1
|
Cofiño-Fabres C, Boonen T, Rivera-Arbeláez JM, Rijpkema M, Blauw L, Rensen PCN, Schwach V, Ribeiro MC, Passier R. Micro-Engineered Heart Tissues On-Chip with Heterotypic Cell Composition Display Self-Organization and Improved Cardiac Function. Adv Healthc Mater 2024; 13:e2303664. [PMID: 38471185 DOI: 10.1002/adhm.202303664] [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: 10/26/2023] [Revised: 01/30/2024] [Indexed: 03/14/2024]
Abstract
Advanced in vitro models that recapitulate the structural organization and function of the human heart are highly needed for accurate disease modeling, more predictable drug screening, and safety pharmacology. Conventional 3D Engineered Heart Tissues (EHTs) lack heterotypic cell complexity and culture under flow, whereas microfluidic Heart-on-Chip (HoC) models in general lack the 3D configuration and accurate contractile readouts. In this study, an innovative and user-friendly HoC model is developed to overcome these limitations, by culturing human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs), endothelial (ECs)- and smooth muscle cells (SMCs), together with human cardiac fibroblasts (FBs), underflow, leading to self-organized miniaturized micro-EHTs (µEHTs) with a CM-EC interface reminiscent of the physiological capillary lining. µEHTs cultured under flow display enhanced contractile performance and conduction velocity. In addition, the presence of the EC layer altered drug responses in µEHT contraction. This observation suggests a potential barrier-like function of ECs, which may affect the availability of drugs to the CMs. These cardiac models with increased physiological complexity, will pave the way to screen for therapeutic targets and predict drug efficacy.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Tom Boonen
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - José M Rivera-Arbeláez
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- BIOS Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Minke Rijpkema
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Lisanne Blauw
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Marcelo C Ribeiro
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, 2300 RC, The Netherlands
| |
Collapse
|
2
|
Sanchez Armengol E, Hock N, Saribal S, To D, Summonte S, Veider F, Kali G, Bernkop-Schnürch A, Laffleur F. Unveiling the potential of biomaterials and their synergistic fusion in tissue engineering. Eur J Pharm Sci 2024; 196:106761. [PMID: 38580169 DOI: 10.1016/j.ejps.2024.106761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/17/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Inspired by nature, tissue engineering aims to employ intricate mechanisms for advanced clinical interventions, unlocking inherent biological potential and propelling medical breakthroughs. Therefore, medical, and pharmaceutical fields are growing interest in tissue and organ replacement, repair, and regeneration by this technology. Three primary mechanisms are currently used in tissue engineering: transplantation of cells (I), injection of growth factors (II) and cellular seeding in scaffolds (III). However, to develop scaffolds presenting highest potential, reinforcement with polymeric materials is growing interest. For instance, natural and synthetic polymers can be used. Regardless, chitosan and keratin are two biopolymers presenting great biocompatibility, biodegradability and non-antigenic properties for tissue engineering purposes offering restoration and revitalization. Therefore, combination of chitosan and keratin has been studied and results exhibit highly porous scaffolds providing optimal environment for tissue cultivation. This review aims to give an historical as well as current overview of tissue engineering, presenting mechanisms used and polymers involved in the field.
Collapse
Affiliation(s)
- Eva Sanchez Armengol
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Nathalie Hock
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria; ITM Isotope Technologies Munich SE, Walther-von-Dyck Str. 4, 85748, Garching bei Munich, Germany
| | - Sila Saribal
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Dennis To
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Simona Summonte
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria; ThioMatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - Florina Veider
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria; Sandoz, Biochemiestraße 10, 6250, Kundl, Austria
| | - Gergely Kali
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Flavia Laffleur
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
3
|
Schuftan D, Kooh YKG, Guo J, Sun Y, Aryan L, Stottlemire B, Berkland C, Genin GM, Huebsch N. Dynamic control of contractile resistance to iPSC-derived micro-heart muscle arrays. J Biomed Mater Res A 2024; 112:534-548. [PMID: 37952251 PMCID: PMC10922390 DOI: 10.1002/jbm.a.37642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Many types of cardiovascular disease are linked to the mechanical forces placed on the heart. However, our understanding of how mechanical forces exactly affect the cellular biology of the heart remains incomplete. In vitro models based on cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CM) enable researchers to develop medium to high-throughput systems to study cardiac mechanobiology at the cellular level. Previous models have been developed to enable the study of mechanical forces, such as cardiac afterload. However, most of these models require exogenous extracellular matrix (ECM) to form cardiac tissues. Recently, a system was developed to simulate changes in afterload by grafting ECM-free micro-heart muscle arrays to elastomeric substrates of discrete stiffnesses. In the present study, we extended this system by combining the elastomer-grafted tissue arrays with a magnetorheological elastomeric substrate. This system allows iPSC-CM based micro-heart muscle arrays to experience dynamic changes in contractile resistance to mimic dynamically altered afterload. Acute changes in substrate stiffness led to acute changes in the calcium dynamics and contractile forces, illustrating the system's ability to dynamically elicit changes in tissue mechanics by dynamically changing contractile resistance.
Collapse
Affiliation(s)
- David Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yasaman Kargar Gaz Kooh
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingxuan Guo
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yuwen Sun
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryce Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Guy M. Genin
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Das A, Nikhil A, Shiekh PA, Yadav B, Jagavelu K, Kumar A. Ameliorating impaired cardiac function in myocardial infarction using exosome-loaded gallic-acid-containing polyurethane scaffolds. Bioact Mater 2024; 33:324-340. [PMID: 38076649 PMCID: PMC10701288 DOI: 10.1016/j.bioactmat.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) can be tackled by implanting cardiac patches which provide mechanical support to the heart. However, most tissue-engineered scaffolds face difficulty in attenuating oxidative stress, maintaining mechanical stability, and regenerating damaged cardiomyocytes. Here, we fabricated elastic cryogels using polyurethane modified with antioxidant gallic acid in its backbone (PUGA) and further coated them with decellularized extracellular matrix (dECM) to improve adhesiveness, biocompatibility and hemocompatibility. The scaffold was functionalized with exosomes (EXO) isolated from adipose-derived stem cells having regenerative potential. PUGA-dECM + EXO was tested in a rat model with induced MI where echocardiography after 8 weeks of implantation showed significant recovery in treatment group. Histological analysis revealed a decrease in fibrosis after application of patch and promotion of angiogenesis with reduced oxidative stress was shown by immunostaining. Expression of cardiac tissue contractile function marker was also observed in treatment groups. Thus, the proposed biomaterial has a promising application to be utilized as a patch for cardiac regeneration. More detailed studies with larger animal species are needed for using these observations for specific applications.
Collapse
Affiliation(s)
- Ankita Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Aman Nikhil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Babita Yadav
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Kumaravelu Jagavelu
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre of Excellence for Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| |
Collapse
|
5
|
Baena-Montes JM, Kraśny MJ, O’Halloran M, Dunne E, Quinlan LR. In Vitro Models for Improved Therapeutic Interventions in Atrial Fibrillation. J Pers Med 2023; 13:1237. [PMID: 37623487 PMCID: PMC10455620 DOI: 10.3390/jpm13081237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Atrial fibrillation is the most common type of cardiac arrhythmias in humans, mostly caused by hyper excitation of specific areas in the atrium resulting in dyssynchronous atrial contractions, leading to severe consequences such as heart failure and stroke. Current therapeutics aim to target this condition through both pharmacological and non-pharmacological approaches. To test and validate any of these treatments, an appropriate preclinical model must be carefully chosen to refine and optimise the therapy features to correctly reverse this condition. A broad range of preclinical models have been developed over the years, with specific features and advantages to closely mimic the pathophysiology of atrial fibrillation. In this review, currently available models are described, from traditional animal models and in vitro cell cultures to state-of-the-art organoids and organs-on-a-chip. The advantages, applications and limitations of each model are discussed, providing the information to select the appropriate model for each research application.
Collapse
Affiliation(s)
- Jara M. Baena-Montes
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
| | - Marcin J. Kraśny
- Smart Sensors Lab, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Martin O’Halloran
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Electrical & Electronic Engineering, School of Engineering, University of Galway, H91 TK33 Galway, Ireland
| | - Eoghan Dunne
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R. Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
- CÚRAM SFI Centre for Research in Medical Devices, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
6
|
Dwyer KD, Kant RJ, Soepriatna AH, Roser SM, Daley MC, Sabe SA, Xu CM, Choi BR, Sellke FW, Coulombe KLK. One Billion hiPSC-Cardiomyocytes: Upscaling Engineered Cardiac Tissues to Create High Cell Density Therapies for Clinical Translation in Heart Regeneration. Bioengineering (Basel) 2023; 10:587. [PMID: 37237658 PMCID: PMC10215511 DOI: 10.3390/bioengineering10050587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Despite the overwhelming use of cellularized therapeutics in cardiac regenerative engineering, approaches to biomanufacture engineered cardiac tissues (ECTs) at clinical scale remain limited. This study aims to evaluate the impact of critical biomanufacturing decisions-namely cell dose, hydrogel composition, and size-on ECT formation and function-through the lens of clinical translation. ECTs were fabricated by mixing human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) and human cardiac fibroblasts into a collagen hydrogel to engineer meso-(3 × 9 mm), macro- (8 × 12 mm), and mega-ECTs (65 × 75 mm). Meso-ECTs exhibited a hiPSC-CM dose-dependent response in structure and mechanics, with high-density ECTs displaying reduced elastic modulus, collagen organization, prestrain development, and active stress generation. Scaling up, cell-dense macro-ECTs were able to follow point stimulation pacing without arrhythmogenesis. Finally, we successfully fabricated a mega-ECT at clinical scale containing 1 billion hiPSC-CMs for implantation in a swine model of chronic myocardial ischemia to demonstrate the technical feasibility of biomanufacturing, surgical implantation, and engraftment. Through this iterative process, we define the impact of manufacturing variables on ECT formation and function as well as identify challenges that must still be overcome to successfully accelerate ECT clinical translation.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
| | - Rajeev J. Kant
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
| | - Arvin H. Soepriatna
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
| | - Stephanie M. Roser
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
| | - Mark C. Daley
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
| | - Sharif A. Sabe
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Cynthia M. Xu
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Frank W. Sellke
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Kareen L. K. Coulombe
- School of Engineering, Brown University Center for Biomedical Engineering, Providence, RI 02912, USA; (K.D.D.)
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
7
|
Mandal K, Sangabathuni S, Haghniaz R, Kawakita S, Mecwan M, Nakayama A, Zhang X, Edalati M, Huang W, Lopez Hernandez A, Jucaud V, Dokmeci MR, Khademhosseini A. Oxygen-generating microparticles downregulate HIF-1α expression, increase cardiac contractility, and mitigate ischemic injury. Acta Biomater 2023; 159:211-225. [PMID: 36669549 PMCID: PMC9992239 DOI: 10.1016/j.actbio.2023.01.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/20/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Myocardial hypoxia is the low oxygen tension in the heart tissue implicated in many diseases, including ischemia, cardiac dysfunction, or after heart procurement for transplantation. Oxygen-generating microparticles have recently emerged as a potential strategy for supplying oxygen to sustain cell survival, growth, and tissue functionality in hypoxia. Here, we prepared oxygen-generating microparticles with poly D,L-lactic-co-glycolic acid, and calcium peroxide (CPO), which yielded a continuous morphology capable of sustained oxygen release for up to 24 h. We demonstrated that CPO microparticles increased primary rat cardiomyocyte metabolic activity while not affecting cell viability during hypoxia. Moreover, hypoxia-inducible factor (HIF)-1α, which is upregulated during hypoxia, can be downregulated by delivering oxygen using CPO microparticles. Single-cell traction force microscopy data demonstrated that the reduced energy generated by hypoxic cells could be restored using CPO microparticles. We engineered cardiac tissues that showed higher contractility in the presence of CPO microparticles compared to hypoxic cells. Finally, we observed reduced myocardial injuries in ex vivo rabbit hearts treated with CPO microparticles. In contrast, an acute early myocardial injury was observed for the hearts treated with control saline solution in hypoxia. In conclusion, CPO microparticles improved cell and tissue contractility and gene expression while reducing hypoxia-induced myocardial injuries in the heart. STATEMENT OF SIGNIFICANCE: Oxygen-releasing microparticles can reduce myocardial ischemia, allograft rejection, or irregular heartbeats after heart transplantation. Here we present biodegradable oxygen-releasing microparticles that are capable of sustained oxygen release for more than 24 hrs. We then studied the impact of sustained oxygen release from microparticles on gene expresseion and cardiac cell and tissue function. Previous studies have not measured cardiac tissue or cell mechanics during hypoxia, which is important for understanding proper cardiac function and beating. Using traction force microscopy and an engineered tissue-on-a-chip, we demonstrated that our oxygen-releasing microparticles improve cell and tissue contractility during hypoxia while downregulating the HIF-1α expression level. Finally, using the microparticles, we showed reduced myocardial injuries in rabbit heart tissue, confirming the potential of the particles to be used for organ transplantation or tissue engineering.
Collapse
Affiliation(s)
- Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Sivakoti Sangabathuni
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA; Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Xuexiang Zhang
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Masoud Edalati
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Wei Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Ana Lopez Hernandez
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Mehmet R Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| |
Collapse
|
8
|
Sesena-Rubfiaro A, Prajapati NJ, Paolino L, Lou L, Cotayo D, Pandey P, Shaver M, Hutcheson J, Agarwal A, He J. Membrane Remodeling of Human-Engineered Cardiac Tissue by Chronic Electric Stimulation. ACS Biomater Sci Eng 2023; 9:1644-1655. [PMID: 36765460 PMCID: PMC10542861 DOI: 10.1021/acsbiomaterials.2c01370] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) show immature features, but these are improved by integration into 3D cardiac constructs. In addition, it has been demonstrated that physical manipulations such as electrical stimulation (ES) are highly effective in improving the maturation of human-engineered cardiac tissue (hECT) derived from hiPSC-CMs. Here, we continuously applied an ES in capacitive coupling configuration, which is below the pacing threshold, to millimeter-sized hECTs for 1-2 weeks. Meanwhile, the structural and functional developments of the hECTs were monitored and measured using an array of assays. Of particular note, a nanoscale imaging technique, scanning ion conductance microscopy (SICM), has been used to directly image membrane remodeling of CMs at different locations on the tissue surface. Periodic crest/valley patterns with a distance close to the sarcomere length appeared on the membrane of CMs near the edge of the tissue after ES, suggesting the enhanced transverse tubulation network. The SICM observation is also supported by the fluorescence images of the transverse tubulation network and α-actinin. Correspondingly, essential cardiac functions such as calcium handling and contraction force generation were improved. Our study provides evidence that chronic subthreshold ES can still improve the structural and functional developments of hECTs.
Collapse
Affiliation(s)
| | - Navin J. Prajapati
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Lia Paolino
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
| | - Lihua Lou
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Daniel Cotayo
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Popular Pandey
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Mohammed Shaver
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
| | - Joshua Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami FL 33199, USA
| | - Arvind Agarwal
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Jin He
- Department of Physics, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami FL 33199, USA
| |
Collapse
|
9
|
Kałużna E, Nadel A, Zimna A, Rozwadowska N, Kolanowski T. Modeling the human heart ex vivo-current possibilities and strive for future applications. J Tissue Eng Regen Med 2022; 16:853-874. [PMID: 35748158 PMCID: PMC9796015 DOI: 10.1002/term.3335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
Collapse
Affiliation(s)
- Ewelina Kałużna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Nadel
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Zimna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | | | | |
Collapse
|
10
|
Mehrotra S, de Melo BAG, Miscuglio M, Kiaee K, Shin SR, Mandal BB. Mimicking Native Heart Tissue Physiology and Pathology in Silk Fibroin Constructs through a Perfusion-Based Dynamic Mechanical Stimulation Microdevice. Adv Healthc Mater 2022; 11:e2101678. [PMID: 34971210 PMCID: PMC11041525 DOI: 10.1002/adhm.202101678] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/23/2021] [Indexed: 11/07/2022]
Abstract
In vitro cardiomyocyte (CM) maturation is an imperative step to replicate native heart tissue-like structures as cardiac tissue grafts or as drug screening platforms. CMs are known to interpret biophysical cues such as stiffness, topography, external mechanical stimulation or dynamic perfusion load through mechanotransduction and change their behavior, organization, and maturation. In this regard, a silk-based cardiac tissue (CT) coupled with a dynamic perfusion-based mechanical stimulation platform (DMM) for achieving maturation and functionality in vitro is tried to be delivered. Silk fibroin (SF) is used to fabricate lamellar scaffolds to provide native tissue-like anisotropic architecture and is found to be nonimmunogenic and biocompatible allowing cardiomyocyte attachment and growth in vitro. Further, the scaffolds display excellent mechanical properties by their ability to undergo cyclic compressions without any deformation when places in the DMM. Gradient compression strains (5% to 20%), mimicking the native physiological and pathological conditions, are applied to the cardiomyocyte culture seeded on lamellar silk scaffolds in the DMM. A strain-dependent difference in cardiomyocyte maturation, gene expression, sarcomere elongation, and extracellular matrix formation is observed. These silk-based CTs matured in the DMM can open up several avenues toward the development of host-specific grafts and in vitro models for drug screening.
Collapse
Affiliation(s)
- Shreya Mehrotra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Bruna Alice Gomes de Melo
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, 04039-002, Brazil
| | - Mario Miscuglio
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
- Department of Electrical and Computer Engineering, George Washington University, Washington, DC, 20052, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| |
Collapse
|
11
|
Do Human iPSC-Derived Cardiomyocytes Cultured on PLA Scaffolds Induce Expression of CD28/CTLA-4 by T Lymphocytes? J Funct Biomater 2022; 13:jfb13010006. [PMID: 35076538 PMCID: PMC8788528 DOI: 10.3390/jfb13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/29/2021] [Accepted: 01/07/2022] [Indexed: 02/05/2023] Open
Abstract
Many research groups have developed various types of tissue-engineered cardiac constructs. However, the immunological properties of such artificial tissues are not yet fully understood. Previously, we developed microfiber scaffolds carrying human iPSC-derived cardiomyocytes (hiPSC-CM). In this work, we evaluated the ability of these tissue-engineered constructs to activate the expression of CD28 and CTLA-4 proteins on T lymphocytes, which are early markers of the immune response. For this purpose, electrospun PLA microfiber scaffolds were seeded with hiPSC-CM and cultured for 2 weeks. Allogeneic mononuclear cells were then co-cultured for 48 h with three groups of samples: bare scaffolds, pure cardiomyocyte culture and tissue-engineered constructs, followed by analysis of CD28/CTLA-4 expression on T lymphocytes using flow cytometry. PLA scaffolds and concanavalin A stimulation (positive control) statistically significantly increased CD28 expression on CD4+ T cells (up to 61.3% and 66.3%) CD8+ T cells (up to 17.8% and 21.7%). CD28/CTLA-4 expression was not increased when T lymphocytes were co-cultured with cardiac tissue-engineered constructs and iPSC-CM monolayers. Thus, iPSC-CM in monolayers and on PLA microfiber scaffolds did not induce T cell activation, which suggests that such cardiac constructs would not be a cause of rejection after implantation.
Collapse
|
12
|
Bomkamp C, Skaalure SC, Fernando GF, Ben‐Arye T, Swartz EW, Specht EA. Scaffolding Biomaterials for 3D Cultivated Meat: Prospects and Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102908. [PMID: 34786874 PMCID: PMC8787436 DOI: 10.1002/advs.202102908] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/12/2021] [Indexed: 05/03/2023]
Abstract
Cultivating meat from stem cells rather than by raising animals is a promising solution to concerns about the negative externalities of meat production. For cultivated meat to fully mimic conventional meat's organoleptic and nutritional properties, innovations in scaffolding technology are required. Many scaffolding technologies are already developed for use in biomedical tissue engineering. However, cultivated meat production comes with a unique set of constraints related to the scale and cost of production as well as the necessary attributes of the final product, such as texture and food safety. This review discusses the properties of vertebrate skeletal muscle that will need to be replicated in a successful product and the current state of scaffolding innovation within the cultivated meat industry, highlighting promising scaffold materials and techniques that can be applied to cultivated meat development. Recommendations are provided for future research into scaffolds capable of supporting the growth of high-quality meat while minimizing production costs. Although the development of appropriate scaffolds for cultivated meat is challenging, it is also tractable and provides novel opportunities to customize meat properties.
Collapse
Affiliation(s)
- Claire Bomkamp
- The Good Food Institute1380 Monroe St. NW #229WashingtonDC20010USA
| | | | | | - Tom Ben‐Arye
- The Good Food Institute1380 Monroe St. NW #229WashingtonDC20010USA
| | - Elliot W. Swartz
- The Good Food Institute1380 Monroe St. NW #229WashingtonDC20010USA
| | | |
Collapse
|
13
|
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
|
14
|
Roth BJ. Bidomain modeling of electrical and mechanical properties of cardiac tissue. BIOPHYSICS REVIEWS 2021; 2:041301. [PMID: 38504719 PMCID: PMC10903405 DOI: 10.1063/5.0059358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/15/2021] [Indexed: 03/21/2024]
Abstract
Throughout the history of cardiac research, there has been a clear need to establish mathematical models to complement experimental studies. In an effort to create a more complete picture of cardiac phenomena, the bidomain model was established in the late 1970s to better understand pacing and defibrillation in the heart. This mathematical model has seen ongoing use in cardiac research, offering mechanistic insight that could not be obtained from experimental pursuits. Introduced from a historical perspective, the origins of the bidomain model are reviewed to provide a foundation for researchers new to the field and those conducting interdisciplinary research. The interplay of theory and experiment with the bidomain model is explored, and the contributions of this model to cardiac biophysics are critically evaluated. Also discussed is the mechanical bidomain model, which is employed to describe mechanotransduction. Current challenges and outstanding questions in the use of the bidomain model are addressed to give a forward-facing perspective of the model in future studies.
Collapse
Affiliation(s)
- Bradley J. Roth
- Department of Physics, Oakland University, Rochester, Michigan 48309, USA
| |
Collapse
|
15
|
Tamargo MA, Nash TR, Fleischer S, Kim Y, Vila OF, Yeager K, Summers M, Zhao Y, Lock R, Chavez M, Costa T, Vunjak-Novakovic G. milliPillar: A Platform for the Generation and Real-Time Assessment of Human Engineered Cardiac Tissues. ACS Biomater Sci Eng 2021; 7:5215-5229. [PMID: 34668692 DOI: 10.1021/acsbiomaterials.1c01006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Engineered cardiac tissues derived from human induced pluripotent stem cells (iPSCs) are increasingly used for drug discovery, pharmacology and in models of development and disease. While there are numerous platforms to engineer cardiac tissues, they often require expensive and nonconventional equipment and utilize complex video-processing algorithms. As a result, only specialized academic laboratories have been able to harness this technology. In addition, methodologies and tissue features have been challenging to reproduce between different groups and models. Here, we describe a facile technology (milliPillar) that covers the entire pipeline required for studies of engineered cardiac tissues. We include methodologies for (i) platform fabrication, (ii) cardiac tissue generation, (iii) electrical stimulation, (iv) automated real-time data acquisition, and (v) advanced video analyses. We validate these methodologies and demonstrate the versatility of the platform by showcasing the fabrication of tissues in different hydrogel materials and using cardiomyocytes derived from different iPSC lines in combination with different types of stromal cells. We also validate the long-term culture of tissues within the platform and provide protocols for automated analysis of force generation and calcium flux using both brightfield and fluorescence imaging. Lastly, we demonstrate the compatibility of the milliPillar platform with electromechanical stimulation to enhance cardiac tissue function. We expect that this resource will provide a valuable and user-friendly tool for the generation and real-time assessment of engineered human cardiac tissues for basic and translational studies.
Collapse
Affiliation(s)
- Manuel Alejandro Tamargo
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States.,Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Trevor Ray Nash
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States.,Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Olaia Fernandez Vila
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States.,Gladstone Institutes, San Francisco, California 94158, United States
| | - Keith Yeager
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Max Summers
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Yimu Zhao
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Miguel Chavez
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Troy Costa
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States.,Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, New York 10032, United States
| |
Collapse
|
16
|
Khalil NN, McCain ML. Engineering the Cellular Microenvironment of Post-infarct Myocardium on a Chip. Front Cardiovasc Med 2021; 8:709871. [PMID: 34336962 PMCID: PMC8316619 DOI: 10.3389/fcvm.2021.709871] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Myocardial infarctions are one of the most common forms of cardiac injury and death worldwide. Infarctions cause immediate necrosis in a localized region of the myocardium, which is followed by a repair process with inflammatory, proliferative, and maturation phases. This repair process culminates in the formation of scar tissue, which often leads to heart failure in the months or years after the initial injury. In each reparative phase, the infarct microenvironment is characterized by distinct biochemical, physical, and mechanical features, such as inflammatory cytokine production, localized hypoxia, and tissue stiffening, which likely each contribute to physiological and pathological tissue remodeling by mechanisms that are incompletely understood. Traditionally, simplified two-dimensional cell culture systems or animal models have been implemented to elucidate basic pathophysiological mechanisms or predict drug responses following myocardial infarction. However, these conventional approaches offer limited spatiotemporal control over relevant features of the post-infarct cellular microenvironment. To address these gaps, Organ on a Chip models of post-infarct myocardium have recently emerged as new paradigms for dissecting the highly complex, heterogeneous, and dynamic post-infarct microenvironment. In this review, we describe recent Organ on a Chip models of post-infarct myocardium, including their limitations and future opportunities in disease modeling and drug screening.
Collapse
Affiliation(s)
- Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Canonico F, Chirivi M, Maiullari F, Milan M, Rizzi R, Arcudi A, Galli M, Pane M, Gowran A, Pompilio G, Mercuri E, Crea F, Bearzi C, D'Amario D. Focus on the road to modelling cardiomyopathy in muscular dystrophy. Cardiovasc Res 2021; 118:1872-1884. [PMID: 34254111 DOI: 10.1093/cvr/cvab232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/07/2021] [Indexed: 11/13/2022] Open
Abstract
Alterations in the DMD gene, which codes for the protein dystrophin, cause forms of dystrophinopathies such as Duchenne muscular dystrophy, an X-linked disease. Cardiomyopathy linked to DMD mutations is becoming the leading cause of death in patients with dystrophinopathy. Since phenotypic pathophysiological mechanisms are not fully understood, the improvement and development of new disease models, considering their relative advantages and disadvantages, is essential. The application of genetic engineering approaches on induced pluripotent stem cells, such as gene editing technology, enables the development of physiologically relevant human cell models for in vitro dystrophinopathy studies. The combination of induced pluripotent stem cells-derived cardiovascular cell types and 3 D bioprinting technologies hold great promise for the study of dystrophin-linked cardiomyopathy. This combined approach enables the assessment of responses to physical or chemical stimuli, and the influence of pharmaceutical approaches. The critical objective of in vitro microphysiological systems is to more accurately reproduce the microenvironment observed in vivo. Ground-breaking methodology involving the connection of multiple microphysiological systems comprised of different tissues would represent a move toward precision body-on-chip disease modelling could lead to a critical expansion in what is known about inter-organ responses to disease and novel therapies that have the potential to replace animal models. In this review, we will focus on the generation, development, and application of current cellular, animal and potential for bio-printed models, in the study of the pathophysiological mechanisms underlying dystrophin-linked cardiomyopathy in the direction of personalized medicine.
Collapse
Affiliation(s)
- Francesco Canonico
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Maila Chirivi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Fabio Maiullari
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Marika Milan
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy.,Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Segrate, Milan, Italy
| | - Alessandra Arcudi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Mattia Galli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Marika Pane
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Women, Children and Public Health Sciences, Rome, Italy
| | - Aoife Gowran
- Centro Cardiologico Monzino IRCCS, Unit of Vascular Biology and Regenerative Medicine, Milan, Italy
| | - Giulio Pompilio
- Centro Cardiologico Monzino IRCCS, Unit of Vascular Biology and Regenerative Medicine, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Eugenio Mercuri
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Women, Children and Public Health Sciences, Rome, Italy
| | - Filippo Crea
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Claudia Bearzi
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy.,Institute of Genetic and Biomedical Research, National Research Council (IRGB-CNR), Milan, Italy
| | - Domenico D'Amario
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| |
Collapse
|
18
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
19
|
Tavakol DN, Fleischer S, Vunjak-Novakovic G. Harnessing organs-on-a-chip to model tissue regeneration. Cell Stem Cell 2021; 28:993-1015. [PMID: 34087161 DOI: 10.1016/j.stem.2021.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue engineering has markedly matured since its early beginnings in the 1980s. In addition to the original goal to regenerate damaged organs, the field has started to explore modeling of human physiology "in a dish." Induced pluripotent stem cell (iPSC) technologies now enable studies of organ regeneration and disease modeling in a patient-specific context. We discuss the potential of "organ-on-a-chip" systems to study regenerative therapies with focus on three distinct organ systems: cardiac, respiratory, and hematopoietic. We propose that the combinatorial studies of human tissues at these two scales would help realize the translational potential of tissue engineering.
Collapse
Affiliation(s)
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Medicine, Columbia University, New York, NY.
| |
Collapse
|
20
|
Jiang X, Cheng H, Huang J, Cui C, Zhu Y, Lin Y, Miao W, Liu H, Chen H, Ju W, Chen M. Construction of chamber-specific engineered cardiac tissues in vitro with human iPSC-derived cardiomyocytes and human foreskin fibroblasts. J Biosci Bioeng 2021; 132:198-205. [PMID: 34074596 DOI: 10.1016/j.jbiosc.2021.04.012] [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: 02/09/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022]
Abstract
Human-induced pluripotent stem cell (hiPSC) technology and directed cardiac differentiation technology can provide a continuous supply of cells for disease modeling, drug screening, and cell therapy. However, two-dimensional (2D) cells often fail to faithfully reflect the physiological structure and function of the heart. Considering the contractile function is the most critical and easy-to-understand function of cardiomyocytes, the engineered cardiac tissues (ECT) with mechanical properties may serve as an appropriate three-dimensional (3D) platform for drug evaluation. At present, there are various methods to generate ECTs, some of which are quite costly. In the present study, we proposed that human foreskin fibroblast (HFF) cells, as a cost-effective and accessible cell source, can promote the compaction and remodeling of ECTs. The HFFs derived ECTs displayed stable structural and functional characteristics with a higher performance-to-price ratio. Moreover, both ECTs made from atrial and ventricular cardiomyocytes showed an excellent drug response, demonstrating that the ECT with HFFs as an easy and reliable platform for drug evaluation.
Collapse
Affiliation(s)
- Xiaohong Jiang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hongyi Cheng
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiayi Huang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chang Cui
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yue Zhu
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yongping Lin
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weilun Miao
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hailei Liu
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hongwu Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weizhu Ju
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Minglong Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
21
|
Jayne RK, Karakan MÇ, Zhang K, Pierce N, Michas C, Bishop DJ, Chen CS, Ekinci KL, White AE. Direct laser writing for cardiac tissue engineering: a microfluidic heart on a chip with integrated transducers. LAB ON A CHIP 2021; 21:1724-1737. [PMID: 33949395 DOI: 10.1039/d0lc01078b] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We have developed a microfluidic platform for engineering cardiac microtissues in highly-controlled microenvironments. The platform is fabricated using direct laser writing (DLW) lithography and soft lithography, and contains four separate devices. Each individual device houses a cardiac microtissue and is equipped with an integrated strain actuator and a force sensor. Application of external pressure waves to the platform results in controllable time-dependent forces on the microtissues. Conversely, oscillatory forces generated by the microtissues are transduced into measurable electrical outputs. We demonstrate the capabilities of this platform by studying the response of cardiac microtissues derived from human induced pluripotent stem cells (hiPSC) under prescribed mechanical loading and pacing. This platform will be used for fundamental studies and drug screening on cardiac microtissues.
Collapse
Affiliation(s)
- Rachael K Jayne
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA
| | - M Çağatay Karakan
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Noelle Pierce
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Christos Michas
- Photonics Center, Boston University, Boston, MA 02215, USA and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - David J Bishop
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA and Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA and Department of Physics, Boston University, Boston, MA 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Kamil L Ekinci
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Alice E White
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA and Department of Physics, Boston University, Boston, MA 02215, USA
| |
Collapse
|
22
|
Cho S, Lee C, Skylar-Scott MA, Heilshorn SC, Wu JC. Reconstructing the heart using iPSCs: Engineering strategies and applications. J Mol Cell Cardiol 2021; 157:56-65. [PMID: 33895197 DOI: 10.1016/j.yjmcc.2021.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have emerged as a key component of cardiac tissue engineering, enabling studies of cardiovascular disease mechanisms, drug responses, and developmental processes in human 3D tissue models assembled from isogenic cells. Since the very first engineered heart tissues were introduced more than two decades ago, a wide array of iPSC-derived cardiac spheroids, organoids, and heart-on-a-chip models have been developed incorporating the latest available technologies and materials. In this review, we will first outline the fundamental biological building blocks required to form a functional unit of cardiac muscle, including iPSC-derived cells differentiated by soluble factors (e.g., small molecules), extracellular matrix scaffolds, and exogenous biophysical maturation cues. We will then summarize the different fabrication approaches and strategies employed to reconstruct the heart in vitro at varying scales and geometries. Finally, we will discuss how these platforms, with continued improvements in scalability and tissue maturity, can contribute to both basic cardiovascular research and clinical applications in the future.
Collapse
Affiliation(s)
- Sangkyun Cho
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Chelsea Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Mark A Skylar-Scott
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Betty Irene Moore Children's Heart Center, Stanford Children's Health, Stanford, CA 94025, USA
| | - Sarah C Heilshorn
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA.
| |
Collapse
|
23
|
Shi H, Wang C, Ma Z. Stimuli-responsive biomaterials for cardiac tissue engineering and dynamic mechanobiology. APL Bioeng 2021; 5:011506. [PMID: 33688616 PMCID: PMC7929620 DOI: 10.1063/5.0025378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
Since the term "smart materials" was put forward in the 1980s, stimuli-responsive biomaterials have been used as powerful tools in tissue engineering, mechanobiology, and clinical applications. For the purpose of myocardial repair and regeneration, stimuli-responsive biomaterials are employed to fabricate hydrogels and nanoparticles for targeted delivery of therapeutic drugs and cells, which have been proved to alleviate disease progression and enhance tissue regeneration. By reproducing the sophisticated and dynamic microenvironment of the native heart, stimuli-responsive biomaterials have also been used to engineer dynamic culture systems to understand how cardiac cells and tissues respond to progressive changes in extracellular microenvironments, enabling the investigation of dynamic cell mechanobiology. Here, we provide an overview of stimuli-responsive biomaterials used in cardiovascular research applications, with a specific focus on cardiac tissue engineering and dynamic cell mechanobiology. We also discuss how these smart materials can be utilized to mimic the dynamic microenvironment during heart development, which might provide an opportunity to reveal the fundamental mechanisms of cardiomyogenesis and cardiac maturation.
Collapse
Affiliation(s)
| | | | - Zhen Ma
- Author to whom correspondence should be addressed:
| |
Collapse
|
24
|
Jorba I, Mostert D, Hermans LH, van der Pol A, Kurniawan NA, Bouten CV. In Vitro Methods to Model Cardiac Mechanobiology in Health and Disease. Tissue Eng Part C Methods 2021; 27:139-151. [PMID: 33514281 PMCID: PMC7984657 DOI: 10.1089/ten.tec.2020.0342] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022] Open
Abstract
In vitro cardiac modeling has taken great strides in the past decade. While most cell and engineered tissue models have focused on cell and tissue contractile function as readouts, mechanobiological cues from the cell environment that affect this function, such as matrix stiffness or organization, are less well explored. In this study, we review two-dimensional (2D) and three-dimensional (3D) models of cardiac function that allow for systematic manipulation or precise control of mechanobiological cues under simulated (patho)physiological conditions while acquiring multiple readouts of cell and tissue function. We summarize the cell types used in these models and highlight the importance of linking 2D and 3D models to address the multiscale organization and mechanical behavior. Finally, we provide directions on how to advance in vitro modeling for cardiac mechanobiology using next generation hydrogels that mimic mechanical and structural environmental features at different length scales and diseased cell types, along with the development of new tissue fabrication and readout techniques. Impact statement Understanding the impact of mechanobiology in cardiac (patho)physiology is essential for developing effective tissue regeneration and drug discovery strategies and requires detailed cause-effect studies. The development of three-dimensional in vitro models allows for such studies with high experimental control, while integrating knowledge from complementary cell culture models and in vivo studies for this purpose. Complemented by the use of human-induced pluripotent stem cells, with or without predisposed genetic diseases, these in vitro models will offer promising outlooks to delineate the impact of mechanobiological cues on human cardiac (patho)physiology in a dish.
Collapse
Affiliation(s)
- Ignasi Jorba
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Dylan Mostert
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Leon H.L. Hermans
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Atze van der Pol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Carlijn V.C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| |
Collapse
|
25
|
Hendrickson T, Mancino C, Whitney L, Tsao C, Rahimi M, Taraballi F. Mimicking cardiac tissue complexity through physical cues: A review on cardiac tissue engineering approaches. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102367. [PMID: 33549819 DOI: 10.1016/j.nano.2021.102367] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/06/2021] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Cardiovascular diseases are the number one killer in the world.1,2 Currently, there are no clinical treatments to regenerate damaged cardiac tissue, leaving patients to develop further life-threatening cardiac complications. Cardiac tissue has multiple functional demands including vascularization, contraction, and conduction that require many synergic components to properly work. Most of these functions are a direct result of the cardiac tissue structure and composition, and, for this reason, tissue engineering strongly proposed to develop substitute engineered heart tissues (EHTs). EHTs usually have combined pluripotent stem cells and supporting scaffolds with the final aim to repair or replace the damaged native tissue. However, as simple as this idea is, indeed, it resulted, after many attempts in the field, to be very challenging. Without design complexity, EHTs remain unable to mature fully and integrate into surrounding heart tissue resulting in minimal in vivo effects.3 Lately, there has been a growing body of evidence that a complex, multifunctional approach through implementing scaffold designs, cellularization, and molecular release appears to be essential in the development of a functional cardiac EHTs.4-6 This review covers the advancements in EHTs developments focusing on how to integrate contraction, conduction, and vascularization mimics and how combinations have resulted in improved designs thus warranting further investigation to develop a clinically applicable treatment.
Collapse
Affiliation(s)
- Troy Hendrickson
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA; Texas A&M MD/PhD Program, Texas A&M Health Science Center, College Station, TX, USA
| | - Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, (MI), Italy
| | - Lauren Whitney
- Texas A&M Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Chris Tsao
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA
| | - Maham Rahimi
- Department of Cardiovascular Surgery, Houston Methodist, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
26
|
Tadevosyan K, Iglesias-García O, Mazo MM, Prósper F, Raya A. Engineering and Assessing Cardiac Tissue Complexity. Int J Mol Sci 2021; 22:ijms22031479. [PMID: 33540699 PMCID: PMC7867236 DOI: 10.3390/ijms22031479] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiac tissue engineering is very much in a current focus of regenerative medicine research as it represents a promising strategy for cardiac disease modelling, cardiotoxicity testing and cardiovascular repair. Advances in this field over the last two decades have enabled the generation of human engineered cardiac tissue constructs with progressively increased functional capabilities. However, reproducing tissue-like properties is still a pending issue, as constructs generated to date remain immature relative to native adult heart. Moreover, there is a high degree of heterogeneity in the methodologies used to assess the functionality and cardiac maturation state of engineered cardiac tissue constructs, which further complicates the comparison of constructs generated in different ways. Here, we present an overview of the general approaches developed to generate functional cardiac tissues, discussing the different cell sources, biomaterials, and types of engineering strategies utilized to date. Moreover, we discuss the main functional assays used to evaluate the cardiac maturation state of the constructs, both at the cellular and the tissue levels. We trust that researchers interested in developing engineered cardiac tissue constructs will find the information reviewed here useful. Furthermore, we believe that providing a unified framework for comparison will further the development of human engineered cardiac tissue constructs displaying the specific properties best suited for each particular application.
Collapse
Affiliation(s)
- Karine Tadevosyan
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Center for Networked Biomedical Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| |
Collapse
|
27
|
Modulating hESC-derived cardiomyocyte and endothelial cell function with triple-helical peptides for heart tissue engineering. Biomaterials 2020; 269:120612. [PMID: 33385684 PMCID: PMC7884910 DOI: 10.1016/j.biomaterials.2020.120612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 11/21/2022]
Abstract
In this study, we investigated the role of cardiomyocyte (CM) and endothelial cell (EC) specific interactions with collagen in the assembly of an operational myocardium in vitro. Engineered cardiac patches represent valuable tools for myocardial repair following infarction and are generally constituted of a suitable biomaterial populated by CMs and supportive cell types. Among those, ECs are required for tissue vascularization and positively modulate CM function. To direct the function of human embryonic stem cell (hESC)-derived CM and EC seeded on biomaterials, we replicated cell-collagen interactions, which regulate cellular behaviour in the native myocardium, using triple-helical peptides (THPs) that are ligands for collagen-binding proteins. THPs enhanced proliferation and activity of CMs and ECs separately and in co-culture, drove CM maturation and enabled coordinated cellular contraction on collagen films. These results highlight the importance of collagen interactions on cellular response and establish THP-functionalized biomaterials as novel tools to produce engineered cardiac tissues.
Collapse
|
28
|
Jeong YJ, Kim DS, Kim JY, Oyunbaatar NE, Shanmugasundaram A, Kim ES, Lee DW. On-stage bioreactor platform integrated with nano-patterned and gold-coated PDMS diaphragm for live cell stimulation and imaging. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111355. [PMID: 33254975 DOI: 10.1016/j.msec.2020.111355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/03/2020] [Accepted: 08/05/2020] [Indexed: 12/26/2022]
Abstract
Over the years, several in-vitro biosensing platforms have been developed for enhancing the maturation of the cultured cells. However, most of the proposed platforms met with limited success due to its inability for live-cell imaging, complicated fabrication, and not being advantageous from an economic perspective due to a higher price. To overcome the drawbacks of the current state-of-the-art, herein, we developed a next-generation stage-top incubator (STI) incorporated with nano grooves patterned PDMS diaphragm (NGPPD). The proposed device consists of a miniatured STI, the NGPPD functional well plates, and a mechanical stimulator. A thin layer of gold (Au) is deposited on the NGPPD to enhanced myogenic differentiation, cell maturation, and cell-cell interactions. The nano grooves are integrated on the PDMS surface to align the cardiomyocytes in the grooved direction during the culture period. The cardiomyocytes cultivated on the Au-deposited NGPPD are stimulated topographically and mechanically during the cultivation period. The enhanced cardiomyocytes maturation cultured on the Au-deposited NGPPD is experimentally demonstrated using immunofluorescence staining and PCR analysis.
Collapse
Affiliation(s)
- Yun-Jin Jeong
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Su Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jong Yun Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Arunkumar Shanmugasundaram
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea; Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
29
|
Zhang W, Huang G, Xu F. Engineering Biomaterials and Approaches for Mechanical Stretching of Cells in Three Dimensions. Front Bioeng Biotechnol 2020; 8:589590. [PMID: 33154967 PMCID: PMC7591716 DOI: 10.3389/fbioe.2020.589590] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical stretch is widely experienced by cells of different tissues in the human body and plays critical roles in regulating their behaviors. Numerous studies have been devoted to investigating the responses of cells to mechanical stretch, providing us with fruitful findings. However, these findings have been mostly observed from two-dimensional studies and increasing evidence suggests that cells in three dimensions may behave more closely to their in vivo behaviors. While significant efforts and progresses have been made in the engineering of biomaterials and approaches for mechanical stretching of cells in three dimensions, much work remains to be done. Here, we briefly review the state-of-the-art researches in this area, with focus on discussing biomaterial considerations and stretching approaches. We envision that with the development of advanced biomaterials, actuators and microengineering technologies, more versatile and predictive three-dimensional cell stretching models would be available soon for extensive applications in such fields as mechanobiology, tissue engineering, and drug screening.
Collapse
Affiliation(s)
- Weiwei Zhang
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Guoyou Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing University, Chongqing, China
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
30
|
Lavine KJ, Greenberg MJ. Beyond genomics-technological advances improving the molecular characterization and precision treatment of heart failure. Heart Fail Rev 2020; 26:405-415. [PMID: 32885327 DOI: 10.1007/s10741-020-10021-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/04/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major cause of heart failure and cardiovascular mortality. In the past 20 years, there has been an overwhelming focus on developing therapeutics that target common downstream disease pathways thought to be involved in all forms of heart failure independent of the initial etiology. While this strategy is effective at the population level, individual responses vary tremendously and only approximately one third of patients receive benefit from modern heart failure treatments. In this perspective, we propose that DCM should be considered as a collection of diseases with a common phenotype of left ventricular dilation and systolic dysfunction rather than a single disease entity, and that mechanism-based classification of disease subtypes will revolutionize our understanding and clinical approach towards DCM. We discuss how these efforts are central to realizing the potential of precision medicine and how they are empowered by the development of new tools that allow investigators to strategically employ genomic and transcriptomic information. Finally, we outline an investigational strategy to (1) define DCM at the patient level, (2) develop new tools to model and mechanistically dissect subtypes of human heart failure, and (3) harness these insights for the development of precision therapeutics.
Collapse
Affiliation(s)
- Kory J Lavine
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8086, St. Louis, MO, 63110, USA.
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8231, St. Louis, MO, 63110, USA.
| |
Collapse
|
31
|
Montero P, Flandes-Iparraguirre M, Musquiz S, Pérez Araluce M, Plano D, Sanmartín C, Orive G, Gavira JJ, Prosper F, Mazo MM. Cells, Materials, and Fabrication Processes for Cardiac Tissue Engineering. Front Bioeng Biotechnol 2020; 8:955. [PMID: 32850768 PMCID: PMC7431658 DOI: 10.3389/fbioe.2020.00955] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease is the number one killer worldwide, with myocardial infarction (MI) responsible for approximately 1 in 6 deaths. The lack of endogenous regenerative capacity, added to the deleterious remodelling programme set into motion by myocardial necrosis, turns MI into a progressively debilitating disease, which current pharmacological therapy cannot halt. The advent of Regenerative Therapies over 2 decades ago kick-started a whole new scientific field whose aim was to prevent or even reverse the pathological processes of MI. As a highly dynamic organ, the heart displays a tight association between 3D structure and function, with the non-cellular components, mainly the cardiac extracellular matrix (ECM), playing both fundamental active and passive roles. Tissue engineering aims to reproduce this tissue architecture and function in order to fabricate replicas able to mimic or even substitute damaged organs. Recent advances in cell reprogramming and refinement of methods for additive manufacturing have played a critical role in the development of clinically relevant engineered cardiovascular tissues. This review focuses on the generation of human cardiac tissues for therapy, paying special attention to human pluripotent stem cells and their derivatives. We provide a perspective on progress in regenerative medicine from the early stages of cell therapy to the present day, as well as an overview of cellular processes, materials and fabrication strategies currently under investigation. Finally, we summarise current clinical applications and reflect on the most urgent needs and gaps to be filled for efficient translation to the clinical arena.
Collapse
Affiliation(s)
- Pilar Montero
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - María Flandes-Iparraguirre
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - Saioa Musquiz
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
| | - María Pérez Araluce
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- University Institute for Regenerative Medicine and Oral Implantology – UIRMI (UPV/EHU – Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, Singapore, Singapore
| | - Juan José Gavira
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Cardiology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
32
|
Massai D, Pisani G, Isu G, Rodriguez Ruiz A, Cerino G, Galluzzi R, Pisanu A, Tonoli A, Bignardi C, Audenino AL, Marsano A, Morbiducci U. Bioreactor Platform for Biomimetic Culture and in situ Monitoring of the Mechanical Response of in vitro Engineered Models of Cardiac Tissue. Front Bioeng Biotechnol 2020; 8:733. [PMID: 32766218 PMCID: PMC7381147 DOI: 10.3389/fbioe.2020.00733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
In the past two decades, relevant advances have been made in the generation of engineered cardiac constructs to be used as functional in vitro models for cardiac research or drug testing, and with the ultimate but still challenging goal of repairing the damaged myocardium. To support cardiac tissue generation and maturation in vitro, the application of biomimetic physical stimuli within dedicated bioreactors is crucial. In particular, cardiac-like mechanical stimulation has been demonstrated to promote development and maturation of cardiac tissue models. Here, we developed an automated bioreactor platform for tunable cyclic stretch and in situ monitoring of the mechanical response of in vitro engineered cardiac tissues. To demonstrate the bioreactor platform performance and to investigate the effects of cyclic stretch on construct maturation and contractility, we developed 3D annular cardiac tissue models based on neonatal rat cardiac cells embedded in fibrin hydrogel. The constructs were statically pre-cultured for 5 days and then exposed to 4 days of uniaxial cyclic stretch (sinusoidal waveform, 10% strain, 1 Hz) within the bioreactor. Explanatory biological tests showed that cyclic stretch promoted cardiomyocyte alignment, maintenance, and maturation, with enhanced expression of typical mature cardiac markers compared to static controls. Moreover, in situ monitoring showed increasing passive force of the constructs along the dynamic culture. Finally, only the stretched constructs were responsive to external electrical pacing with synchronous and regular contractile activity, further confirming that cyclic stretching was instrumental for their functional maturation. This study shows that the proposed bioreactor platform is a reliable device for cyclic stretch culture and in situ monitoring of the passive mechanical response of the cultured constructs. The innovative feature of acquiring passive force measurements in situ and along the culture allows monitoring the construct maturation trend without interrupting the culture, making the proposed device a powerful tool for in vitro investigation and ultimately production of functional engineered cardiac constructs.
Collapse
Affiliation(s)
- Diana Massai
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Giuseppe Pisani
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giuseppe Isu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andres Rodriguez Ruiz
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Giulia Cerino
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Renato Galluzzi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Alessia Pisanu
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andrea Tonoli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Cristina Bignardi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Alberto L Audenino
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Anna Marsano
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| |
Collapse
|
33
|
Whole Organ Engineering: Approaches, Challenges, and Future Directions. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10124277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
End-stage organ failure remains a leading cause of morbidity and mortality across the globe. The only curative treatment option currently available for patients diagnosed with end-stage organ failure is organ transplantation. However, due to a critical shortage of organs, only a fraction of these patients are able to receive a viable organ transplantation. Those patients fortunate enough to receive a transplant must then be subjected to a lifelong regimen of immunosuppressant drugs. The concept of whole organ engineering offers a promising alternative to organ transplantation that overcomes these limitations. Organ engineering is a discipline that merges developmental biology, anatomy, physiology, and cellular interactions with enabling technologies such as advanced biomaterials and biofabrication to create bioartificial organs that recapitulate native organs in vivo. There have been numerous developments in bioengineering of whole organs over the past two decades. Key technological advancements include (1) methods of whole organ decellularization and recellularization, (2) three-dimensional bioprinting, (3) advanced stem cell technologies, and (4) the ability to genetically modify tissues and cells. These advancements give hope that organ engineering will become a commercial reality in the next decade. In this review article, we describe the foundational principles of whole organ engineering, discuss key technological advances, and provide an overview of current limitations and future directions.
Collapse
|
34
|
Guo J, Huebsch N. Modeling the Response of Heart Muscle to Mechanical Stimulation In Vitro. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43152-020-00007-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Mandrycky CJ, Williams NP, Batalov I, El-Nachef D, de Bakker BS, Davis J, Kim DH, DeForest CA, Zheng Y, Stevens KR, Sniadecki NJ. Engineering Heart Morphogenesis. Trends Biotechnol 2020; 38:835-845. [PMID: 32673587 DOI: 10.1016/j.tibtech.2020.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
Recent advances in stem cell biology and tissue engineering have laid the groundwork for building complex tissues in a dish. We propose that these technologies are ready for a new challenge: recapitulating cardiac morphogenesis in vitro. In development, the heart transforms from a simple linear tube to a four-chambered organ through a complex process called looping. Here, we re-examine heart tube looping through the lens of an engineer and argue that the linear heart tube is an advantageous starting point for tissue engineering. We summarize the structures, signaling pathways, and stresses in the looping heart, and evaluate approaches that could be used to build a linear heart tube and guide it through the process of looping.
Collapse
Affiliation(s)
- Christian J Mandrycky
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Nisa P Williams
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Ivan Batalov
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Danny El-Nachef
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Bernadette S de Bakker
- Clinical Anatomy and Embryology, Department of Medical Biology, AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer Davis
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Deok-Ho Kim
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine/Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Cole A DeForest
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Ying Zheng
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kelly R Stevens
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Nathan J Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
36
|
Mao M, He J, Li Z, Han K, Li D. Multi-directional cellular alignment in 3D guided by electrohydrodynamically-printed microlattices. Acta Biomater 2020; 101:141-151. [PMID: 31669696 DOI: 10.1016/j.actbio.2019.10.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 01/30/2023]
Abstract
Recapitulating aligned cellular architectures of native tissues in vitro is important to engineer artificial tissue analogs with desired biological functions. Here a novel strategy is presented to direct three-dimensional (3D) cellular alignment by embedding cell/collagen hydrogel into the predefined electrohydrodynamically-printed microlattices. The cell/collagen hydrogel, originally filled within the printed microlattices uniformly, was found to gradually develop into densely-populated and highly-aligned bands along the longitudinal direction of the printed microlattices. The cellular alignment was highly dependent on the height, spacing and orientation of the microlattices. The presented method was applicable to multiple cell types including primary cardiomyocytes and the gaps formed between the aligned bands and the lateral walls of the microlattice facilitated the subsequent seeding and rapid alignment of other cell types which enables to engineer anisotropic multicellular tissue constructs. The engineered cardiac patches expressed mature cardiomyocyte-specific phenotypes and exhibited synchronous contractive activities. Multilayer cellular alignment with varied orientation in 3D collagen hydrogel was successfully achieved by using electrohydrodynamically-printed microlattices with layer-specific orientations. This exploration offers a promising way to engineer complex 3D tissue constructs with predefined cellular alignments. STATEMENT OF SIGNIFICANCE: Fabrication of biomimetic highly-aligned complex cellular architectures has a great significance to recapitulate the unique mechanical and physiological functions of the engineered tissues (e.g., heart tissue, neuron, muscle). Here, we introduced a novel strategy to direct 3D cellular alignment by embedding cell/collagen hydrogel into the predefined electrohydrodynamically-printed microlattices without any external stimuli. The microscopical study of the dynamic alignment process of cells and collagen fibers contributed to exploring the mechanism of autonomous formation of highly-aligned cellular bands. Multilayer cellular alignment with varied orientation in 3D collagen hydrogel was successfully achieved by using the microlattices with layer-specific orientations, which showed a promising way to engineer complex 3D tissue constructs with predefined cellular alignments.
Collapse
|
37
|
Garoffolo G, Pesce M. Mechanotransduction in the Cardiovascular System: From Developmental Origins to Homeostasis and Pathology. Cells 2019; 8:cells8121607. [PMID: 31835742 PMCID: PMC6953076 DOI: 10.3390/cells8121607] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
With the term ‘mechanotransduction’, it is intended the ability of cells to sense and respond to mechanical forces by activating intracellular signal transduction pathways and the relative phenotypic adaptation. While a known role of mechanical stimuli has been acknowledged for developmental biology processes and morphogenesis in various organs, the response of cells to mechanical cues is now also emerging as a major pathophysiology determinant. Cells of the cardiovascular system are typically exposed to a variety of mechanical stimuli ranging from compression to strain and flow (shear) stress. In addition, these cells can also translate subtle changes in biophysical characteristics of the surrounding matrix, such as the stiffness, into intracellular activation cascades with consequent evolution toward pro-inflammatory/pro-fibrotic phenotypes. Since cellular mechanotransduction has a potential readout on long-lasting modifications of the chromatin, exposure of the cells to mechanically altered environments may have similar persisting consequences to those of metabolic dysfunctions or chronic inflammation. In the present review, we highlight the roles of mechanical forces on the control of cardiovascular formation during embryogenesis, and in the development and pathogenesis of the cardiovascular system.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, 4, I-20138 Milan, Italy;
- PhD Program in Translational and Molecular Medicine DIMET, Università di Milano - Bicocca, 20126 Milan, Italy
- Correspondence:
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, 4, I-20138 Milan, Italy;
| |
Collapse
|
38
|
Ribeiro AJS, Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Brock M, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Drug-Induced Inotropic Effects in Early Drug Development. Part 2: Designing and Fabricating Microsystems for Assaying Cardiac Contractility With Physiological Relevance Using Human iPSC-Cardiomyocytes. Front Pharmacol 2019; 10:934. [PMID: 31555128 PMCID: PMC6727630 DOI: 10.3389/fphar.2019.00934] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
Contractility of the myocardium engines the pumping function of the heart and is enabled by the collective contractile activity of its muscle cells: cardiomyocytes. The effects of drugs on the contractility of human cardiomyocytes in vitro can provide mechanistic insight that can support the prediction of clinical cardiac drug effects early in drug development. Cardiomyocytes differentiated from human-induced pluripotent stem cells have high potential for overcoming the current limitations of contractility assays because they attach easily to extracellular materials and last long in culture, while having human- and patient-specific properties. Under these conditions, contractility measurements can be non-destructive and minimally invasive, which allow assaying sub-chronic effects of drugs. For this purpose, the function of cardiomyocytes in vitro must reflect physiological settings, which is not observed in cultured cardiomyocytes derived from induced pluripotent stem cells because of the fetal-like properties of their contractile machinery. Primary cardiomyocytes or tissues of human origin fully represent physiological cellular properties, but are not easily available, do not last long in culture, and do not attach easily to force sensors or mechanical actuators. Microengineered cellular systems with a more mature contractile function have been developed in the last 5 years to overcome this limitation of stem cell-derived cardiomyocytes, while simultaneously measuring contractile endpoints with integrated force sensors/actuators and image-based techniques. Known effects of engineered microenvironments on the maturity of cardiomyocyte contractility have also been discovered in the development of these systems. Based on these discoveries, we review here design criteria of microengineered platforms of cardiomyocytes derived from pluripotent stem cells for measuring contractility with higher physiological relevance. These criteria involve the use of electromechanical, chemical and morphological cues, co-culture of different cell types, and three-dimensional cellular microenvironments. We further discuss the use and the current challenges for developing and improving these novel technologies for predicting clinical effects of drugs based on contractility measurements with cardiomyocytes differentiated from induced pluripotent stem cells. Future research should establish contexts of use in drug development for novel contractility assays with stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
39
|
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: 1.4] [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
|
40
|
Zhao G, Bao X, Huang G, Xu F, Zhang X. Differential Effects of Directional Cyclic Stretching on the Functionalities of Engineered Cardiac Tissues. ACS APPLIED BIO MATERIALS 2019; 2:3508-3519. [DOI: 10.1021/acsabm.9b00414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Guoxu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- School of Material Science and Chemical Engineering, Xi’an Technological University, Xi’an 710021, People’s Republic of China
| | - Xuejiao Bao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
| |
Collapse
|
41
|
Rodriguez ML, Beussman KM, Chun KS, Walzer MS, Yang X, Murry CE, Sniadecki NJ. Substrate Stiffness, Cell Anisotropy, and Cell-Cell Contact Contribute to Enhanced Structural and Calcium Handling Properties of Human Embryonic Stem Cell-Derived Cardiomyocytes. ACS Biomater Sci Eng 2019; 5:3876-3888. [PMID: 33438427 DOI: 10.1021/acsbiomaterials.8b01256] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can be utilized to understand the mechanisms underlying the development and progression of heart disease, as well as to develop better interventions and treatments for this disease. However, these cells are structurally and functionally immature, which undermines some of their adequacy in modeling adult heart tissue. Previous studies with immature cardiomyocytes have shown that altering substrate stiffness, cell anisotropy, and/or cell-cell contact can enhance the contractile and structural maturation of hPSC-CMs. In this study, the structural and calcium handling properties of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were enhanced by exposure to a downselected combination of these three maturation stimuli. First, hESC-CMs were seeded onto substrates composed of two commercial formulations of polydimethylsiloxane (PDMS), Sylgard 184 and Sylgard 527, whose stiffness ranged from 5 kPa to 101 kPa. Upon analyzing the morphological and calcium transient properties of these cells, it was concluded that a 21 kPa substrate yielded cells with the highest degree of maturation. Next, these PDMS substrates were microcontact-printed with laminin to force the cultured cells into rod-shaped geometries using line patterns that were 12, 18, or 24 μm in width. We found that cells on the 18 and 24 μm pattern widths had structural and functional properties that were superior to those on the 12 μm pattern. The hESC-CMs were then seeded onto these line-stamped surfaces at a density of 500 000 cells per 25-mm-diameter substrate, to enable the formation of cell-cell contacts at their distal ends. We discovered that this combination of culture conditions resulted in cells that were more structurally and functionally mature than those that were only exposed to one or two stimuli. Our results suggest that downselecting a combination of mechanobiological stimuli could prove to be an effective means of maturing hPSC-CMs in vitro.
Collapse
Affiliation(s)
- Marita L Rodriguez
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Kevin M Beussman
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Katherine S Chun
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Melissa S Walzer
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States
| | - Xiulan Yang
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Charles E Murry
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States.,Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, United States
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
42
|
Marin TM, de Carvalho Indolfo N, Rocco SA, Basei FL, de Carvalho M, de Almeida Gonçalves K, Pagani E. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact 2019; 299:59-76. [DOI: 10.1016/j.cbi.2018.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 11/10/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022]
|
43
|
Herrmann FEM, Lehner A, Koenig F, Hollweck T, Fano C, Dauner M, Eissner G, Hagl C, Akra B. A feasibility study of a multimodal stimulation bioreactor for the conditioning of stem cell seeded cardiac patches via electrical impulses and pulsatile perfusion. Biomed Mater Eng 2018; 30:37-48. [PMID: 30530957 DOI: 10.3233/bme-181031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/OBJECTIVE Ischemic heart disease is a major cause of mortality worldwide. Myocardial tissue engineering aims to create transplantable units of myocardium for the treatment of myocardial necrosis caused by ischemic heart disease - bioreactors are used to condition these bioartificial tissues before application. METHODS Our group developed a multimodal bioreactor consisting of a linear drive motor for pulsatile flow generation (500 ml/min) and an external pacemaker for electrical stimulation (10 mA, 3 V at 60 Hz) using LinMot-Talk Software to synchronize these modes of stimulation. Polyurethane scaffolds were seeded with 0.750 × 106 mesenchymal stem cells from umbilical cord tissue per cm2 and stimulated in our system for 72 h, then evaluated. RESULTS After conditioning histology showed that the patches consisted of a cell multilayer surviving stimulation without major damage by the multimodal stimulation, scanning electron microscopy showed a confluent cell layer with no cell-cell interspaces visible. No cell viability issues could be identified via Syto9-Propidium Iodide staining. CONCLUSIONS This bioreactor allows mechanical stimulation via pulsatile flow and electrical stimulation through a pacemaker. Our stem cell-polyurethane constructs displayed survival after conditioning. This system shows feasibility in preliminary tests.
Collapse
Affiliation(s)
- Florian Ernst Martin Herrmann
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| | - Anja Lehner
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany.,Department of Pediatric Cardiology and Pediatric Intensive Care, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| | - Fabian Koenig
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| | - Trixi Hollweck
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| | - Cornelia Fano
- German Institutes of Textile and Fiber Research (DITF), Denkendorf, Germany
| | - Martin Dauner
- German Institutes of Textile and Fiber Research (DITF), Denkendorf, Germany
| | - Guenther Eissner
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Christian Hagl
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| | - Bassil Akra
- Department of Cardiac Surgery, Laboratory for Tissue Engineering, Grosshadern Medical Centre, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
44
|
Mannhardt I, Warncke C, Trieu HK, Müller J, Eschenhagen T. Piezo-bending actuators for isometric or auxotonic contraction analysis of engineered heart tissue. J Tissue Eng Regen Med 2018; 13:3-11. [PMID: 30334614 DOI: 10.1002/term.2755] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/28/2018] [Accepted: 09/24/2018] [Indexed: 01/02/2023]
Abstract
Engineered heart tissue (EHT) has proven as valuable tool for disease modelling, drug safety screening, and cardiac repair. Especially in combination with the stem cell technology, these in vitro models of the human heart have generated interest not only of basic cardiovascular researchers but also of regulatory authorities responsible for drug safety. A main limitation of 3D-based assays for evaluating cardiotoxicity is their limited throughput. We integrated piezo-bending actuators in a 24-well system for the generation of strip-like rat and human EHT attached to hollow, elastic silicone posts. Muscle contractions of EHTs induced a measurable electrical current in the piezo-bending actuators that could be analysed for contraction amplitude, frequency, and contraction and relaxation kinetics. Compared with the standard video-optical analysis of contractile activity, the new system allows for (a) the analysis of several tissues in parallel, (b) switching between auxotonic and isometric contractions by inserting a stiff metal post in the silicone post opposing the piezo actuator, (c) continuous measurement over days with low data volume (megabyte), (d) automated measurement without the necessity of adjustment of tissue position for video-optical analysis, (e) reduced complexity and costs, (f) high sensitivity of contraction detection, (g) calculation of absolute contraction force, and (h) suitability for variable tissue geometries. The new set-up for contraction analysis based on piezo-bending actuators is a promising new method for the parallel screening of EHT for pharmacological drug effects and other applications of muscle tissue engineering (e.g., skeletal muscle engineering or cardiac repair).
Collapse
Affiliation(s)
- Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Warncke
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Hoc Khiem Trieu
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Jörg Müller
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
45
|
Greenberg MJ, Daily NJ, Wang A, Conway MK, Wakatsuki T. Genetic and Tissue Engineering Approaches to Modeling the Mechanics of Human Heart Failure for Drug Discovery. Front Cardiovasc Med 2018; 5:120. [PMID: 30283789 PMCID: PMC6156537 DOI: 10.3389/fcvm.2018.00120] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022] Open
Abstract
Heart failure is the leading cause of death in the western world and as such, there is a great need for new therapies. Heart failure has a variable presentation in patients and a complex etiology; however, it is fundamentally a condition that affects the mechanics of cardiac contraction, preventing the heart from generating sufficient cardiac output under normal operating pressures. One of the major issues hindering the development of new therapies has been difficulties in developing appropriate in vitro model systems of human heart failure that recapitulate the essential changes in cardiac mechanics seen in the disease. Recent advances in stem cell technologies, genetic engineering, and tissue engineering have the potential to revolutionize our ability to model and study heart failure in vitro. Here, we review how these technologies are being applied to develop personalized models of heart failure and discover novel therapeutics.
Collapse
Affiliation(s)
- Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, United States
| | | | - Ann Wang
- InvivoSciences Inc., Madison, WI, United States
| | | | | |
Collapse
|
46
|
Jara Avaca M, Gruh I. Bioengineered Cardiac Tissue Based on Human Stem Cells for Clinical Application. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:117-146. [PMID: 29218360 DOI: 10.1007/10_2017_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Engineered cardiac tissue might enable novel therapeutic strategies for the human heart in a number of acquired and congenital diseases. With recent advances in stem cell technologies, namely the availability of pluripotent stem cells, the generation of potentially autologous tissue grafts has become a realistic option. Nevertheless, a number of limitations still have to be addressed before clinical application of engineered cardiac tissue based on human stem cells can be realized. We summarize current progress and pending challenges regarding the optimal cell source, cardiomyogenic lineage specification, purification, safety of genetic cell engineering, and genomic stability. Cardiac cells should be combined with clinical grade scaffold materials for generation of functional myocardial tissue in vitro. Scale-up to clinically relevant dimensions is mandatory, and tissue vascularization is most probably required both for preclinical in vivo testing in suitable large animal models and for clinical application. Graphical Abstract.
Collapse
Affiliation(s)
- Monica Jara Avaca
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Vascular and Transplantation Surgery (HTTG), Hannover Medical School (MHH) & Cluster of Excellence REBIRTH, Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Vascular and Transplantation Surgery (HTTG), Hannover Medical School (MHH) & Cluster of Excellence REBIRTH, Hannover, Germany.
| |
Collapse
|
47
|
Debbi L, Drori S, Tzlil S. The Influence of the Timing of Cyclic Load Application on Cardiac Cell Contraction. Front Physiol 2018; 9:917. [PMID: 30072912 PMCID: PMC6058596 DOI: 10.3389/fphys.2018.00917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/22/2018] [Indexed: 02/01/2023] Open
Abstract
Cardiac cells are subjected to mechanical load during each heart-beat. Normal heart load is essential for physiological development and cardiac function. At the same time, excessive load can induce pathologies such as cardiac hypertrophy. While the forces working on the heart as an organ are well-understood, information regarding stretch response at the cellular level is limited. Since cardiac stretch-response depends on the amplitude and pattern of the applied load as well as its timing during the beating cycle, the directionality of load application and its phase relative to action potential generation must be controlled precisely. Here, we design a new experimental setup, which enables high-resolution fluorescence imaging of cultured cardiac cells under cyclic uniaxial mechanical load and electrical stimulation. Cyclic stretch was applied in different phases relative to the electrical stimulus and the effect on cardiac cell beating was monitored. The results show a clear phase-dependent response and provide insight into cardiac response to excessive loading conditions.
Collapse
Affiliation(s)
- Lior Debbi
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Stavit Drori
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shelly Tzlil
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
48
|
Abstract
Some of the most significant leaps in the history of modern civilization-the development of article in China, the steam engine, which led to the European industrial revolution, and the era of computers-have occurred when science converged with engineering. Recently, the convergence of human pluripotent stem cell technology with biomaterials and bioengineering have launched a new medical innovation: functional human engineered tissue, which promises to revolutionize the treatment of failing organs including most critically, the heart. This compendium covers recent, state-of-the-art developments in the fields of cardiovascular tissue engineering, as well as the needs and challenges associated with the clinical use of these technologies. We have not attempted to provide an exhaustive review in stem cell biology and cardiac cell therapy; many other important and influential reports are certainly merit but already been discussed in several recent reviews. Our scope is limited to the engineered tissues that have been fabricated to repair or replace components of the heart (eg, valves, vessels, contractile tissue) that have been functionally compromised by diseases or developmental abnormalities. In particular, we have focused on using an engineered myocardial tissue to mitigate deficiencies in contractile function.
Collapse
Affiliation(s)
- Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Wuqiang Zhu
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada (M.R.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering and Department of Medicine, Columbia University, New York, NY (G.V.-N.)
| |
Collapse
|
49
|
Leonard A, Bertero A, Powers JD, Beussman KM, Bhandari S, Regnier M, Murry CE, Sniadecki NJ. Afterload promotes maturation of human induced pluripotent stem cell derived cardiomyocytes in engineered heart tissues. J Mol Cell Cardiol 2018; 118:147-158. [PMID: 29604261 DOI: 10.1016/j.yjmcc.2018.03.016] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/07/2018] [Accepted: 03/26/2018] [Indexed: 12/30/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) grown in engineered heart tissue (EHT) can be used for drug screening, disease modeling, and heart repair. However, the immaturity of hiPSC-CMs currently limits their use. Because mechanical loading increases during development and facilitates cardiac maturation, we hypothesized that afterload would promote maturation of EHTs. To test this we developed a system in which EHTs are suspended between a rigid post and a flexible one, whose resistance to contraction can be modulated by applying braces of varying length. These braces allow us to adjust afterload conditions over two orders of magnitude by increasing the flexible post resistance from 0.09 up to 9.2 μN/μm. After three weeks in culture, optical tracking of post deflections revealed that auxotonic twitch forces increased in correlation with the degree of afterload, whereas twitch velocities decreased with afterload. Consequently, the power and work of the EHTs were maximal under intermediate afterloads. When studied isometrically, the inotropy of EHTs increased with afterload up to an intermediate resistance (0.45 μN/μm) and then plateaued. Applied afterload increased sarcomere length, cardiomyocyte area and elongation, which are hallmarks of maturation. Furthermore, progressively increasing the level of afterload led to improved calcium handling, increased expression of several key markers of cardiac maturation, including a shift from fetal to adult ventricular myosin heavy chain isoforms. However, at the highest afterload condition, markers of pathological hypertrophy and fibrosis were also upregulated, although the bulk tissue stiffness remained the same for all levels of applied afterload tested. Together, our results indicate that application of moderate afterloads can substantially improve the maturation of hiPSC-CMs in EHTs, while high afterload conditions may mimic certain aspects of human cardiac pathology resulting from elevated mechanical overload.
Collapse
Affiliation(s)
- Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA
| | - Alessandro Bertero
- Department of Pathology, University of Washington, Seattle 98109, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA
| | - Joseph D Powers
- Department of Bioengineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA
| | - Kevin M Beussman
- Department of Mechanical Engineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA
| | - Shiv Bhandari
- Department of Medicine, University of Washington, Seattle 98195, WA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA
| | - Charles E Murry
- Department of Pathology, University of Washington, Seattle 98109, WA, USA; Department of Bioengineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA; Department of Medicine, University of Washington, Seattle 98195, WA, USA; Division of Cardiology, University of Washington, Seattle 98195, WA, USA.
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle 98107, WA, USA; Department of Bioengineering, University of Washington, Seattle 98107, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle 98109, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle 98109, WA, USA.
| |
Collapse
|
50
|
Mills RJ, Voges HK, Porrello ER, Hudson JE. Disease modeling and functional screening using engineered heart tissue. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|