1
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [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: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
2
|
El-Nashar H, Sabry M, Tseng YT, Francis N, Latif N, Parker KH, Moore JE, Yacoub MH. Multiscale structure and function of the aortic valve apparatus. Physiol Rev 2024; 104:1487-1532. [PMID: 37732828 PMCID: PMC11495199 DOI: 10.1152/physrev.00038.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023] Open
Abstract
Whereas studying the aortic valve in isolation has facilitated the development of life-saving procedures and technologies, the dynamic interplay of the aortic valve and its surrounding structures is vital to preserving their function across the wide range of conditions encountered in an active lifestyle. Our view is that these structures should be viewed as an integrated functional unit, here referred to as the aortic valve apparatus (AVA). The coupling of the aortic valve and root, left ventricular outflow tract, and blood circulation is crucial for AVA's functions: unidirectional flow out of the left ventricle, coronary perfusion, reservoir function, and support of left ventricular function. In this review, we explore the multiscale biological and physical phenomena that underlie the simultaneous fulfillment of these functions. A brief overview of the tools used to investigate the AVA, such as medical imaging modalities, experimental methods, and computational modeling, specifically fluid-structure interaction (FSI) simulations, is included. Some pathologies affecting the AVA are explored, and insights are provided on treatments and interventions that aim to maintain quality of life. The concepts explained in this article support the idea of AVA being an integrated functional unit and help identify unanswered research questions. Incorporating phenomena through the molecular, micro, meso, and whole tissue scales is crucial for understanding the sophisticated normal functions and diseases of the AVA.
Collapse
Affiliation(s)
- Hussam El-Nashar
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Malak Sabry
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Biomedical Engineering, King's College London, London, United Kingdom
| | - Yuan-Tsan Tseng
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nadine Francis
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Najma Latif
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kim H Parker
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - James E Moore
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Magdi H Yacoub
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Bouhrira N, Vite A, Margulies KB. Distinct cytoskeletal regulators of mechanical memory in cardiac fibroblasts and cardiomyocytes. Basic Res Cardiol 2024; 119:277-289. [PMID: 38349539 DOI: 10.1007/s00395-023-01030-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 04/12/2024]
Abstract
Recognizing that cells "feel" and respond to their mechanical environment, recent studies demonstrate that many cells exhibit a phenomenon of "mechanical memory" in which features induced by prior mechanical cues persist after the mechanical stimulus has ceased. While there is a general recognition that different cell types exhibit different responses to changes in extracellular matrix stiffening, the phenomenon of mechanical memory within myocardial cell types has received little attention to date. To probe the dynamics of mechanical memory in cardiac fibroblasts (CFs) and cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs), we employed a magnetorheological elastomer (MRE) cell culture substrate with tunable and reversible stiffness spanning the range from normal to diseased myocardium. In CFs, using increased cell area and increases in α-smooth muscle actin as markers of cellular responses to matrix stiffening, we found that induction of mechanical memory required seven days of stiff priming. Both induction and maintenance of persistent CF activation were blocked with the F-actin inhibitor cytochalasin D, while inhibitors of microtubule detyrosination had no impact on CFs. In iPSC-CMs, mechanical memory was invoked after only 24 h of stiff priming. Moreover, mechanical memory induction and maintenance were microtubule-dependent in CMs with no dependence on F-actin. Overall, these results identify the distinct temporal dynamics of mechanical memory in CFs and iPSC-CMs with different cytoskeletal mediators responsible for inducing and maintaining the stiffness-activated phenotype. Due to its flexibility, this model is broadly applicable to future studies interrogating mechanotransduction and mechanical memory in the heart and might inform strategies for attenuating the impact of load-induced pathology and excess myocardial stiffness.
Collapse
Affiliation(s)
- Nesrine Bouhrira
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Alexia Vite
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Kenneth B Margulies
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Chen X, Liu S, Han M, Long M, Li T, Hu L, Wang L, Huang W, Wu Y. Engineering Cardiac Tissue for Advanced Heart-On-A-Chip Platforms. Adv Healthc Mater 2024; 13:e2301338. [PMID: 37471526 DOI: 10.1002/adhm.202301338] [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] [Received: 04/26/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Cardiovascular disease is a major cause of mortality worldwide, and current preclinical models including traditional animal models and 2D cell culture models have limitations in replicating human native heart physiology and response to drugs. Heart-on-a-chip (HoC) technology offers a promising solution by combining the advantages of cardiac tissue engineering and microfluidics to create in vitro 3D cardiac models, which can mimic key aspects of human microphysiological systems and provide controllable microenvironments. Herein, recent advances in HoC technologies are introduced, including engineered cardiac microtissue construction in vitro, microfluidic chip fabrication, microenvironmental stimulation, and real-time feedback systems. The development of cardiac tissue engineering methods is focused for 3D microtissue preparation, advanced strategies for HoC fabrication, and current applications of these platforms. Major challenges in HoC fabrication are discussed and the perspective on the potential for these platforms is provided to advance research and clinical applications.
Collapse
Affiliation(s)
- Xinyi Chen
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sitian Liu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingying Han
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Meng Long
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Li
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lanlan Hu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ling Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
5
|
Brown SJ, Šoltić D, Synowsky SA, Shirran SL, Chilcott E, Shorrock HK, Gillingwater TH, Yáñez-Muñoz RJ, Schneider B, Bowerman M, Fuller HR. AAV9-mediated SMN gene therapy rescues cardiac desmin but not lamin A/C and elastin dysregulation in Smn2B/- spinal muscular atrophy mice. Hum Mol Genet 2023; 32:2950-2965. [PMID: 37498175 PMCID: PMC10549791 DOI: 10.1093/hmg/ddad121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
Structural, functional and molecular cardiac defects have been reported in spinal muscular atrophy (SMA) patients and mouse models. Previous quantitative proteomics analyses demonstrated widespread molecular defects in the severe Taiwanese SMA mouse model. Whether such changes are conserved across different mouse models, including less severe forms of the disease, has yet to be established. Here, using the same high-resolution proteomics approach in the less-severe Smn2B/- SMA mouse model, 277 proteins were found to be differentially abundant at a symptomatic timepoint (post-natal day (P) 18), 50 of which were similarly dysregulated in severe Taiwanese SMA mice. Bioinformatics analysis linked many of the differentially abundant proteins to cardiovascular development and function, with intermediate filaments highlighted as an enriched cellular compartment in both datasets. Lamin A/C was increased in the cardiac tissue, whereas another intermediate filament protein, desmin, was reduced. The extracellular matrix (ECM) protein, elastin, was also robustly decreased in the heart of Smn2B/- mice. AAV9-SMN1-mediated gene therapy rectified low levels of survival motor neuron protein and restored desmin levels in heart tissues of Smn2B/- mice. In contrast, AAV9-SMN1 therapy failed to correct lamin A/C or elastin levels. Intermediate filament proteins and the ECM have key roles in cardiac function and their dysregulation may explain cardiac impairment in SMA, especially since mutations in genes encoding these proteins cause other diseases with cardiac aberration. Cardiac pathology may need to be considered in the long-term care of SMA patients, as it is unclear whether currently available treatments can fully rescue peripheral pathology in SMA.
Collapse
Affiliation(s)
- Sharon J Brown
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Darija Šoltić
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Silvia A Synowsky
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Ellie Chilcott
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Hannah K Shorrock
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Bernard Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Melissa Bowerman
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| |
Collapse
|
6
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
7
|
El-Husseiny HM, Mady EA, El-Dakroury WA, Doghish AS, Tanaka R. Stimuli-responsive hydrogels: smart state of-the-art platforms for cardiac tissue engineering. Front Bioeng Biotechnol 2023; 11:1174075. [PMID: 37449088 PMCID: PMC10337592 DOI: 10.3389/fbioe.2023.1174075] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Biomedicine and tissue regeneration have made significant advancements recently, positively affecting the whole healthcare spectrum. This opened the way for them to develop their applications for revitalizing damaged tissues. Thus, their functionality will be restored. Cardiac tissue engineering (CTE) using curative procedures that combine biomolecules, biomimetic scaffolds, and cells plays a critical part in this path. Stimuli-responsive hydrogels (SRHs) are excellent three-dimensional (3D) biomaterials for tissue engineering (TE) and various biomedical applications. They can mimic the intrinsic tissues' physicochemical, mechanical, and biological characteristics in a variety of ways. They also provide for 3D setup, adequate aqueous conditions, and the mechanical consistency required for cell development. Furthermore, they function as competent delivery platforms for various biomolecules. Many natural and synthetic polymers were used to fabricate these intelligent platforms with innovative enhanced features and specialized capabilities that are appropriate for CTE applications. In the present review, different strategies employed for CTE were outlined. The light was shed on the limitations of the use of conventional hydrogels in CTE. Moreover, diverse types of SRHs, their characteristics, assembly and exploitation for CTE were discussed. To summarize, recent development in the construction of SRHs increases their potential to operate as intelligent, sophisticated systems in the reconstruction of degenerated cardiac tissues.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Walaa A. El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Egypt
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
8
|
Sajid N. Topography and mechanical measurements of primary Schwann cells and neuronal cells with atomic force microscope for understanding and controlling nerve growth. Micron 2023; 167:103427. [PMID: 36805164 DOI: 10.1016/j.micron.2023.103427] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023]
Abstract
Peripheral nerve injuries require a piece of substantial information for a satisfactory treatment. The prior peripheral nerve injury knowledge, can improve nerve repair, and its growth at molecular and cellular level. In this study, we employed an atomic force microscope (AFM) to investigate the topography and mechanical properties of the primary Schwann cells and neuronal cells. Tapping mode images and contact points force-volume maps provide the cells topography. Two different probes were used to acquire the micro and nanomechanical properties of the primary Schwann cells, NG-108-15 neuronal cells, and growth cones. Moreover, the sharp probe was only used to investigate neurites nanomechanics. A significant difference in the elastic moduli found between primary Schwann cells, and neuronal cells, with both probes, with consistent results. The elastic moduli of the growth cones were found higher, than the neuronal cells and primary Schwann cells, with both probes. Furthermore, the modulus variations were also found between neurites. These results have significant implications for a better understanding of the peripheral nerve system (PNS) in terms of defining the optimal pattern surface and nerve guidance conduits.
Collapse
Affiliation(s)
- Nusrat Sajid
- Department of Physics, COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan.
| |
Collapse
|
9
|
Williams DF. The plasticity of biocompatibility. Biomaterials 2023; 296:122077. [PMID: 36907003 DOI: 10.1016/j.biomaterials.2023.122077] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
Biocompatibility concerns the phenomena that occur within the interactions between biomaterials and human patients, which ultimately control the performance of many facets of medical technology. It involves aspects of materials science, many different forms of engineering and nanotechnology, chemistry, biophysics, molecular and cellular biology, immunology, pathology and a myriad of clinical applications. It is not surprising that an overarching framework of mechanisms of biocompatibility has been difficult to elucidate and validate. This essay discusses one fundamental reason for this; we have tended to consider biocompatibility pathways as essentially linear sequences of events which follow well-understood processes of materials science and biology. The reality, however, is that the pathways may involve a great deal of plasticity, in which many additional idiosyncratic factors, including those of genetic, epigenetic and viral origin, exert influence, as do complex mechanical, physical and pharmacological variables. Plasticity is an inherent core feature of the performance of synthetic materials; here we follow the more recent biological applications of plasticity concepts into the sphere of biocompatibility pathways. A straightforward linear pathway may result in successful outcomes for many patients; we may describe this in terms of classic biocompatibility pathways. In other situations, which usually command much more attention because of their unsuccessful outcomes, these plasticity-driven processes follow alternative biocompatibility pathways; often, the variability in outcomes with identical technologies is due to biological plasticity rather than material or device deficiency.
Collapse
Affiliation(s)
- David F Williams
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
10
|
Carotenuto F, Politi S, Ul Haq A, De Matteis F, Tamburri E, Terranova ML, Teodori L, Pasquo A, Di Nardo P. From Soft to Hard Biomimetic Materials: Tuning Micro/Nano-Architecture of Scaffolds for Tissue Regeneration. MICROMACHINES 2022; 13:mi13050780. [PMID: 35630247 PMCID: PMC9144100 DOI: 10.3390/mi13050780] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/30/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022]
Abstract
Failure of tissues and organs resulting from degenerative diseases or trauma has caused huge economic and health concerns around the world. Tissue engineering represents the only possibility to revert this scenario owing to its potential to regenerate or replace damaged tissues and organs. In a regeneration strategy, biomaterials play a key role promoting new tissue formation by providing adequate space for cell accommodation and appropriate biochemical and biophysical cues to support cell proliferation and differentiation. Among other physical cues, the architectural features of the biomaterial as a kind of instructive stimuli can influence cellular behaviors and guide cells towards a specific tissue organization. Thus, the optimization of biomaterial micro/nano architecture, through different manufacturing techniques, is a crucial strategy for a successful regenerative therapy. Over the last decades, many micro/nanostructured biomaterials have been developed to mimic the defined structure of ECM of various soft and hard tissues. This review intends to provide an overview of the relevant studies on micro/nanostructured scaffolds created for soft and hard tissue regeneration and highlights their biological effects, with a particular focus on striated muscle, cartilage, and bone tissue engineering applications.
Collapse
Affiliation(s)
- Felicia Carotenuto
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Correspondence: (F.C.); (P.D.N.)
| | - Sara Politi
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Arsalan Ul Haq
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
| | - Fabio De Matteis
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento Ingegneria Industriale, Università Degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Emanuela Tamburri
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Maria Letizia Terranova
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Laura Teodori
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
| | - Alessandra Pasquo
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
| | - Paolo Di Nardo
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Correspondence: (F.C.); (P.D.N.)
| |
Collapse
|
11
|
Truncation of the N-terminus of cardiac troponin I initiates adaptive remodeling of the myocardial proteosome via phosphorylation of mechano-sensitive signaling pathways. Mol Cell Biochem 2022; 477:1803-1815. [PMID: 35316461 DOI: 10.1007/s11010-022-04414-3] [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: 10/15/2021] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The cardiac isoform of troponin I has a unique N-terminal extension (~ 1-30 amino acids), which contributes to the modulation of cardiac contraction and relaxation. Hearts of various species including humans produce a truncated variant of cardiac troponin I (cTnI-ND) deleting the first ~ 30 amino acids as an adaption in pathophysiological conditions. In this study, we investigated the impact of cTnI-ND chronic expression in transgenic mouse hearts compared to wildtype (WT) controls (biological n = 8 in each group). We aimed to determine the global phosphorylation effects of cTnI-ND on the cardiac proteome, thereby determining the signaling pathways that have an impact on cardiac function. The samples were digested and isobarically labeled and equally mixed for relative quantification via nanoLC-MS/MS. The peptides were then enriched for phospho-peptides and bioinformatic analysis was done with Ingenuity Pathway Analysis (IPA). We found approximately 77% replacement of the endogenous intact cTnI with cTnI-ND in the transgenic mouse hearts with 1674 phospho-proteins and 2971 non-modified proteins. There were 73 significantly altered phospho-proteins; bioinformatic analysis identified the top canonical pathways as associated with integrin, protein kinase A, RhoA, and actin cytoskeleton signaling. Among the 73 phospho-proteins compared to controls cTnI-ND hearts demonstrated a significant decrease in paxillin and YAP1, which are known to play a role in cell mechano-sensing pathways. Our data indicate that cTnI-ND modifications in the sarcomere are sufficient to initiate changes in the phospho-signaling profile that may underly the chronic-adaptive response associated with cTnI cleavage in response to stressors by modifying mechano-sensitive signaling pathways.
Collapse
|
12
|
Balakrishnan G, Song J, Mou C, Bettinger CJ. Recent Progress in Materials Chemistry to Advance Flexible Bioelectronics in Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106787. [PMID: 34751987 PMCID: PMC8917047 DOI: 10.1002/adma.202106787] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/15/2021] [Indexed: 05/09/2023]
Abstract
Designing bioelectronic devices that seamlessly integrate with the human body is a technological pursuit of great importance. Bioelectronic medical devices that reliably and chronically interface with the body can advance neuroscience, health monitoring, diagnostics, and therapeutics. Recent major efforts focus on investigating strategies to fabricate flexible, stretchable, and soft electronic devices, and advances in materials chemistry have emerged as fundamental to the creation of the next generation of bioelectronics. This review summarizes contemporary advances and forthcoming technical challenges related to three principal components of bioelectronic devices: i) substrates and structural materials, ii) barrier and encapsulation materials, and iii) conductive materials. Through notable illustrations from the literature, integration and device fabrication strategies and associated challenges for each material class are highlighted.
Collapse
Affiliation(s)
| | - Jiwoo Song
- Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA
| | - Chenchen Mou
- Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA
| | | |
Collapse
|
13
|
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
|
14
|
Singh JP, Young JL. The cardiac nanoenvironment: form and function at the nanoscale. Biophys Rev 2021; 13:625-636. [PMID: 34765045 PMCID: PMC8555021 DOI: 10.1007/s12551-021-00834-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Mechanical forces in the cardiovascular system occur over a wide range of length scales. At the whole organ level, large scale forces drive the beating heart as a synergistic unit. On the microscale, individual cells and their surrounding extracellular matrix (ECM) exhibit dynamic reciprocity, with mechanical feedback moving bidirectionally. Finally, in the nanometer regime, molecular features of cells and the ECM show remarkable sensitivity to mechanical cues. While small, these nanoscale properties are in many cases directly responsible for the mechanosensitive signaling processes that elicit cellular outcomes. Given the inherent challenges in observing, quantifying, and reconstituting this nanoscale environment, it is not surprising that this landscape has been understudied compared to larger length scales. Here, we aim to shine light upon the cardiac nanoenvironment, which plays a crucial role in maintaining physiological homeostasis while also underlying pathological processes. Thus, we will highlight strategies aimed at (1) elucidating the nanoscale components of the cardiac matrix, and (2) designing new materials and biosystems capable of mimicking these features in vitro.
Collapse
Affiliation(s)
- Jashan P Singh
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore
| | - Jennifer L Young
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117575 Singapore, Singapore
| |
Collapse
|
15
|
Cortella LRX, Cestari IA, Lahuerta RD, Araña MC, Soldera M, Rank A, Lasagni AF, Cestari IN. Conditioning of hiPSC-derived cardiomyocytes using surface topography obtained with high throughput technology. Biomed Mater 2021; 16. [PMID: 34412045 DOI: 10.1088/1748-605x/ac1f73] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 08/19/2021] [Indexed: 12/31/2022]
Abstract
Surface functionalization of polymers aims to introduce novel properties that favor bioactive responses. We have investigated the possibility of surface functionalization of polyethylene terephthalate (PET) sheets by the combination of laser ablation with hot embossing and the application of such techniques in the field of stem cell research. We investigated the response of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to topography in the low micrometer range. HiPSC-CMs are expected to offer new therapeutic tools for myocardial replacement or regeneration after an infarct or other causes of cardiac tissue loss. However, hiPSC-CMs are phenotypically immature compared to myocytes in the adult myocardium, hampering their clinical application. We aimed to develop and test a high-throughput technique for surface structuring that would improve hiPSC-CMs structural maturation. We used laser ablation with a ps-laser source in combination with nanoimprint lithography to fabricate large areas of homogeneous micron- to submicron line-like pattern with a spatial period of 3 µm on the PET surface. We evaluated cell morphology, alignment, sarcomeric myofibrils assembly, and calcium transients to evaluate phenotypic changes associated with culturing hiPSC-CMs on functionalized PET. Surface functionalization through hot embossing was able to generate, at low cost, low micrometer features on the PET surface that influenced the hiPSC-CMs phenotype, suggesting improved structural and functional maturation. This technique may be relevant for high-throughput technologies that require conditioning of hiPSC-CMs and may be useful for the production of these cells for drug screening and disease modeling applications with lower costs.
Collapse
Affiliation(s)
- Lucas R X Cortella
- Bioengineering Department, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Enéas de Carvalho Aguiar, 44, 05403-900 São Paulo, Brazil
| | - Idágene A Cestari
- Bioengineering Department, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Enéas de Carvalho Aguiar, 44, 05403-900 São Paulo, Brazil
| | - Ricardo D Lahuerta
- Bioengineering Department, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Enéas de Carvalho Aguiar, 44, 05403-900 São Paulo, Brazil
| | - Matheus C Araña
- Bioengineering Department, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Enéas de Carvalho Aguiar, 44, 05403-900 São Paulo, Brazil
| | - Marcos Soldera
- Institute for Manufacturing Technology, Technische Universität Dresden, George-Baehr-Str. 3c, 01069 Dresden, Germany.,PROBIEN-CONICET, Dto. de Electrotecnia, Universidad Nacional del Comahue, Buenos Aires 1400, 8300 Neuquén, Argentina
| | - Andreas Rank
- Institute for Manufacturing Technology, Technische Universität Dresden, George-Baehr-Str. 3c, 01069 Dresden, Germany
| | - Andrés F Lasagni
- Institute for Manufacturing Technology, Technische Universität Dresden, George-Baehr-Str. 3c, 01069 Dresden, Germany.,Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Winterbergstr. 28, 01277 Dresden, Germany
| | - Ismar N Cestari
- Bioengineering Department, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Enéas de Carvalho Aguiar, 44, 05403-900 São Paulo, Brazil
| |
Collapse
|
16
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
17
|
Worku MG. Pluripotent and Multipotent Stem Cells and Current Therapeutic Applications: Review. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2021; 14:3-7. [PMID: 33880040 PMCID: PMC8052119 DOI: 10.2147/sccaa.s304887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022]
Abstract
There is numerous evidence for the presence of stem cells, which is important for the treatment of a wide variety of disease conditions. Stem cells have a great therapeutic effect on different degenerative diseases through the development of specialized cells. Embryonic stem (ES) cells are derived from preimplantation embryos, which have a natural karyotype. This cell has the capacity of proliferation indefinitely and undifferentiated. Stem cells are very crucial for the treatment of different chronic and degenerative diseases. For instance, stem cell clinical trials have been done for ischemic heart disease. Also, the olfactory cells for spinal cord lesions and human fetal pancreatic cells for diabetes mellitus are the other clinical importance of stem cell therapy. Extracellular matrix (ECM) and other environmental factors influence the fate and activity of stem cells.
Collapse
Affiliation(s)
- Misganaw Gebrie Worku
- Department of Human Anatomy, University of Gondar, College of Medicine and Health Science, School of Medicine, Gondar, Ethiopia
| |
Collapse
|
18
|
Abstract
ABSTRACT Congenital microtia is a severe physiological defect and is among the most common craniofacial defects. It is characterized by severe auricle dysplasia, external auditory canal atresia or stenosis, and middle ear malformation, though inner ear development is mostly normal with some hearing occurring through bone conduction. Auricular reconstruction is the only treatment for congenital microtia. In this study, the authors integrated messenger ribonucleic acid and mass spectrometry data of cartilage obtained from the affected and unaffected sides of 16 unilateral microtia patients who had undergone ear reconstruction surgery. The authors next performed functional analyses to investigate differences in the proteome of the affected and unaffected ears to elicit molecular pathways involved in microtia pathogenesis. The authors collected 16 pairs samples. Proteomic and transcriptomic analyses identified 47 genes that were differentially expressed in affected and unaffected cartilage. Integrated pathway analysis implicated the involvement of genes related to cell adhesion, extracellular matrix organization, and cell migration in disease progression. Through the integration of gene and protein expression data in human primary chondrocytes, the authors identified molecular markers of microtia progression that were replicated across independent datasets and that have translational potential.
Collapse
|
19
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
20
|
Quadri F, Soman SS, Vijayavenkataraman S. Progress in cardiovascular bioprinting. Artif Organs 2021; 45:652-664. [PMID: 33432583 DOI: 10.1111/aor.13913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/13/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease has been the leading cause of death globally for the past 15 years. Following a major cardiac disease episode, the ideal treatment would be the replacement of the damaged tissue, due to the limited regenerative capacity of cardiac tissues. However, we suffer from a chronic organ donor shortage which causes approximately 20 people to die each day waiting to receive an organ. Bioprinting of tissues and organs can potentially alleviate this burden by fabricating low cost tissue and organ replacements for cardiac patients. Clinical adoption of bioprinting in cardiovascular medicine is currently limited by the lack of systematic demonstration of its effectiveness, high costs, and the complexity of the workflow. Here, we give a concise review of progress in cardiovascular bioprinting and its components. We further discuss the challenges and future prospects of cardiovascular bioprinting in clinical applications.
Collapse
Affiliation(s)
- Faisal Quadri
- Division of Science, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Soja Saghar Soman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Sanjairaj Vijayavenkataraman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, USA
| |
Collapse
|
21
|
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
|
22
|
DePalma SJ, Davidson CD, Stis AE, Helms AS, Baker BM. Microenvironmental determinants of organized iPSC-cardiomyocyte tissues on synthetic fibrous matrices. Biomater Sci 2021; 9:93-107. [PMID: 33325920 PMCID: PMC7971708 DOI: 10.1039/d0bm01247e] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) show great potential for engineering myocardium to study cardiac disease and create regenerative therapies. However, iPSC-CMs typically possess a late embryonic stage phenotype, with cells failing to exhibit markers of mature adult tissue. This is due in part to insufficient knowledge and control of microenvironmental cues required to facilitate the organization and maturation of iPSC-CMs. Here, we employed a cell-adhesive, mechanically tunable synthetic fibrous extracellular matrix (ECM) consisting of electrospun dextran vinyl sulfone (DVS) fibers and examined how biochemical, architectural, and mechanical properties of the ECM impact iPSC-CM tissue assembly and subsequent function. Exploring a multidimensional parameter space spanning cell-adhesive ligand, seeding density, fiber alignment, and stiffness, we found that fibronectin-functionalized DVS matrices composed of highly aligned fibers with low stiffness optimally promoted the organization of functional iPSC-CM tissues. Tissues generated on these matrices demonstrated improved calcium handling and increased end-to-end localization of N-cadherin as compared to micropatterned fibronectin lines or fibronectin-coated glass. Furthermore, DVS matrices supported long-term culture (45 days) of iPSC-CMs; N-cadherin end-to-end localization and connexin43 expression both increased as a function of time in culture. In sum, these findings demonstrate the importance of recapitulating the fibrous myocardial ECM in engineering structurally organized and functional iPSC-CM tissues.
Collapse
Affiliation(s)
- Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
23
|
Demolder A, von Kodolitsch Y, Muiño-Mosquera L, De Backer J. Myocardial Function, Heart Failure and Arrhythmia in Marfan Syndrome: A Systematic Literature Review. Diagnostics (Basel) 2020; 10:E751. [PMID: 32992882 PMCID: PMC7599866 DOI: 10.3390/diagnostics10100751] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 11/17/2022] Open
Abstract
Marfan syndrome (MFS) is a heritable systemic connective tissue disease with important cardiovascular involvement, including aortic root dilatation and mitral valve prolapse. Life expectancy in patients with MFS is mainly determined by cardiovascular complications, among which aortic dissection or rupture are most dreaded. In recent years, heart failure and ventricular arrhythmia have drawn attention as extra-aortic cardiovascular manifestations and as additional reported causes of death. Imaging studies have provided data supporting a primary myocardial impairment in the absence of valvular disease or cardiovascular surgery, while studies using ambulatory ECG have demonstrated an increased susceptibility to ventricular arrhythmia. In this paper, current literature was reviewed in order to provide insights in characteristics, pathophysiology and evolution of myocardial function, heart failure and ventricular arrhythmia in MFS.
Collapse
Affiliation(s)
- Anthony Demolder
- Centre for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium; (L.M.-M.); (J.D.B.)
| | | | - Laura Muiño-Mosquera
- Centre for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium; (L.M.-M.); (J.D.B.)
- Department of Paediatrics, Division of Paediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie De Backer
- Centre for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium; (L.M.-M.); (J.D.B.)
- Department of Cardiology, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
24
|
Bigotti MG, Skeffington KL, Jones FP, Caputo M, Brancaccio A. Agrin-Mediated Cardiac Regeneration: Some Open Questions. Front Bioeng Biotechnol 2020; 8:594. [PMID: 32612983 PMCID: PMC7308530 DOI: 10.3389/fbioe.2020.00594] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/15/2020] [Indexed: 01/07/2023] Open
Abstract
After cardiac injury, the mammalian adult heart has a very limited capacity to regenerate, due to the inability of fully differentiated cardiomyocytes (CMs) to efficiently proliferate. This has been directly linked to the extracellular matrix (ECM) surrounding and connecting cardiomyocytes, as its increasing rigidity during heart maturation has a crucial impact over the proliferative capacity of CMs. Very recent studies using mouse models have demonstrated how the ECM protein agrin might promote heart regeneration through CMs de-differentiation and proliferation. In maturing CMs, this proteoglycan would act as an inducer of a specific molecular pathway involving ECM receptor(s) within the transmembrane dystrophin-glycoprotein complex (DGC) as well as intracellular Yap, an effector of the Hippo pathway involved in the replication/regeneration program of CMs. According to the mechanism proposed, during mice heart development agrin gets progressively downregulated and ultimately replaced by other ECM proteins eventually leading to loss of proliferation/ regenerative capacity in mature CMs. Although the role played by the agrin-DGC-YAP axis during human heart development remains still largely to be defined, this scenario opens up fascinating and promising therapeutic avenues. Herein, we discuss the currently available relevant information on this system, with a view to explore how the fundamental understanding of the regenerative potential of this cellular program can be translated into therapeutic treatment of injured human hearts.
Collapse
Affiliation(s)
- Maria Giulia Bigotti
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol, United Kingdom.,School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Katie L Skeffington
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Ffion P Jones
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Andrea Brancaccio
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.,Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Rome, Italy
| |
Collapse
|
25
|
Werner M, Kurniawan NA, Bouten CVC. Cellular Geometry Sensing at Different Length Scales and its Implications for Scaffold Design. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E963. [PMID: 32098110 PMCID: PMC7078773 DOI: 10.3390/ma13040963] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Geometrical cues provided by the intrinsic architecture of tissues and implanted biomaterials have a high relevance in controlling cellular behavior. Knowledge of how cells sense and subsequently respond to complex geometrical cues of various sizes and origins is needed to understand the role of the architecture of the extracellular environment as a cell-instructive parameter. This is of particular interest in the field of tissue engineering, where the success of scaffold-guided tissue regeneration largely depends on the formation of new tissue in a native-like organization in order to ensure proper tissue function. A well-considered internal scaffold design (i.e., the inner architecture of the porous structure) can largely contribute to the desired cell and tissue organization. Advances in scaffold production techniques for tissue engineering purposes in the last years have provided the possibility to accurately create scaffolds with defined macroscale external and microscale internal architectures. Using the knowledge of how cells sense geometrical cues of different size ranges can drive the rational design of scaffolds that control cellular and tissue architecture. This concise review addresses the recently gained knowledge of the sensory mechanisms of cells towards geometrical cues of different sizes (from the nanometer to millimeter scale) and points out how this insight can contribute to informed architectural scaffold designs.
Collapse
Affiliation(s)
- Maike Werner
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Nicholas A. Kurniawan
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
26
|
Zhao Y, Rafatian N, Wang EY, Wu Q, Lai BFL, Lu RX, Savoji H, Radisic M. Towards chamber specific heart-on-a-chip for drug testing applications. Adv Drug Deliv Rev 2020; 165-166:60-76. [PMID: 31917972 PMCID: PMC7338250 DOI: 10.1016/j.addr.2019.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Modeling of human organs has long been a task for scientists in order to lower the costs of therapeutic development and understand the pathological onset of human disease. For decades, despite marked differences in genetics and etiology, animal models remained the norm for drug discovery and disease modeling. Innovative biofabrication techniques have facilitated the development of organ-on-a-chip technology that has great potential to complement conventional animal models. However, human organ as a whole, more specifically the human heart, is difficult to regenerate in vitro, in terms of its chamber specific orientation and its electrical functional complexity. Recent progress with the development of induced pluripotent stem cell differentiation protocols, made recapitulating the complexity of the human heart possible through the generation of cells representative of atrial & ventricular tissue, the sinoatrial node, atrioventricular node and Purkinje fibers. Current heart-on-a-chip approaches incorporate biological, electrical, mechanical, and topographical cues to facilitate tissue maturation, therefore improving the predictive power for the chamber-specific therapeutic effects targeting adult human. In this review, we will give a summary of current advances in heart-on-a-chip technology and provide a comprehensive outlook on the challenges involved in the development of human physiologically relevant heart-on-a-chip.
Collapse
Affiliation(s)
- Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University of Health Network, Toronto, Ontario M5G 2N2, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Benjamin F L Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
27
|
Alassaf A, Tansik G, Mayo V, Wubker L, Carbonero D, Agarwal A. Engineering anisotropic cardiac monolayers on microelectrode arrays for non-invasive analyses of electrophysiological properties. Analyst 2020; 145:139-149. [DOI: 10.1039/c9an01339c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Engineering cardiac tissues with physiological architectural and mechanical properties on microelectrode arrays enables long term culture and non-invasive collection of electrophysiological readouts.
Collapse
Affiliation(s)
- Ahmad Alassaf
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Department of Medical Equipment Technology
| | - Gulistan Tansik
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Vera Mayo
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Laura Wubker
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Daniel Carbonero
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Dr. John T Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami
| |
Collapse
|
28
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
29
|
Wanjare M, Kawamura M, Hu C, Alcazar C, Wang H, Woo YJ, Huang NF. Vascularization of Engineered Spatially Patterned Myocardial Tissue Derived From Human Pluripotent Stem Cells in vivo. Front Bioeng Biotechnol 2019; 7:208. [PMID: 31552234 PMCID: PMC6733921 DOI: 10.3389/fbioe.2019.00208] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Tissue engineering approaches to regenerate myocardial tissue after disease or injury is promising. Integration with the host vasculature is critical to the survival and therapeutic efficacy of engineered myocardial tissues. To create more physiologically oriented engineered myocardial tissue with organized cellular arrangements and endothelial interactions, randomly oriented or parallel-aligned microfibrous polycaprolactone scaffolds were seeded with human pluripotent stem cell-derived cardiomyocytes (iCMs) and/or endothelial cells (iECs). The resultant engineered myocardial tissues were assessed in a subcutaneous transplantation model and in a myocardial injury model to evaluate the effect of scaffold anisotropy and endothelial interactions on vascular integration of the engineered myocardial tissue. Here we demonstrated that engineered myocardial tissue composed of randomly oriented scaffolds seeded with iECs promoted the survival of iECs for up to 14 days. However, engineered myocardial tissue composed of aligned scaffolds preferentially guided the organization of host capillaries along the direction of the microfibers. In a myocardial injury model, epicardially transplanted engineered myocardial tissues composed of randomly oriented scaffolds seeded with iCMs augmented microvessel formation leading to a significantly higher arteriole density after 4 weeks, compared to engineered tissues derived from aligned scaffolds. These findings that the scaffold microtopography imparts differential effect on revascularization, in which randomly oriented scaffolds promote pro-survival and pro-angiogenic effects, and aligned scaffolds direct the formation of anisotropic vessels. These findings suggest a dominant role of scaffold topography over endothelial co-culture in modulating cellular survival, vascularization, and microvessel architecture.
Collapse
Affiliation(s)
- Maureen Wanjare
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Masashi Kawamura
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Caroline Hu
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Cynthia Alcazar
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Ngan F Huang
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
30
|
Camacho P, Busari H, Seims KB, Schwarzenberg P, Dailey HL, Chow LW. 3D printing with peptide-polymer conjugates for single-step fabrication of spatially functionalized scaffolds. Biomater Sci 2019; 7:4237-4247. [PMID: 31393469 DOI: 10.1039/c9bm00887j] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Biodegradable polymer-based scaffolds are widely used to provide support during early stages of regeneration and can be functionalized with various chemical groups or bioactive cues to promote desired cellular behavior. However, these scaffolds are often modified post-fabrication, which can lead to undesired changes and homogeneously distributed chemistries that fail to mimic the spatial biochemical organization found in native tissues. To address these challenges, surface functionalization can be achieved by 3D printing with pre-functionalized biodegradable polymers, such as peptide-modified polymer conjugates, to control the deposition of preferred chemistries. Peptide-PCL conjugates were synthesized with the canonical cell adhesion peptide motif RGDS or its negative control RGES and 3D printed into scaffolds displaying one or both peptides. The peptides were also modified with bioorthogonal groups, biotin and azide, to visualize peptide concentration and location by labeling with complementary fluorophores. Peptide concentration on the scaffold surface increased with increasing peptide-PCL conjugate concentration added to the ink prior to 3D printing, and scaffolds printed with the highest RGDS(biotin)-PCL concentrations showed a significant increase in NIH3T3 fibroblast adhesion. To demonstrate spatial control of peptide functionalization, multiple printer heads were used to print both peptide-PCL conjugates into the same construct in alternating patterns. Cells preferentially attached and spread on RGDS(biotin)-PCL fibers compared to RGES(azide)-PCL fibers, illustrating how spatial functionalization can be used to influence local cell behavior within a single biomaterial. This presents a versatile platform to generate multifunctional biomaterials that can mimic the biochemical organization found in native tissues to support functional regeneration.
Collapse
Affiliation(s)
- Paula Camacho
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA.
| | - Hafiz Busari
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, USA
| | - Kelly B Seims
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, USA
| | | | - Hannah L Dailey
- Department of Mechanical Engineering and Mechanics, Bethlehem, PA, USA
| | - Lesley W Chow
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA. and Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, USA
| |
Collapse
|
31
|
Heikkinen JJ, Peltola E, Wester N, Koskinen J, Laurila T, Franssila S, Jokinen V. Fabrication of Micro- and Nanopillars from Pyrolytic Carbon and Tetrahedral Amorphous Carbon. MICROMACHINES 2019; 10:E510. [PMID: 31370267 PMCID: PMC6723446 DOI: 10.3390/mi10080510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022]
Abstract
Pattern formation of pyrolyzed carbon (PyC) and tetrahedral amorphous carbon (ta-C) thin films were investigated at micro- and nanoscale. Micro- and nanopillars were fabricated from both materials, and their biocompatibility was studied with cell viability tests. Carbon materials are known to be very challenging to pattern. Here we demonstrate two approaches to create biocompatible carbon features. The microtopographies were 2 μ m or 20 μ m pillars (1:1 aspect ratio) with three different pillar layouts (square-grid, hexa-grid, or random-grid orientation). The nanoscale topography consisted of random nanopillars fabricated by maskless anisotropic etching. The PyC structures were fabricated with photolithography and embossing techniques in SU-8 photopolymer which was pyrolyzed in an inert atmosphere. The ta-C is a thin film coating, and the structures for it were fabricated on silicon substrates. Despite different fabrication methods, both materials were formed into comparable micro- and nanostructures. Mouse neural stem cells were cultured on the samples (without any coatings) and their viability was evaluated with colorimetric viability assay. All samples expressed good biocompatibility, but the topography has only a minor effect on viability. Two μ m pillars in ta-C shows increased cell count and aggregation compared to planar ta-C reference sample. The presented materials and fabrication techniques are well suited for applications that require carbon chemistry and benefit from large surface area and topography, such as electrophysiological and -chemical sensors for in vivo and in vitro measurements.
Collapse
Affiliation(s)
- Joonas J Heikkinen
- Department of Chemistry and Materials Science, Aalto University, Tietotie 3, 02150 Espoo, Finland.
| | - Emilia Peltola
- Department of Electrical Engineering and Automation, Aalto University, Tietotie 3, 02150 Espoo, Finland
| | - Niklas Wester
- Department of Chemistry and Materials Science, Aalto University, Kemistintie 1, 02150 Espoo, Finland
| | - Jari Koskinen
- Department of Chemistry and Materials Science, Aalto University, Kemistintie 1, 02150 Espoo, Finland
| | - Tomi Laurila
- Department of Electrical Engineering and Automation, Aalto University, Tietotie 3, 02150 Espoo, Finland
| | - Sami Franssila
- Department of Chemistry and Materials Science, Aalto University, Tietotie 3, 02150 Espoo, Finland
| | - Ville Jokinen
- Department of Chemistry and Materials Science, Aalto University, Tietotie 3, 02150 Espoo, Finland.
| |
Collapse
|
32
|
Simeonov S, Schäffer TE. Ultrafast Imaging of Cardiomyocyte Contractions by Combining Scanning Ion Conductance Microscopy with a Microelectrode Array. Anal Chem 2019; 91:9648-9655. [PMID: 31247725 DOI: 10.1021/acs.analchem.9b01092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Beating cardiomyocytes undergo fast morphodynamics during the contraction-relaxation cycle. However, imaging these morphodynamics with a high spatial and temporal resolution is difficult, owing to a lack of suitable techniques. Here, we combine scanning ion conductance microscopy (SICM) with a microelectrode array (MEA) to image the three-dimensional (3D) topography of cardiomyocytes during a contraction-relaxation cycle with 1 μm spatial and 1 ms time resolution. We record the vertical motion of cardiomyocytes at many locations across a cell by SICM and synchronize these data using the simultaneously recorded action potential by the MEA as a time reference. This allows us to reconstruct the time-resolved 3D morphology of cardiomyocytes during a full contraction-relaxation cycle with a raw data rate of 200 μs/frame and to generate spatially resolved images of contractile parameters (maximum displacement, time delay, asymmetry factor). We use the MEA-SICM setup to visualize the effect of blebbistatin, a myosin II inhibitor, on the morphodynamics of contractions. Further, we find an upper limit of 0.02% for cell volume changes during an action potential. The results show that MEA-SICM provides an ultrafast imaging platform for investigating the functional interplay of cardiomyocyte electrophysiology and mechanics.
Collapse
Affiliation(s)
- Stefan Simeonov
- Institute of Applied Physics , University of Tübingen , Auf der Morgenstelle 10 , 72076 Tübingen , Germany
| | - Tilman E Schäffer
- Institute of Applied Physics , University of Tübingen , Auf der Morgenstelle 10 , 72076 Tübingen , Germany
| |
Collapse
|
33
|
Corbin EA, Vite A, Peyster EG, Bhoopalam M, Brandimarto J, Wang X, Bennett AI, Clark AT, Cheng X, Turner KT, Musunuru K, Margulies KB. Tunable and Reversible Substrate Stiffness Reveals a Dynamic Mechanosensitivity of Cardiomyocytes. ACS APPLIED MATERIALS & INTERFACES 2019; 11:20603-20614. [PMID: 31074953 DOI: 10.1021/acsami.9b02446] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
New directions in material applications have allowed for the fresh insight into the coordination of biophysical cues and regulators. Although the role of the mechanical microenvironment on cell responses and mechanics is often studied, most analyses only consider static environments and behavior, however, cells and tissues are themselves dynamic materials that adapt in myriad ways to alterations in their environment. Here, we introduce an approach, through the addition of magnetic inclusions into a soft poly(dimethylsiloxane) elastomer, to fabricate a substrate that can be stiffened nearly instantaneously in the presence of cells through the use of a magnetic gradient to investigate short-term cellular responses to dynamic stiffening or softening. This substrate allows us to observe time-dependent changes, such as spreading, stress fiber formation, Yes-associated protein translocation, and sarcomere organization. The identification of temporal dynamic changes on a short time scale suggests that this technology can be more broadly applied to study targeted mechanisms of diverse biologic processes, including cell division, differentiation, tissue repair, pathological adaptations, and cell-death pathways. Our method provides a unique in vitro platform for studying the dynamic cell behavior by better mimicking more complex and realistic microenvironments. This platform will be amenable to future studies aimed at elucidating the mechanisms underlying mechanical sensing and signaling that influence cellular behaviors and interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Andy T Clark
- Department of Physics , Bryn Mawr College , Bryn Mawr , Pennsylvania 19010 , United States
| | - Xuemei Cheng
- Department of Physics , Bryn Mawr College , Bryn Mawr , Pennsylvania 19010 , United States
| | | | | | | |
Collapse
|
34
|
Avazmohammadi R, Soares JS, Li DS, Raut SS, Gorman RC, Sacks MS. A Contemporary Look at Biomechanical Models of Myocardium. Annu Rev Biomed Eng 2019; 21:417-442. [PMID: 31167105 PMCID: PMC6626320 DOI: 10.1146/annurev-bioeng-062117-121129] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Understanding and predicting the mechanical behavior of myocardium under healthy and pathophysiological conditions are vital to developing novel cardiac therapies and promoting personalized interventions. Within the past 30 years, various constitutive models have been proposed for the passive mechanical behavior of myocardium. These models cover a broad range of mathematical forms, microstructural observations, and specific test conditions to which they are fitted. We present a critical review of these models, covering both phenomenological and structural approaches, and their relations to the underlying structure and function of myocardium. We further explore the experimental and numerical techniques used to identify the model parameters. Next, we provide a brief overview of continuum-level electromechanical models of myocardium, with a focus on the methods used to integrate the active and passive components of myocardial behavior. We conclude by pointing to future directions in the areas of optimal form as well as new approaches for constitutive modeling of myocardium.
Collapse
Affiliation(s)
- Reza Avazmohammadi
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA;
| | - João S Soares
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA;
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, USA
| | - David S Li
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA;
| | - Samarth S Raut
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA;
| | - Robert C Gorman
- Gorman Cardiovascular Research Group, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA;
| |
Collapse
|
35
|
Hosoyama K, Ahumada M, Goel K, Ruel M, Suuronen EJ, Alarcon EI. Electroconductive materials as biomimetic platforms for tissue regeneration. Biotechnol Adv 2019; 37:444-458. [DOI: 10.1016/j.biotechadv.2019.02.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/03/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
|
36
|
Yang GH, Lee J, Kim G. The fabrication of uniaxially aligned micro-textured polycaprolactone struts and application for skeletal muscle tissue regeneration. Biofabrication 2019; 11:025005. [DOI: 10.1088/1758-5090/ab0098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
37
|
Martin LJ, Pilipenko V, Benson DW. Role of Segregation for Variant Discovery in Multiplex Families Ascertained by Probands With Left Sided Cardiovascular Malformations. Front Genet 2019; 9:729. [PMID: 30687393 PMCID: PMC6336695 DOI: 10.3389/fgene.2018.00729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/22/2018] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular malformations (CVM) are common birth defects (incidence of 2-5/100 live births). Although a genetic basis is established, in most cases the cause remains unknown. Analysis of whole exome sequencing (WES) in left sided CVM case and trio series has identified large numbers of potential variants but evidence of causality has remained elusive except in a small percentage of cases. We sought to determine whether variant segregation in families would aid in novel gene discovery. The objective was to compare conventional and co-segregation approaches for WES in multiplex families. WES was performed on 52 individuals from 4 multiplex families ascertained by probands with hypoplastic left heart syndrome (HLHS). We identified rare variants with informatics support (RVIS, minor allele frequency ≤0.01 and Combined Annotation Dependent Depletion score ≥20) in probands. Non-RVIS variants did not meet these criteria. Family specific two point logarithm of the odds (LOD) scores identified co-segregating variants (C-SV) using a dominant model and 80% penetrance. In families, 702 RVIS in 668 genes were identified, but only 1 RVIS was also a C-SV (LOD ≥ 1). On the other hand, there were 109 non-RVIS variants with LOD ≥ 1. Among 110 C-SV, 97% were common (MAF > 1%). These results suggest that conventional variant identification methods focused on RVIS, miss most C-SV. For diseases such as left sided CVM, which exhibit strong familial transmission, co-segregation can identify novel candidates.
Collapse
Affiliation(s)
- Lisa J Martin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Valentina Pilipenko
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - D Woodrow Benson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
38
|
Hosoyama K, Ahumada M, McTiernan CD, Davis DR, Variola F, Ruel M, Liang W, Suuronen EJ, Alarcon EI. Nanoengineered Electroconductive Collagen-Based Cardiac Patch for Infarcted Myocardium Repair. ACS APPLIED MATERIALS & INTERFACES 2018; 10:44668-44677. [PMID: 30508481 DOI: 10.1021/acsami.8b18844] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We have prepared and tested in vivo a novel nanoengineered hybrid electroconductive cardiac patch for treating the infarcted myocardium. Of the prepared and tested patches, only those containing spherical nanogold were able to increase connexin-43 expression in neonatal rat cardiomyocytes cultured under electrical stimulation. In vivo data indicated that only nano-gold-containing patches were able to recover cardiac function. Histological analysis also revealed that connexin-43 levels and blood vessel density were increased, while the scar size was reduced for animals that received the nanogold patch. Thus, our study indicates that the incorporation of electroconductive properties into a collagen-based cardiac patch can improve its therapeutic potential for treating myocardial infarction.
Collapse
Affiliation(s)
- Katsuhiro Hosoyama
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
| | - Manuel Ahumada
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Bionanomaterials Chemistry and Engineering Laboratory & Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Christopher D McTiernan
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Bionanomaterials Chemistry and Engineering Laboratory & Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Fabio Variola
- Department of Mechanical Engineering , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Marc Ruel
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Wenbin Liang
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
- University of Ottawa Heart Institute, Cardiac Electrophysiology Lab , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
| | - Erik J Suuronen
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Emilio I Alarcon
- University of Ottawa Heart Institute, Division of Cardiac Surgery , University of Ottawa , Ottawa , Ontario K1Y 4W7 , Canada
- Bionanomaterials Chemistry and Engineering Laboratory & Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| |
Collapse
|
39
|
Jamilpour N, Nam KH, Gregorio CC, Wong PK. Probing Collective Mechanoadaptation in Cardiomyocyte Development by Plasma Lithography Patterned Elastomeric Substrates. ACS Biomater Sci Eng 2018; 5:3808-3816. [DOI: 10.1021/acsbiomaterials.8b00815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Nima Jamilpour
- Department of Aerospace and Mechanical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Ki-Hwan Nam
- Department of Aerospace and Mechanical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Optical Instrumentation Development Team, Korea Basic Science Institute (KBSI), 169-148 Gwahak-ro Yuseong-gu, Daejeon 34133, Rep. of Korea
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine, Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona 85721, United States
| | - Pak Kin Wong
- Department of Aerospace and Mechanical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Departments of Biomedical Engineering, Mechanical Engineering and Surgery, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
40
|
Joshi J, Brennan D, Beachley V, Kothapalli CR. Cardiomyogenic differentiation of human bone marrow-derived mesenchymal stem cell spheroids within electrospun collagen nanofiber mats. J Biomed Mater Res A 2018; 106:3303-3312. [PMID: 30242963 DOI: 10.1002/jbm.a.36530] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/26/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022]
Abstract
Collagen is the major structural protein in myocardium and contributes to tissue strength and integrity, cellular orientation, and cell-cell and cell-matrix interactions. Significant post-myocardial infarction related loss of cardiomyocytes and cardiac tissue, and their subsequent replacement with fibrous scar tissue, negatively impacts endogenous tissue repair and regeneration capabilities. To overcome such limitations, tissue engineers are working toward developing a 3D cardiac patch which not only mimics the structural, functional, and biological hierarchy of the native cardiac tissue, but also could deliver autologous stem cells and encourage their homing and differentiation. In this study, we examined the utility of electrospun, randomly-oriented, type-I collagen nanofiber (dia = 789 ± 162 nm) mats on the cardiomyogenic differentiation of human bone marrow-derived mesenchymal stem cells (BM-MSC) spheroids, in the presence or absence of 10 μM 5-azacytidine (aza). Results showed that these scaffolds are biocompatible and enable time-dependent evolution of early (GATA binding protein 4: GATA4), late (cardiac troponin I: cTnI), and mature (myosin heavy chain: MHC) cardiomyogenic markers, with a simultaneous reduction in CD90 (stemness) expression, independent of aza-treatment. Aza-exposure improved connexin-4 expression and sustained sarcomeric α-actin expression, but provided only transient improvement in cardiac troponin T (cTnT) expression. Cell orientation and alignment significantly improved in these nanofiber scaffolds over time and with aza-exposure. Although further quantitative in vitro and in vivo studies are needed to establish the clinical applicability of such stem-cell laden collagen nanofiber mats as cardiac patches for cardiac tissue regeneration, our results underscore the benefits of 3D milieu provided by electrospun collagen nanofiber mats, aza, and spheroids on the survival, cardiac differentiation and maturation of human BM-MSCs. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3303-3312, 2018.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio, 44115
| | - David Brennan
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, 08028
| | - Vince Beachley
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, 08028
| | | |
Collapse
|
41
|
Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, Yu ZX, Fisher JP, Mohiuddin M, Zhang LG. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv Drug Deliv Rev 2018; 132:252-269. [PMID: 30053441 PMCID: PMC6226324 DOI: 10.1016/j.addr.2018.07.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Compared to traditional therapeutic strategies, three-dimensional (3D) bioprinting is one of the most advanced techniques for creating complicated cardiovascular implants with biomimetic features, which are capable of recapitulating both the native physiochemical and biomechanical characteristics of the cardiovascular system. The present review provides an overview of the cardiovascular system, as well as describes the principles of, and recent advances in, 3D bioprinting cardiovascular tissues and models. Moreover, this review will focus on the applications of 3D bioprinting technology in cardiovascular repair/regeneration and pharmacological modeling, further discussing current challenges and perspectives.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, USA
| | | | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
42
|
Elucidating molecular events underlying topography mediated cardiomyogenesis of stem cells on 3D nanofibrous scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 88:104-114. [DOI: 10.1016/j.msec.2018.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/27/2018] [Accepted: 03/14/2018] [Indexed: 12/28/2022]
|
43
|
Multi-length scale bioprinting towards simulating microenvironmental cues. Biodes Manuf 2018; 1:77-88. [PMID: 30546920 PMCID: PMC6267274 DOI: 10.1007/s42242-018-0014-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/02/2018] [Indexed: 02/08/2023]
Abstract
It is envisaged that the creation of cellular environments at multiple length scales, that recapitulate in vivo bioactive and structural roles, may hold the key to creating functional, complex tissues in the laboratory. This review considers recent advances in biofabrication and bioprinting techniques across different length scales. Particular focus is placed on 3D printing of hydrogels and fabrication of biomaterial fibres that could extend the feature resolution and material functionality of soft tissue constructs. The outlook from this review discusses how one might create and simulate microenvironmental cues in vitro. A fabrication platform that integrates the competencies of different biofabrication technologies is proposed. Such a multi-process, multiscale fabrication strategy may ultimately translate engineering capability into an accessible life sciences toolkit, fulfilling its potential to deliver in vitro disease models and engineered tissue implants.
Collapse
|
44
|
|
45
|
Pasqualini FS, Agarwal A, O'Connor BB, Liu Q, Sheehy SP, Parker KK. Traction force microscopy of engineered cardiac tissues. PLoS One 2018; 13:e0194706. [PMID: 29590169 PMCID: PMC5874032 DOI: 10.1371/journal.pone.0194706] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/08/2018] [Indexed: 01/08/2023] Open
Abstract
Cardiac tissue development and pathology have been shown to depend sensitively on microenvironmental mechanical factors, such as extracellular matrix stiffness, in both in vivo and in vitro systems. We present a novel quantitative approach to assess cardiac structure and function by extending the classical traction force microscopy technique to tissue-level preparations. Using this system, we investigated the relationship between contractile proficiency and metabolism in neonate rat ventricular myocytes (NRVM) cultured on gels with stiffness mimicking soft immature (1 kPa), normal healthy (13 kPa), and stiff diseased (90 kPa) cardiac microenvironments. We found that tissues engineered on the softest gels generated the least amount of stress and had the smallest work output. Conversely, cardiomyocytes in tissues engineered on healthy- and disease-mimicking gels generated significantly higher stresses, with the maximal contractile work measured in NRVM engineered on gels of normal stiffness. Interestingly, although tissues on soft gels exhibited poor stress generation and work production, their basal metabolic respiration rate was significantly more elevated than in other groups, suggesting a highly ineffective coupling between energy production and contractile work output. Our novel platform can thus be utilized to quantitatively assess the mechanotransduction pathways that initiate tissue-level structural and functional remodeling in response to substrate stiffness.
Collapse
Affiliation(s)
- Francesco Silvio Pasqualini
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Ashutosh Agarwal
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States of America
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute, Miami, FL, United States of America
| | - Blakely Bussie O'Connor
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Qihan Liu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
46
|
Rose JC, Gehlen DB, Haraszti T, Köhler J, Licht CJ, De Laporte L. Biofunctionalized aligned microgels provide 3D cell guidance to mimic complex tissue matrices. Biomaterials 2018; 163:128-141. [PMID: 29459322 DOI: 10.1016/j.biomaterials.2018.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 12/27/2022]
Abstract
Natural healing is based on highly orchestrated processes, in which the extracellular matrix plays a key role. To resemble the native cell environment, we introduce an artificial extracellular matrix (aECM) with the capability to template hierarchical and anisotropic structures in situ, allowing a minimally-invasive application via injection. Synthetic, magnetically responsive, rod-shaped microgels are locally aligned and fixed by a biocompatible surrounding hydrogel, creating a hybrid anisotropic hydrogel (Anisogel), of which the physical, mechanical, and chemical properties can be tailored. The microgels are rendered cell-adhesive with GRGDS and incorporated either inside a cell-adhesive fibrin or bioinert poly(ethylene glycol) hydrogel to strongly interact with fibroblasts. GRGDS-modified microgels inside a fibrin-based Anisogel enhance fibroblast alignment and lead to a reduction in fibronectin production, indicating successful replacement of structural proteins. In addition, YAP-translocation to the nucleus increases with the concentration of microgels, indicating cellular sensing of the overall anisotropic mechanical properties of the Anisogel. For bioinert surrounding PEG hydrogels, GRGDS-microgels are required to support cell proliferation and fibronectin production. In contrast to fibroblasts, primary nerve growth is not significantly affected by the biomodification of the microgels. In conclusion, this approach opens new opportunities towards advanced and complex aECMs for tissue regeneration.
Collapse
Affiliation(s)
- Jonas C Rose
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - David B Gehlen
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Tamás Haraszti
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Jens Köhler
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | | | - Laura De Laporte
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany.
| |
Collapse
|
47
|
Nawroth JC, Scudder LL, Halvorson RT, Tresback J, Ferrier JP, Sheehy SP, Cho A, Kannan S, Sunyovszki I, Goss JA, Campbell PH, Parker KK. Automated fabrication of photopatterned gelatin hydrogels for organ-on-chips applications. Biofabrication 2018; 10:025004. [PMID: 29337695 PMCID: PMC6221195 DOI: 10.1088/1758-5090/aa96de] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Organ-on-chip platforms aim to improve preclinical models for organ-level responses to novel drug compounds. Heart-on-a-chip assays in particular require tissue engineering techniques that rely on labor-intensive photolithographic fabrication or resolution-limited 3D printing of micropatterned substrates, which limits turnover and flexibility of prototyping. We present a rapid and automated method for large scale on-demand micropatterning of gelatin hydrogels for organ-on-chip applications using a novel biocompatible laser-etching approach. Fast and automated micropatterning is achieved via photosensitization of gelatin using riboflavin-5'phosphate followed by UV laser-mediated photoablation of the gel surface in user-defined patterns only limited by the resolution of the 15 μm wide laser focal point. Using this photopatterning approach, we generated microscale surface groove and pillar structures with feature dimensions on the order of 10-30 μm. The standard deviation of feature height was 0.3 μm, demonstrating robustness and reproducibility. Importantly, the UV-patterning process is non-destructive and does not alter gelatin micromechanical properties. Furthermore, as a quality control step, UV-patterned heart chip substrates were seeded with rat or human cardiac myocytes, and we verified that the resulting cardiac tissues achieved structural organization, contractile function, and long-term viability comparable to manually patterned gelatin substrates. Start-to-finish, UV-patterning shortened the time required to design and manufacture micropatterned gelatin substrates for heart-on-chip applications by up to 60% compared to traditional lithography-based approaches, providing an important technological advance enroute to automated and continuous manufacturing of organ-on-chips.
Collapse
Affiliation(s)
- Janna C. Nawroth
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Lisa L. Scudder
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Ryan T. Halvorson
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jason Tresback
- Center for Nanoscale Systems, Harvard University, Cambridge, MA, USA
| | - John P. Ferrier
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Alex Cho
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Suraj Kannan
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Ilona Sunyovszki
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Josue A. Goss
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Patrick H. Campbell
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| |
Collapse
|
48
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
49
|
The Rapidly Evolving Concept of Whole Heart Engineering. Stem Cells Int 2017; 2017:8920940. [PMID: 29250121 PMCID: PMC5700515 DOI: 10.1155/2017/8920940] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/12/2017] [Indexed: 01/10/2023] Open
Abstract
Whole heart engineering represents an incredible journey with as final destination the challenging aim to solve end-stage cardiac failure with a biocompatible and living organ equivalent. Its evolution started in 2008 with rodent organs and is nowadays moving closer to clinical application thanks to scaling-up strategies to human hearts. This review will offer a comprehensive examination on the important stages to be reached for the bioengineering of the whole heart, by describing the approaches of organ decellularization, repopulation, and maturation so far applied and the novel technologies of potential interest. In addition, it will carefully address important demands that still need to be satisfied in order to move to a real clinical translation of the whole bioengineering heart concept.
Collapse
|
50
|
Caddeo S, Boffito M, Sartori S. Tissue Engineering Approaches in the Design of Healthy and Pathological In Vitro Tissue Models. Front Bioeng Biotechnol 2017; 5:40. [PMID: 28798911 PMCID: PMC5526851 DOI: 10.3389/fbioe.2017.00040] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022] Open
Abstract
In the tissue engineering (TE) paradigm, engineering and life sciences tools are combined to develop bioartificial substitutes for organs and tissues, which can in turn be applied in regenerative medicine, pharmaceutical, diagnostic, and basic research to elucidate fundamental aspects of cell functions in vivo or to identify mechanisms involved in aging processes and disease onset and progression. The complex three-dimensional (3D) microenvironment in which cells are organized in vivo allows the interaction between different cell types and between cells and the extracellular matrix, the composition of which varies as a function of the tissue, the degree of maturation, and health conditions. In this context, 3D in vitro models can more realistically reproduce a tissue or organ than two-dimensional (2D) models. Moreover, they can overcome the limitations of animal models and reduce the need for in vivo tests, according to the "3Rs" guiding principles for a more ethical research. The design of 3D engineered tissue models is currently in its development stage, showing high potential in overcoming the limitations of already available models. However, many issues are still opened, concerning the identification of the optimal scaffold-forming materials, cell source and biofabrication technology, and the best cell culture conditions (biochemical and physical cues) to finely replicate the native tissue and the surrounding environment. In the near future, 3D tissue-engineered models are expected to become useful tools in the preliminary testing and screening of drugs and therapies and in the investigation of the molecular mechanisms underpinning disease onset and progression. In this review, the application of TE principles to the design of in vitro 3D models will be surveyed, with a focus on the strengths and weaknesses of this emerging approach. In addition, a brief overview on the development of in vitro models of healthy and pathological bone, heart, pancreas, and liver will be presented.
Collapse
Affiliation(s)
- Silvia Caddeo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, Amsterdam, Netherlands
| | - Monica Boffito
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Susanna Sartori
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|