MJ, Chirico N, de Ruijter M, Hrynevich A, Dokter I, Sluijter JPG, Malda J, van Mil A, Castilho M. Convergence of melt electrowriting and extrusion-based bioprinting for vascular patterning of a myocardial construct. Biofabrication 2023; 15:035025. [PMID: 37343567 DOI: 10.1088/1758-5090/ace07f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/21/2023] [Indexed: 06/23/2023]
Abstract
To progress cardiac tissue engineering strategies closer to the clinic, thicker constructs are required to meet the functional need following a cardiac event. Consequently, pre-vascularization of these constructs needs to be investigated to ensure survival and optimal performance of implantable engineered heart tissue. The aim of this research is to investigate the potential of combining extrusion-based bioprinting (EBB) and melt electrowriting for the fabrication of a myocardial construct with a precisely patterned pre-vascular pathway. Gelatin methacryloyl (GelMA) was investigated as a base hydrogel for the respective myocardial and vascular bioinks with collagen, Matrigel and fibrinogen as interpenetrating polymers to support myocardial functionality. Subsequently, extrusion-based printability and viability were investigated to determine the optimal processing parameters for printing into melt electrowritten meshes. Finally, an anatomically inspired vascular pathway was implemented in a dual EBB set-up into melt electrowritten meshes, creating a patterned pre-vascularized myocardial construct. It was determined that a blend of 5% GelMA and 0.8 mg·ml-1collagen with a low crosslinked density was optimal for myocardial cellular arrangement and alignment within the constructs. For the vascular fraction, the optimized formulation consisted of 5% GelMA, 0.8 mg·ml-1collagen and 1 mg·ml-1fibrinogen with a higher crosslinked density, which led to enhanced vascular cell connectivity. Printability assessment confirmed that the optimized bioinks could effectively fill the microfiber mesh while supporting cell viability (∼70%). Finally, the two bioinks were applied using a dual EBB system for the fabrication of a pre-vascular pathway with the shape of a left anterior descending artery within a myocardial construct, whereby the distinct cell populations could be visualized in their respective patterns up to D14. This research investigated the first step towards developing a thick engineered cardiac tissue construct in which a pre-vascularization pathway is fabricated within a myocardial construct.
Collapse
Affiliation(s)
- Madison Jade Ainsworth
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nino Chirico
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mylène de Ruijter
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Andrei Hrynevich
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - Inge Dokter
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alain van Mil
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
17
|
Sun Q, Yao J, Zhang Z, Li J, Zhang X, Wang H, Du X, Li M, Zhao Y. Facile fabrication of biocompatible injectable blended polymeric hydrogel with bioactive nanoformulation to improving cardiac tissue regeneration efficiency after myocardial infarction for nursing care potential applications. Nanotoxicology 2023; 17:432-448. [PMID: 37724376 DOI: 10.1080/17435390.2023.2252921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/17/2023] [Indexed: 09/20/2023]
Abstract
Recent years, cardiac vascular disease has arisen owing to acute myocardial infarction (MI) and heart failure leading to death worldwide. Various treatments are available for MI in modern medicine such as implantation of devices, pharmaceutical therapy, and transplantation of organs, nonetheless, it has many complications in finding an organ donor, devices for stenosis, high intrusiveness and long-time hospitalization. To overcome these problems, we have designed and developed a novel hydrogel material with a combination of Se NPs loaded poly(ethylene glycol)/tannic acid (PEG/TA) hydrogel for the treatment of acute MI repair. Herein, Se NPs were characterized by effective analytical and spectroscopic techniques. In vitro cell compatibility and anti-oxidant analyses were examined on human cardiomyocytes in different concentrations of Se NPs and appropriate Se NPs loaded hydrogel samples to demonstrate its greater suitability for in vivo cardiac applications. In vivo investigations of MI mice models injected with Se hydrogels established that LV wall thickness was conserved significantly from the value of 235.6 µm to 390 µm. In addition, the relative scar thickness (33.6%) and infarct size (17.1%) of the MI model were enormously reduced after injection of Se hydrogel when compared to the Se NPs and control (MI) sample, respectively, which confirmed that Se introduced hydrogel have greatly influenced on the restoration of the infarcted heart. Based on the investigated results of the nanoformulation samples, it could be a promising material for future generations treatment of acute myocardial infarction and cardiac repair applications.
Collapse
Affiliation(s)
- Qinqin Sun
- Department of Outpatient Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, TaiyuanChina
- Department of Outpatient Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jia Yao
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhijun Zhang
- Department of Cardiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Juan Li
- Department of Outpatient Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, TaiyuanChina
- Department of Outpatient Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xue Zhang
- Department of Surgery Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Wang
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xufang Du
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Min Li
- Department of Breast Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P.R. China
- Department of Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ying Zhao
- Department of Outpatient Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, TaiyuanChina
- Department of Outpatient Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
18
|
Electrically conductive scaffolds mimicking the hierarchical structure of cardiac myofibers. Sci Rep 2023; 13:2863. [PMID: 36804588 PMCID: PMC9938142 DOI: 10.1038/s41598-023-29780-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Electrically conductive scaffolds, mimicking the unique directional alignment of muscle fibers in the myocardium, are fabricated using the 3D printing micro-stereolithography technique. Polyethylene glycol diacrylate (photo-sensitive polymer), Irgacure 819 (photo-initiator), curcumin (dye) and polyaniline (conductive polymer) are blended to make the conductive ink that is crosslinked using free radical photo-polymerization reaction. Curcumin acts as a liquid filter and prevents light from penetrating deep into the photo-sensitive solution and plays a central role in the 3D printing process. The obtained scaffolds demonstrate well defined morphology with an average pore size of 300 ± 15 μm and semi-conducting properties with a conductivity of ~ 10-6 S/m. Cyclic voltammetry analyses detect the electroactivity and highlight how the electron transfer also involve an ionic diffusion between the polymer and the electrolyte solution. Scaffolds reach their maximum swelling extent 30 min after immersing in the PBS at 37 °C and after 4 weeks they demonstrate a slow hydrolytic degradation rate typical of polyethylene glycol network. Conductive scaffolds display tunable conductivity and provide an optimal environment to the cultured mouse cardiac progenitor cells.
Collapse
|
19
|
Assad H, Assad A, Kumar A. Recent Developments in 3D Bio-Printing and Its Biomedical Applications. Pharmaceutics 2023; 15:255. [PMID: 36678884 PMCID: PMC9861443 DOI: 10.3390/pharmaceutics15010255] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The fast-developing field of 3D bio-printing has been extensively used to improve the usability and performance of scaffolds filled with cells. Over the last few decades, a variety of tissues and organs including skin, blood vessels, and hearts, etc., have all been produced in large quantities via 3D bio-printing. These tissues and organs are not only able to serve as building blocks for the ultimate goal of repair and regeneration, but they can also be utilized as in vitro models for pharmacokinetics, drug screening, and other purposes. To further 3D-printing uses in tissue engineering, research on novel, suitable biomaterials with quick cross-linking capabilities is a prerequisite. A wider variety of acceptable 3D-printed materials are still needed, as well as better printing resolution (particularly at the nanoscale range), speed, and biomaterial compatibility. The aim of this study is to provide expertise in the most prevalent and new biomaterials used in 3D bio-printing as well as an introduction to the associated approaches that are frequently considered by researchers. Furthermore, an effort has been made to convey the most pertinent implementations of 3D bio-printing processes, such as tissue regeneration, etc., by providing the most significant research together with a comprehensive list of material selection guidelines, constraints, and future prospects.
Collapse
Affiliation(s)
- Humira Assad
- Department of Chemistry, School of Chemical Engineering and Physical Sciences, Lovely Professional University, Punjab 144001, India
| | - Arvina Assad
- Bibi Halima College of Nursing and Medical Technology, Srinagar 190010, India
| | - Ashish Kumar
- Nalanda College of Engineering, Department of Science and Technology, Government of Bihar, Patna 803108, India
| |
Collapse
|
20
|
Das SL, Sutherland BP, Lejeune E, Eyckmans J, Chen CS. Mechanical response of cardiac microtissues to acute localized injury. Am J Physiol Heart Circ Physiol 2022; 323:H738-H748. [PMID: 36053751 DOI: 10.1152/ajpheart.00305.2022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
After a myocardial infarction (MI), the heart undergoes changes including local remodeling that can lead to regional abnormalities in mechanical and electrical properties, ultimately increasing the risk of arrythmias and heart failure. While these responses have been successfully recapitulated in animal models of MI, local changes in tissue and cell-level mechanics caused by MI remain difficult to study in vivo. Here, we developed an in vitro cardiac microtissue (CMT) injury system which through acute focal injury recapitulates aspects of the regional responses seen following an MI. Using a pulsed laser, cell death was induced in the center of the microtissue causing a loss of calcium signaling and a complete loss of contractile function in the injured region and resulting in a 39% reduction in the CMT's overall force production. After 7 days, the injured area remained void of CMs and showed increased expression of vimentin and fibronectin, two markers for fibrotic remodeling. Interestingly, while the injured region showed minimal recovery, calcium amplitudes in uninjured regions returned to levels comparable to control. Furthermore, overall force production returned to pre-injury levels despite the lack of contractile function in the injured region. Instead, uninjured regions exhibited elevated contractile function, compensating for the loss of function in the injured region, drawing parallels to changes in tissue-level mechanics seen in vivo. Overall, this work presents a new in vitro model to study cardiac tissue remodeling and electromechanical changes after injury.
Collapse
Affiliation(s)
- Shoshana L Das
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Bryan P Sutherland
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, United States
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| |
Collapse
|
21
|
Tetsuka H, Pirrami L, Wang T, Demarchi D, Shin SR. Wirelessly Powered 3D Printed Hierarchical Biohybrid Robots with Multiscale Mechanical Properties. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2202674. [PMID: 36313126 PMCID: PMC9603592 DOI: 10.1002/adfm.202202674] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The integration of flexible and stretchable electronics into biohybrid soft robotics can spur the development of new approaches to fabricate biohybrid soft machines, thus enabling a wide variety of innovative applications. Inspired by flexible and stretchable wireless-based bioelectronic devices, we have developed untethered biohybrid soft robots that can execute swimming motions, which are remotely controllable by the wireless transmission of electrical power into a cell simulator. To this end, wirelessly-powered, stretchable, and lightweight cell stimulators were designed to be integrated into muscle bodies without impeding the robots' underwater swimming abilities. The cell stimulators function by generating controlled monophasic pulses of up to ∼9 V in biological environments. By differentiating induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) directly on the cell stimulators using an accordion-inspired, three-dimensional (3D) printing construct, we have replicated the native myofiber architecture with comparable robustness and enhanced contractibility. Wirelessly modulated electrical frequencies enabled us to control the speed and direction of the biohybrid soft robots. A maximum locomotion speed of ∼580 μm/s was achieved in robots possessing a large body size by adjusting the pacing frequency. This innovative approach will provide a platform for building untethered and biohybrid systems for various biomedical applications.
Collapse
Affiliation(s)
- Hiroyuki Tetsuka
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, Massachusetts, 02139 USA
- Future Mobility Research Department, Toyota Research Institute of North America, Toyota Motor North America, 1555 Woodridge Avenue, Ann Arbor, Michigan, 48105 USA
| | - Lorenzo Pirrami
- iPrint Institute, HEIA-FR, HES-SO University of Applied Sciences and Arts Western Switzerland, Fribourg-1700, Switzerland
| | - Ting Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, Massachusetts, 02139 USA
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, Turin 10129, Italy
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Lansdowne Street, Cambridge, Massachusetts, 02139 USA
| |
Collapse
|
22
|
Murata K, Masumoto H. Systems for the functional evaluation of human heart tissues derived from pluripotent stem cells. Stem Cells 2022; 40:537-545. [PMID: 35303744 PMCID: PMC9216506 DOI: 10.1093/stmcls/sxac022] [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: 01/17/2022] [Accepted: 03/06/2022] [Indexed: 11/13/2022]
Abstract
Human pluripotent stem cells (hPSCs) are expected to be a promising cell source in regenerative medicine and drug discovery for the treatment of various intractable diseases. An approach for creating a three-dimensional (3D) structure from hPSCs that mimics human cardiac tissue functions has made it theoretically possible to conduct drug discovery and cardiotoxicity tests by assessing pharmacological responses in human cardiac tissues by a screening system using a compound library. The myocardium functions as a tissue composed of organized vascular networks, supporting stromal cells and cardiac muscle cells. Considering this, the reconstruction of tissue structure by various cells of cardiovascular lineages, such as vascular cells and cardiac muscle cells, is desirable for the ideal conformation of hPSC-derived cardiac tissues. Heart-on-a-chip, an organ-on-a-chip system to evaluate the physiological pump function of 3D cardiac tissues might hold promise in medical researches such as drug discovery and regenerative medicine. Here, we review various modalities to evaluate the function of human stem cell-derived cardiac tissues and introduce heart-on-a-chip systems that can recapitulate physiological parameters of hPSC-derived cardiac tissues.
Collapse
Affiliation(s)
- Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Bliley J, Tashman JW, Stang MA, Coffin BD, Shiwarksi DJ, Lee A, Hinton TJ, Feinberg AW. FRESH 3D bioprinting a contractile heart tube using human stem cell-derived cardiomyocytes. Biofabrication 2022; 14. [PMID: 35213846 DOI: 10.1088/1758-5090/ac58be] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/25/2022] [Indexed: 11/11/2022]
Abstract
Here we developed a simplified model of the human heart, similar that observed in embryonic development where the heart first starts as a contractile linear tube. To this end, we created a bioinspired model of the human heart tube scaled ~10x larger, consisting of a collagen tube fabricated with high fidelity using freeform reversible of embedding of suspended hydrogels (FRESH) 3D bioprinting. The collagen tubes were cellularized using human stem cell-derived cardiomyocytes and cardiac fibroblasts via a rapid casting approach, with synchronous contractions ~3-4 days after fabrication and maintained for up to one month. Immunofluorescent staining confirmed dense, interconnected networks of sarcomeric α-actinin-positive cardiomyocytes. Electrophysiology was assessed using calcium imaging and demonstrated anisotropic calcium wave propagation along the heart tube with a conduction velocity of ~5 cm/s. Contractility and basic pump function were demonstrated by tracking the movement of fluorescent beads within the lumen to estimate fluid displacement and bead velocity. Results show the ability to displace fluid, but the simple linear design and lack of valves limited mean bead displacement. In summary, we have 3D bioprinted a contractile human heart tube as an initial step toward organ engineering by mimicking the simplified structure observed at early developmental time points.
Collapse
Affiliation(s)
- Jacqueline Bliley
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, UNITED STATES
| | - Joshua W Tashman
- Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, UNITED STATES
| | - Maria A Stang
- Materials Science & Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213-3815, UNITED STATES
| | - Brian D Coffin
- Materials Science & Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213-3815, UNITED STATES
| | - Daniel J Shiwarksi
- Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, UNITED STATES
| | - Andrew Lee
- Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213-3815, UNITED STATES
| | - Thomas J Hinton
- Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213-3815, UNITED STATES
| | - Adam W Feinberg
- Biomedical Engineering, Materials Science & Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pittsburgh, Pennsylvania, 15213-3815, UNITED STATES
| |
Collapse
|
24
|
Wang B, Shah M, Williams LN, de Jongh Curry AL, Hong Y, Zhang G, Liao J. Acellular Myocardial Scaffolds and Slices Fabrication, and Method for Applying Mechanical and Electrical Simulation to Tissue Construct. Methods Mol Biol 2022; 2485:55-70. [PMID: 35618898 PMCID: PMC9811994 DOI: 10.1007/978-1-0716-2261-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiac tissue engineering/regeneration using decellularized myocardium has attracted great research attention due to its potential benefit to myocardial infarction (MI) treatment. Here, we described an optimal decellularization protocol to generate 3D porcine myocardial scaffolds with well-preserved cardiomyocyte lacunae, myocardial slices as a biomimetic cell culture and delivery platform, and a multi-stimulation bioreactor that is able to provide coordinated mechanical and electrical stimulations for facilitating cardiac construct development.
Collapse
Affiliation(s)
- Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mickey Shah
- Department of Biomedical Engineering, The University of Akron, Akron, OH, USA
| | - Lakiesha N Williams
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | | | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, OH, USA.
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA.
| |
Collapse
|
25
|
Jamee R, Araf Y, Naser IB, Promon SK. The promising rise of bioprinting in revolutionalizing medical science: Advances and possibilities. Regen Ther 2021; 18:133-145. [PMID: 34189195 PMCID: PMC8213915 DOI: 10.1016/j.reth.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Bioprinting is a relatively new yet evolving technique predominantly used in regenerative medicine and tissue engineering. 3D bioprinting techniques combine the advantages of creating Extracellular Matrix (ECM)like environments for cells and computer-aided tailoring of predetermined tissue shapes and structures. The essential application of bioprinting is for the regeneration or restoration of damaged and injured tissues by producing implantable tissues and organs. The capability of bioprinting is yet to be fully scrutinized in sectors like the patient-specific spatial distribution of cells, bio-robotics, etc. In this review, currently developed experimental systems and strategies for the bioprinting of different types of tissues as well as for drug delivery and cancer research are explored for potential applications. This review also digs into the most recent opportunities and future possibilities for the efficient implementation of bioprinting to restructure medical and technological practices.
Collapse
Affiliation(s)
- Radia Jamee
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Iftekhar Bin Naser
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Salman Khan Promon
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| |
Collapse
|
26
|
Wang L, Liu Y, Ye G, He Y, Li B, Guan Y, Gong B, Mequanint K, Xing MMQ, Qiu X. Injectable and conductive cardiac patches repair infarcted myocardium in rats and minipigs. Nat Biomed Eng 2021; 5:1157-1173. [PMID: 34593988 DOI: 10.1038/s41551-021-00796-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiac patches can help to restore the electrophysiological properties of the heart after myocardial infarction. However, scaffolds for the repair of heart muscle typically require surgical implantation or, if they are injectable, they are not electrically conductive or do not maintain their shape or function. Here, we report the performance, as demonstrated for the repair of infarcted heart muscle in rats and minipigs, of injectable and conductive scaffolds consisting of methacrylated elastin and gelatin, and carbon nanotubes that display shape-memory behaviour, a hierarchical porous structure and a negligible Poisson's ratio. In rats, the implantation of cell-free patches or patches seeded with rat cardiomyocytes onto the myocardium after ligation of the left anterior descending coronary artery led to functional repair after 4 weeks, as indicated by increases in fractional shortening and the ejection fraction, and by a decrease in the infarcted area. We also observed measures of functional recovery in minipigs with infarcted hearts after the delivery of cell-free patches or patches incorporating cardiomyocytes differentiated from human pluripotent stem cells.
Collapse
Affiliation(s)
- Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Yuqing Liu
- Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Genlan Ye
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Yutong He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Yezhi Guan
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Baoyong Gong
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario, Canada.,School of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Malcolm M Q Xing
- Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Ul Haq A, Carotenuto F, Di Nardo P, Francini R, Prosposito P, Pescosolido F, De Matteis F. Extrinsically Conductive Nanomaterials for Cardiac Tissue Engineering Applications. MICROMACHINES 2021; 12:914. [PMID: 34442536 PMCID: PMC8402139 DOI: 10.3390/mi12080914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/09/2023]
Abstract
Myocardial infarction (MI) is the consequence of coronary artery thrombosis resulting in ischemia and necrosis of the myocardium. As a result, billions of contractile cardiomyocytes are lost with poor innate regeneration capability. This degenerated tissue is replaced by collagen-rich fibrotic scar tissue as the usual body response to quickly repair the injury. The non-conductive nature of this tissue results in arrhythmias and asynchronous beating leading to total heart failure in the long run due to ventricular remodelling. Traditional pharmacological and assistive device approaches have failed to meet the utmost need for tissue regeneration to repair MI injuries. Engineered heart tissues (EHTs) seem promising alternatives, but their non-conductive nature could not resolve problems such as arrhythmias and asynchronous beating for long term in-vivo applications. The ability of nanotechnology to mimic the nano-bioarchitecture of the extracellular matrix and the potential of cardiac tissue engineering to engineer heart-like tissues makes it a unique combination to develop conductive constructs. Biomaterials blended with conductive nanomaterials could yield conductive constructs (referred to as extrinsically conductive). These cell-laden conductive constructs can alleviate cardiac functions when implanted in-vivo. A succinct review of the most promising applications of nanomaterials in cardiac tissue engineering to repair MI injuries is presented with a focus on extrinsically conductive nanomaterials.
Collapse
Affiliation(s)
- Arsalan Ul Haq
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Felicia Carotenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Paolo Di Nardo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- L.L. Levshin Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia
| | - Roberto Francini
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Paolo Prosposito
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Francesca Pescosolido
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Fabio De Matteis
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| |
Collapse
|
28
|
Roshanbinfar K, Esser TU, Engel FB. Stem Cells and Their Cardiac Derivatives for Cardiac Tissue Engineering and Regenerative Medicine. Antioxid Redox Signal 2021; 35:143-162. [PMID: 32993354 DOI: 10.1089/ars.2020.8193] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Heart failure is among the leading causes of morbidity worldwide with a 5-year mortality rate of ∼50%. Therefore, major efforts are invested to reduce heart damage upon injury or maintain and at best restore heart function. Recent Advances: In clinical trials, acellular constructs succeeded in improving cardiac function by stabilizing the infarcted heart. In addition, strategies utilizing stem-cell-derived cardiomyocytes have been developed to improve heart function postmyocardial infarction in small and large animal models. These strategies range from injection of cell-laden hydrogels to unstructured hydrogel-based and complex biofabricated cardiac patches. Importantly, novel methods have been developed to promote differentiation of stem-cell-derived cardiomyocytes to prevascularized cardiac patches. Critical Issues: Despite substantial progress in vascularization strategies for heart-on-the-chip technologies, little advance has been made in generating vascularized cardiac patches with clinically relevant dimensions. In addition, proper electrical coupling between engineered and host tissue to prevent and/or eliminate arrhythmia remains an unresolved issue. Finally, despite advanced approaches to include hierarchical structures in cardiac tissues, engineered tissues do not generate forces in the range of native adult cardiac tissue. Future Directions: It involves utilizing novel materials and advancing biofabrication strategies to generate prevascularized three-dimensional multicellular constructs of clinical relevant size; inclusion of hierarchical structures, electroconductive materials, and biologically active factors to enhance cardiomyocyte differentiation for optimized force generation and vascularization; optimization of bioreactor strategies for tissue maturation. Antioxid. Redox Signal. 35, 143-162.
Collapse
Affiliation(s)
- Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tilman U Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen, MURCE, Erlangen, Germany
| |
Collapse
|
29
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
30
|
Gähwiler EKN, Motta SE, Martin M, Nugraha B, Hoerstrup SP, Emmert MY. Human iPSCs and Genome Editing Technologies for Precision Cardiovascular Tissue Engineering. Front Cell Dev Biol 2021; 9:639699. [PMID: 34262897 PMCID: PMC8273765 DOI: 10.3389/fcell.2021.639699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) originate from the reprogramming of adult somatic cells using four Yamanaka transcription factors. Since their discovery, the stem cell (SC) field achieved significant milestones and opened several gateways in the area of disease modeling, drug discovery, and regenerative medicine. In parallel, the emergence of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) revolutionized the field of genome engineering, allowing the generation of genetically modified cell lines and achieving a precise genome recombination or random insertions/deletions, usefully translated for wider applications. Cardiovascular diseases represent a constantly increasing societal concern, with limited understanding of the underlying cellular and molecular mechanisms. The ability of iPSCs to differentiate into multiple cell types combined with CRISPR-Cas9 technology could enable the systematic investigation of pathophysiological mechanisms or drug screening for potential therapeutics. Furthermore, these technologies can provide a cellular platform for cardiovascular tissue engineering (TE) approaches by modulating the expression or inhibition of targeted proteins, thereby creating the possibility to engineer new cell lines and/or fine-tune biomimetic scaffolds. This review will focus on the application of iPSCs, CRISPR-Cas9, and a combination thereof to the field of cardiovascular TE. In particular, the clinical translatability of such technologies will be discussed ranging from disease modeling to drug screening and TE applications.
Collapse
Affiliation(s)
- Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Marcy Martin
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Bramasta Nugraha
- Molecular Parasitology Lab, Institute of Parasitology, University of Zurich, Zurich, Switzerland
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
31
|
Biotherapeutic-loaded injectable hydrogels as a synergistic strategy to support myocardial repair after myocardial infarction. J Control Release 2021; 335:216-236. [PMID: 34022323 DOI: 10.1016/j.jconrel.2021.05.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) has been considered as the leading cause of cardiovascular-related deaths worldwide. Although traditional therapeutic agents including various bioactive species such as growth factors, stem cells, and nucleic acids have demonstrated somewhat usefulness for the restoration of cardiac functions, the therapeutic efficiency remains unsatisfactory most likely due to the off-target-associated side effects and low localized retention of the used therapeutic agents in the infarcted myocardium, which constitutes a substantial barrier for the effective treatment of MI. Injectable hydrogels are regarded as a minimally invasive technology that can overcome the clinical and surgical limitations of traditional stenting by a modulated sol-gel transition and localized transport of a variety of encapsulated cargoes, leading to enhanced therapeutic efficiency and improved patient comfort and compliance. However, the design of injectable hydrogels for myocardial repair and the mechanism of action of bioactive substance-loaded hydrogels for MI repair remain unclear. To elucidate these points, we summarized the recent progresses made on the use of injectable hydrogels for encapsulation of various therapeutic substances for MI treatment with an emphasis on the mechanism of action of hydrogel systems for myocardial repair. Specifically, the pathogenesis of MI and the rational design of injectable hydrogels for myocardial repair were presented. Next, the mechanisms of various biotherapeutic substance-loaded injectable hydrogels for myocardial repair was discussed. Finally, the potential challenges and future prospects for the use of injectable hydrogels for MI treatment were proposed for the purpose of drawing theoretical guidance on the development of novel therapeutic strategies for efficient treatment of MI.
Collapse
|
32
|
Orsolits B, Kovács Z, Kriston-Vizi J, Merkely B, Földes G. New Modalities of 3D Pluripotent Stem Cell-Based Assays in Cardiovascular Toxicity. Front Pharmacol 2021; 12:603016. [PMID: 33854431 PMCID: PMC8039822 DOI: 10.3389/fphar.2021.603016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/04/2021] [Indexed: 12/04/2022] Open
Abstract
The substantial progress of the human induced pluripotent stem cell (hiPSC) technologies over the last decade has provided us with new opportunities for cardiovascular drug discovery, regenerative medicine, and disease modeling. The combination of hiPSC with 3D culture techniques offers numerous advantages for generating and studying physiological and pathophysiological cardiac models. Cells grown in 3D can overcome many limitations of 2D cell cultures and animal models. Furthermore, it enables the investigation in an architecturally appropriate, complex cellular environment in vitro. Yet, generation and study of cardiac organoids-which may contain versatile cardiovascular cell types differentiated from hiPSC-remain a challenge. The large-scale and high-throughput applications require accurate and standardised models with highly automated processes in culturing, imaging and data collection. Besides the compound spatial structure of organoids, their biological processes also possess different temporal dynamics which require other methods and technologies to detect them. In this review, we summarise the possibilities and challenges of acquiring relevant information from 3D cardiovascular models. We focus on the opportunities during different time-scale processes in dynamic pharmacological experiments and discuss the putative steps toward one-size-fits-all assays.
Collapse
Affiliation(s)
- Barbara Orsolits
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Zsófia Kovács
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - János Kriston-Vizi
- Bioinformatics Image Core (BIONIC), MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Gábor Földes
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
- National Heart and Lung Institute, Imperial Centre for Experimental and Translational Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
33
|
Induced Pluripotent Stem Cells (iPSCs) in Vascular Research: from Two- to Three-Dimensional Organoids. Stem Cell Rev Rep 2021; 17:1741-1753. [PMID: 33738695 PMCID: PMC7972819 DOI: 10.1007/s12015-021-10149-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2021] [Indexed: 01/19/2023]
Abstract
Stem cell technology has been around for almost 30 years and in that time has grown into an enormous field. The stem cell technique progressed from the first successful isolation of mammalian embryonic stem cells (ESCs) in the 1990s, to the production of human induced-pluripotent stem cells (iPSCs) in the early 2000s, to finally culminate in the differentiation of pluripotent cells into highly specialized cell types, such as neurons, endothelial cells (ECs), cardiomyocytes, fibroblasts, and lung and intestinal cells, in the last decades. In recent times, we have attained a new height in stem cell research whereby we can produce 3D organoids derived from stem cells that more accurately mimic the in vivo environment. This review summarizes the development of stem cell research in the context of vascular research ranging from differentiation techniques of ECs and smooth muscle cells (SMCs) to the generation of vascularized 3D organoids. Furthermore, the different techniques are critically reviewed, and future applications of current 3D models are reported.
Collapse
|
34
|
Hnatiuk AP, Briganti F, Staudt DW, Mercola M. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 2021; 28:271-282. [PMID: 33740432 PMCID: PMC8054828 DOI: 10.1016/j.chembiol.2021.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/26/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have emerged as a promising platform for pharmacogenomics and drug development. In cardiology, they make it possible to produce unlimited numbers of patient-specific human cells that reproduce hallmark features of heart disease in the culture dish. Their potential applications include the discovery of mechanism-specific therapeutics, the evaluation of safety and efficacy in a human context before a drug candidate reaches patients, and the stratification of patients for clinical trials. Although this new technology has the potential to revolutionize drug discovery, translational hurdles have hindered its widespread adoption for pharmaceutical development. Here we discuss recent progress in overcoming these hurdles that should facilitate the use of hiPSCs to develop new medicines and individualize therapies for heart disease.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Francesca Briganti
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David W Staudt
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
Banerjee S, Szepes M, Dibbert N, Rios-Camacho JC, Kirschning A, Gruh I, Dräger G. Dextran-based scaffolds for in-situ hydrogelation: Use for next generation of bioartificial cardiac tissues. Carbohydr Polym 2021; 262:117924. [PMID: 33838803 DOI: 10.1016/j.carbpol.2021.117924] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/13/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
In pursuit of a chemically-defined matrix for in vitro cardiac tissue generation, we present dextran (Dex)-derived hydrogels as matrices suitable for bioartificial cardiac tissues (BCT). The dextran hydrogels were generated in situ by using hydrazone formation as the crosslinking reaction. Material properties were flexibly adjusted, by varying the degrees of derivatization and the molecular weight of dextran used. Furthermore, to modulate dextran's bioactivity, cyclic pentapeptide RGD was coupled to its backbone. BCTs were generated by using a blend of modified dextran and human collagen (hColI) in combination with induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and fibroblasts. These hColI + Dex blends with or without RGD supported tissue formation and functional maturation of CMs. Contraction forces (hColI + Dex-RGD: 0.27 ± 0.02 mN; hColI + Dex: 0.26 ± 0.01 mN) and frequencies were comparable to published constructs. Thus, we could demonstrate that, independent of the presence of RGD, our covalently linked dextran hydrogels are a promising matrix for building cardiac grafts.
Collapse
Affiliation(s)
- Samhita Banerjee
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Monika Szepes
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Nick Dibbert
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Julio-Cesar Rios-Camacho
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Andreas Kirschning
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Ina Gruh
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Gerald Dräger
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany.
| |
Collapse
|
36
|
Cacciapuoti M, Johnson B, Kapdia A, Powell S, Gallicano GI. The Role of Neuregulin and Stem Cells as Therapy Post-Myocardial Infarction. Stem Cells Dev 2020; 29:1266-1274. [PMID: 32731805 DOI: 10.1089/scd.2020.0099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease, including myocardial infarction (MI), is a leading cause of morbidity and mortality in the United States. Due to the limited self-renewal capacity of cardiac tissue, MIs can lead to progressive heart disease with a lasting impact on health and quality of life. The recent discovery of cardiac stem cells has incited research into their potential therapeutic applications for patients suffering from cardiovascular disease. Studies have demonstrated the ability of stem cells to both generate cardiac tissues in vitro and aid in the recovery of cardiovascular function in vivo in animal models. However, the long-term efficacy of stem cells as regenerative therapy is still unknown. Exploration of alternative therapies is underway, including the use of cardiac growth factor neuregulin-1 (NRG-1). Research has demonstrated that NRG-1 not only has direct effects on cardiomyocytes (CM) but also acts within the tissues supporting the CM. Transplantation of NRG-1 into ischemic cardiac tissue mitigates the progression of heart failure and can reverse cardiac remodeling. Recent publications have sought to study the combined use of these agents, and while the results are promising, they do warrant further research. This review aims to consider these therapies separately as well as in combination.
Collapse
Affiliation(s)
- Maria Cacciapuoti
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Bria Johnson
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Anjani Kapdia
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Sarah Powell
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - G Ian Gallicano
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
37
|
Yong U, Lee S, Jung S, Jang J. Interdisciplinary approaches to advanced cardiovascular tissue engineering: ECM-based biomaterials, 3D bioprinting, and its assessment. ACTA ACUST UNITED AC 2020. [DOI: 10.1088/2516-1091/abb211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Tsukamoto Y, Akagi T, Akashi M. Supersensitive Layer-by-Layer 3D Cardiac Tissues Fabricated on a Collagen Culture Vessel Using Human-Induced Pluripotent Stem Cells. Tissue Eng Part C Methods 2020; 26:493-502. [PMID: 32873187 DOI: 10.1089/ten.tec.2020.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The fabrication of artificial cardiac tissue is an active area of research due to the shortage of donors for heart transplantation and for drug development. In our previous study, we fabricated vascularized three-dimensional (3D) cardiac tissue by layer-by-layer (LbL) and cell accumulation technique. However, it was not able to develop sufficient function because it was cultured on a hard plastic substrate. Experiment: Herein, we report the fabrication of high-performance 3D cardiac tissue by LbL and cell accumulation technique using a collagen culture vessel. Results: By using a collagen culture vessel, 3D cardiac tissue could be fabricated on a collagen culture vessel and this tissue showed high functionality due to improved interaction with the vessel. In the case of the plastic culture insert, 3D cardiac tissue was found to be peeled off, but this did not occur on the collagen culture vessel. In addition, the 3D cardiac tissue fabricated on a collagen culture vessel showed contraction that was 20 times larger than the tissue fabricated on a plastic culture insert. As a result of evaluation of cardiotoxicity using E-4031, the sensitivity of arrhythmia detection was increased by using collagen culture vessel. Conclusions: These results are expected to contribute to transplantation and drug discovery research as a 3D cardiac tissue model with a function similar to that of the living heart.
Collapse
Affiliation(s)
- Yoshinari Tsukamoto
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Takami Akagi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Mitsuru Akashi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
39
|
Shrestha S, McFadden MJ, Gramolini AO, Santerre JP. Proteome analysis of secretions from human monocyte-derived macrophages post-exposure to biomaterials and the effect of secretions on cardiac fibroblast fibrotic character. Acta Biomater 2020; 111:80-90. [PMID: 32428683 DOI: 10.1016/j.actbio.2020.04.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/31/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022]
Abstract
The use of exogenous biomolecules (BM) for the purpose of repairing and regenerating damaged cardiac tissue can yield serious side effects if used for prolonged periods. As well, such strategies can be cost prohibitive depending on the regiment and period of time applied. Alternatively, autologous monocytes/monocyte-derived macrophages (MDM) can provide a viable path towards generating an endogenous source of stimulatory BM. Biomaterials are often considered as delivery vehicles to generate unique profiles of such BM in tissues or to deliver autologous cells, that can influence the nature of BM produced by the cells. MDM cultured on a degradable polar hydrophobic ionic (D-PHI) polyurethane has previously demonstrated a propensity to increase select anti-inflammatory cytokines, and therefore there is good rationale to further investigate a broader spectrum of the cells' BM in order to provide a more complete proteomic analysis of human MDM secretions induced by D-PHI. Further, it is of interest to assess the potential of such BM to influence cells involved in the reparative state of vital tissues such as those that affect cardiac cell function. Hence, this current study examines the proteomic profile of MDM secretions using mass spectrometry for the first time, along with ELISA, following their culture on D-PHI, and compares them to two important reference materials, poly(lactic-co-glycolic acid) (PLGA) and tissue culture polystyrene (TCPS). Secretions collected from D-PHI cultured MDM led to higher levels of regenerative BM, AGRN, TGFBI and ANXA5, but lower levels of pro-fibrotic BM, MMP7, IL-1β, IL-6 and TNFα, when compared to MDM secretions collected from PLGA and TCPS. In the application to cardiac cell function, the secretion collected from D-PHI cultured MDM led to more human cardiac fibroblast (HCFs) migration. A lower collagen gel contraction induced by MDM secretions collected from D-PHI was supported by gene array analysis for human fibrosis-related genes. The implication of these findings is that more tailored biomaterials such as D-PHI, may lead to a lower pro-inflammatory phenotype of macrophages when used in cardiac tissue constructs, thereby enabling the development of vehicles for the delivery of interventional therapies, or be applied as coatings for sensor implants in cardiac tissue that minimize fibrosis. The general approach of using synthetic biomaterials in order to induce MDM secretions in a manner that will guide favorable regeneration will be critical in making the choice of biomaterials for tissue regeneration work in the future. STATEMENT OF SIGNIFICANCE: Immune modulation strategies currently applied in cardiac tissue repair are mainly based on the delivery of defined exogenous biomolecules. However, the use of such biomolecules may pose wide ranging systemic effects, thereby rendering them clinically less practical. The chemistry of biomaterials (used as a potential targeted delivery modality to circumvent the broad systemic effects of biomolecules) can not only affect acute and chronic toxicity but also alters the timeframe of the wound healing cascade. In this context, monocytes/monocyte-derive macrophages (MDM) can be harnessed as an immune modulating strategy to promote wound healing by an appropriate choice of the biomaterial. However, there are limited reports on the complete proteome analysis of MDM and their reaction of biomaterial related interventions on cardiac tissues and cells. No studies to date have demonstrated the complete proteome of MDM secretions when these cells were cultured on a non-traditional immune modulatory ionomeric polyurethane D-PHI film. This study demonstrated that MDM cultured on D-PHI expressed significantly higher levels of AGRN, TGFBI and ANXA5 but lower levels of MMP7, IL-1β, IL-6 and TNFα when compared to MDM cultured on a well-established degradable biomaterials in the medical field, e.g. PLGA and TCPS, which are often used as the relative standards for cell culture work in the biomaterials field. The implications of these findings have relevance to the repair of cardiac tissues. In another aspect of the work, human cardiac fibroblasts showed significantly lower contractility (low collagen gel contraction and low levels of ACTA2) when cultured in the presence of MDM secretions collected after culturing them on D-PHI compared to PLGA and TCPS. The findings place emphasis on the importance of making the choice of biomaterials for tissue engineering and regenerative medicine applied to their use in cardiac tissue repair.
Collapse
Affiliation(s)
- Suja Shrestha
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada
| | - Meghan J McFadden
- Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1M8, Canada
| | - J Paul Santerre
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada.
| |
Collapse
|
40
|
Obal D, Wu S, McKinstry-Wu A, Tawfik VL. A Guide to Understanding "State-of-the-Art" Basic Research Techniques in Anesthesiology. Anesth Analg 2020; 131:450-463. [PMID: 32371742 DOI: 10.1213/ane.0000000000004801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Perioperative medicine is changing from a "protocol-based" approach to a progressively personalized care model. New molecular techniques and comprehensive perioperative medical records allow for detection of patient-specific phenotypes that may better explain, or even predict, a patient's response to perioperative stress and anesthetic care. Basic science technology has significantly evolved in recent years with the advent of powerful approaches that have translational relevance. It is incumbent on us as a primarily clinical specialty to have an in-depth understanding of rapidly evolving underlying basic science techniques to incorporate such approaches into our own research, critically interpret the literature, and improve future anesthesia patient care. This review focuses on 3 important and most likely practice-changing basic science techniques: next-generation sequencing (NGS), clustered regularly interspaced short palindromic repeat (CRISPR) modulations, and inducible pluripotent stem cells (iPSCs). Each technique will be described, potential advantages and limitations discussed, open questions and challenges addressed, and future developments outlined. We hope to provide insight for practicing physicians when confronted with basic science articles and encourage investigators to apply "state-of-the-art" technology to their future experiments.
Collapse
Affiliation(s)
- Detlef Obal
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California.,Department of Anesthesiology, Perioperative, and Pain Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
| | - Shaogen Wu
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Andrew McKinstry-Wu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vivianne L Tawfik
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
41
|
Abstract
The spectrum of ischemic heart diseases, encompassing acute myocardial infarction to heart failure, represents the leading cause of death worldwide. Although extensive progress in cardiovascular diagnoses and therapy has been made, the prevalence of the disease continues to increase. Cardiac regeneration has a promising perspective for the therapy of heart failure. Recently, extracellular matrix (ECM) has been shown to play an important role in cardiac regeneration and repair after cardiac injury. There is also evidence that the ECM could be directly used as a drug to promote cardiomyocyte proliferation and cardiac regeneration. Increasing evidence supports that applying ECM biomaterials to maintain heart function recovery is an important approach to apply the concept of cardiac regenerative medicine to clinical practice in the future. Here, we will introduce the essential role of cardiac ECM in cardiac regeneration and summarize the approaches of delivering ECM biomaterials to promote cardiac repair in this review.
Collapse
Affiliation(s)
- Haotong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Minghui Bao
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
42
|
|
43
|
Appiah C, Arndt C, Siemsen K, Heitmann A, Staubitz A, Selhuber-Unkel C. Living Materials Herald a New Era in Soft Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807747. [PMID: 31267628 DOI: 10.1002/adma.201807747] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/07/2019] [Indexed: 05/22/2023]
Abstract
Living beings have an unsurpassed range of ways to manipulate objects and interact with them. They can make autonomous decisions and can heal themselves. So far, a conventional robot cannot mimic this complexity even remotely. Classical robots are often used to help with lifting and gripping and thus to alleviate the effects of menial tasks. Sensors can render robots responsive, and artificial intelligence aims at enabling autonomous responses. Inanimate soft robots are a step in this direction, but it will only be in combination with living systems that full complexity will be achievable. The field of biohybrid soft robotics provides entirely new concepts to address current challenges, for example the ability to self-heal, enable a soft touch, or to show situational versatility. Therefore, "living materials" are at the heart of this review. Similarly to biological taxonomy, there is a recent effort for taxonomy of biohybrid soft robotics. Here, an expansion is proposed to take into account not only function and origin of biohybrid soft robotic components, but also the materials. This materials taxonomy key demonstrates visually that materials science will drive the development of the field of soft biohybrid robotics.
Collapse
Affiliation(s)
- Clement Appiah
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Christine Arndt
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Katharina Siemsen
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Anne Heitmann
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Anne Staubitz
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
- Otto-Diels-Institute for Organic Chemistry, University of Kiel, Otto-Hahn-Platz 4, D-24118, Kiel, Germany
| | | |
Collapse
|
44
|
Micro Vacuum Chuck and Tensile Test System for Bio-Mechanical Evaluation of 3D Tissue Constructed of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPS-CM). MICROMACHINES 2019; 10:mi10070487. [PMID: 31331014 PMCID: PMC6680730 DOI: 10.3390/mi10070487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
In this report, we propose a micro vacuum chuck (MVC) which can connect three-dimensional (3D) tissues to a tensile test system by vacuum pressure. Because the MVC fixes the 3D tissue by vacuum pressure generated on multiple vacuum holes, it is expected that the MVC can fix 3D tissue to the system easily and mitigate the damage which can happen by handling during fixing. In order to decide optimum conditions for the size of the vacuum holes and the vacuum pressure, various sized vacuum holes and vacuum pressures were applied to a normal human cardiac fibroblast 3D tissue. From the results, we confirmed that a square shape with 100 µm sides was better for fixing the 3D tissue. Then we mounted our developed MVCs on a specially developed tensile test system and measured the bio-mechanical property (beating force) of cardiac 3D tissue which was constructed of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM); the 3D tissue had been assembled by the layer-by-layer (LbL) method. We measured the beating force of the cardiac 3D tissue and confirmed the measured force followed the Frank-Starling relationship. This indicates that the beating property of cardiac 3D tissue obtained by the LbL method was close to that of native cardiac tissue.
Collapse
|
45
|
Pathak S, Regmi S, Shrestha P, Choi I, Doh KO, Jeong JH. Mesenchymal Stem Cell Capping on ECM-Anchored Caspase Inhibitor-Loaded PLGA Microspheres for Intraperitoneal Injection in DSS-Induced Murine Colitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901269. [PMID: 31018047 DOI: 10.1002/smll.201901269] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/07/2019] [Indexed: 06/09/2023]
Abstract
Mesenchymal stem cells (MSCs) are considered as a promising alternative for the treatment of various inflammatory disorders. However, poor viability and engraftment of MSCs after transplantation are major hurdles in mesenchymal stem cell therapy. Extracellular matrix (ECM)-coated scaffolds provide better cell attachment and mechanical support for MSCs after transplantation. A single-step method for ECM functionalization on poly(lactic-co-glycolic acid) (PLGA) microspheres using a novel compound, dopamine-conjugated poly(ethylene-alt-maleic acid), as a stabilizer during the preparation of microspheres is reported. The dopamine molecules on the surface of microspheres provide active sites for the conjugation of ECM in an aqueous solution. The results reveal that the viability of MSCs improves when they are coated over the ECM-functionalized PLGA microspheres (eMs). In addition, the incorporation of a broad-spectrum caspase inhibitor (IDN6556) into the eMs synergistically increases the viability of MSCs under in vitro conditions. Intraperitoneal injection of the MSC-microsphere hybrid alleviates experimental colitis in a murine model via inhibiting Th1 and Th17 differentiation of CD4+ T cells in colon-draining mesenteric lymph nodes. Therefore, drug-loaded ECM-coated surfaces may be considered as attractive tools for improving viability, proliferation, and functionality of MSCs following transplantation.
Collapse
Affiliation(s)
- Shiva Pathak
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Shobha Regmi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Prakash Shrestha
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Kyoung-Oh Doh
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, 42415, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| |
Collapse
|
46
|
Edalat SG, Jang Y, Kim J, Park Y. Collagen Type I Containing Hybrid Hydrogel Enhances Cardiomyocyte Maturation in a 3D Cardiac Model. Polymers (Basel) 2019; 11:polym11040687. [PMID: 30995718 PMCID: PMC6523216 DOI: 10.3390/polym11040687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/27/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
In vitro maturation of cardiomyocytes in 3D is essential for the development of viable cardiac models for therapeutic and developmental studies. The method by which cardiomyocytes undergoes maturation has significant implications for understanding cardiomyocytes biology. The regulation of the extracellular matrix (ECM) by changing the composition and stiffness is quintessential for engineering a suitable environment for cardiomyocytes maturation. In this paper, we demonstrate that collagen type I, a component of the ECM, plays a crucial role in the maturation of cardiomyocytes. To this end, embryonic stem-cell derived cardiomyocytes were incorporated into Matrigel-based hydrogels with varying collagen type I concentrations of 0 mg, 3 mg, and 6 mg. Each hydrogel was analyzed by measuring the degree of stiffness, the expression levels of MLC2v, TBX18, and pre-miR-21, and the size of the hydrogels. It was shown that among the hydrogel variants, the Matrigel-based hydrogel with 3 mg of collagen type I facilitates cardiomyocyte maturation by increasing MLC2v expression. The treatment of transforming growth factor β1 (TGF-β1) or fibroblast growth factor 4 (FGF-4) on the hydrogels further enhanced the MLC2v expression and thereby cardiomyocyte maturation.
Collapse
Affiliation(s)
- Sam G Edalat
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.
| | - Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.
| | - Jongseong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.
| |
Collapse
|
47
|
Abstract
Bio-actuators that use insect muscular tissue have attracted attention from researchers worldwide because of their small size, self-motive property, self-repairer ability, robustness, and the need for less environment management than mammalian cells. To demonstrate the potential of insect muscular tissue for use as bio-actuators, three types of these robots, a pillar actuator, a walker, and a twizzer, have been designed and fabricated. However, a model of an insect muscular tissue-powered swimming robot that is able to float and swim in a solution has not yet been reported. Therefore, in this paper, we present a prototype of an insect muscular tissue-powered autonomous micro swimming robot that operates at room temperature and requires no temperature and pH maintenance. To design a practical robot body that is capable of swimming by using the force of the insect dorsal vessel (DV), we first measured the contraction force of the DV. Then, the body of the swimming robot was designed, and the design was confirmed by a simulation that used the condition of measured contraction force. After that, we fabricated the robot body using polydimethylpolysiloxane (PDMS). The PDMS body was obtained from a mold that was fabricated by a stereo lithography method. Finally, we carefully attached the DV to the PDMS body to complete the assembly of the swimming robot. As a result, we confirmed the micro swimming robot swam autonomously at an average velocity of 11.7 μm/s using spontaneous contractions of the complete insect DV tissue. These results demonstrated that the insect DV has potential for use as a bio-actuator for floating and swimming in solution.
Collapse
|
48
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
49
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
50
|
Kamm RD, Bashir R, Arora N, Dar RD, Gillette MU, Griffith LG, Kemp ML, Kinlaw K, Levin M, Martin AC, McDevitt TC, Nerem RM, Powers MJ, Saif TA, Sharpe J, Takayama S, Takeuchi S, Weiss R, Ye K, Yevick HG, Zaman MH. Perspective: The promise of multi-cellular engineered living systems. APL Bioeng 2018; 2:040901. [PMID: 31069321 PMCID: PMC6481725 DOI: 10.1063/1.5038337] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
Recent technological breakthroughs in our ability to derive and differentiate induced pluripotent stem cells, organoid biology, organ-on-chip assays, and 3-D bioprinting have all contributed to a heightened interest in the design, assembly, and manufacture of living systems with a broad range of potential uses. This white paper summarizes the state of the emerging field of "multi-cellular engineered living systems," which are composed of interacting cell populations. Recent accomplishments are described, focusing on current and potential applications, as well as barriers to future advances, and the outlook for longer term benefits and potential ethical issues that need to be considered.
Collapse
Affiliation(s)
- Roger D. Kamm
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Rashid Bashir
- University of Illinois at Urbana-Champaign, Urbana, Illinois 61820, USA
| | - Natasha Arora
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Roy D. Dar
- University of Illinois at Urbana-Champaign, Urbana, Illinois 61820, USA
| | | | - Linda G. Griffith
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Melissa L. Kemp
- Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | - Adam C. Martin
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | | | - Robert M. Nerem
- Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Mark J. Powers
- Thermo Fisher Scientific, Frederick, Maryland 21704, USA
| | - Taher A. Saif
- University of Illinois at Urbana-Champaign, Urbana, Illinois 61820, USA
| | - James Sharpe
- EMBL Barcelona, European Molecular Biology Laboratory, Barcelona 08003, Spain
| | | | | | - Ron Weiss
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Kaiming Ye
- Binghamton University, Binghamton, New York 13902, USA
| | - Hannah G. Yevick
- Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | | |
Collapse
|