1
|
Lu D, Fan X. Insights into the prospects of nanobiomaterials in the treatment of cardiac arrhythmia. J Nanobiotechnology 2024; 22:523. [PMID: 39215361 PMCID: PMC11363662 DOI: 10.1186/s12951-024-02805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiac arrhythmia, a disorder of abnormal electrical activity of the heart that disturbs the rhythm of the heart, thereby affecting its normal function, is one of the leading causes of death from heart disease worldwide and causes millions of deaths each year. Currently, treatments for arrhythmia include drug therapy, radiofrequency ablation, cardiovascular implantable electronic devices (CIEDs), including pacemakers, defibrillators, and cardiac resynchronization therapy (CRT). However, these traditional treatments have several limitations, such as the side effects of medication, the risks of device implantation, and the complications of invasive surgery. Nanotechnology and nanomaterials provide safer, effective and crucial treatments to improve the quality of life of patients with cardiac arrhythmia. The large specific surface area, controlled physical and chemical properties, and good biocompatibility of nanobiomaterials make them promising for a wide range of applications, such as cardiovascular drug delivery, tissue engineering, and the diagnosis and therapeutic treatment of diseases. However, issues related to the genotoxicity, cytotoxicity and immunogenicity of nanomaterials remain and require careful consideration. In this review, we first provide a brief overview of cardiac electrophysiology, arrhythmia and current treatments for arrhythmia and discuss the potential applications of nanobiomaterials before focusing on the promising applications of nanobiomaterials in drug delivery and cardiac tissue repair. An in-depth study of the application of nanobiomaterials is expected to provide safer and more effective therapeutic options for patients with cardiac arrhythmia, thereby improving their quality of life.
Collapse
Affiliation(s)
- Dingkun Lu
- Cardiac Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohan Fan
- Cardiac Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Belzil A, Gélinas R, Comtois P. Development of a high-speed imaging system for real time evaluation and monitoring of cardiac engineered tissues. Front Bioeng Biotechnol 2024; 12:1403044. [PMID: 39188370 PMCID: PMC11345265 DOI: 10.3389/fbioe.2024.1403044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Stem cell derived cardiac monolayers have high potential for tissue regeneration, in vitro drug testing and disease modeling. However, current differentiation protocols are still sub-optimal, resulting in cultures with variable yields and properties. We propose a high-speed lenseless imaging system, integrated with an electrical stimulation unit, to optimize the generation of these cultures. This tool relies on the variations of cellular patterns, during contraction, measured by digital imaging. The imaging system can monitor cardiac cell sheet function and structure, providing the necessary tools to quickly evaluate engineered monolayer. It can record high speed videos and capture high resolution images, from which tissue spatial organization and contractile characteristics can be obtained. Validation of the system was performed using cardiomyocytes derived from human induced pluripotent stem cell and neonatal rat cardiomyocytes. The imaging system allows the observation, acquisition and analysis of important data relating to contractile activity development of cardiac cells, making it a promising tool for optimization in cardiac tissue engineering.
Collapse
Affiliation(s)
- Antoine Belzil
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada
- Department of Pharmacology and Physiology, Universite de Montreal, Montréal, QC, Canada
- Institute of Biomedical Engineering, Universite de Montreal, Montréal, QC, Canada
| | - Roselle Gélinas
- Laboratory of Genetics and Genomic Medicine of Inflammation, Montreal Heart Institute, Montréal, QC, Canada
- Department of Medicine, Universite de Montreal, Montréal, QC, Canada
| | - Philippe Comtois
- Department of Pharmacology and Physiology, Universite de Montreal, Montréal, QC, Canada
- Institute of Biomedical Engineering, Universite de Montreal, Montréal, QC, Canada
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
3
|
Xu F, Jin H, Liu L, Yang Y, Cen J, Wu Y, Chen S, Sun D. Architecture design and advanced manufacturing of heart-on-a-chip: scaffolds, stimulation and sensors. MICROSYSTEMS & NANOENGINEERING 2024; 10:96. [PMID: 39006908 PMCID: PMC11239895 DOI: 10.1038/s41378-024-00692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 07/16/2024]
Abstract
Heart-on-a-chip (HoC) has emerged as a highly efficient, cost-effective device for the development of engineered cardiac tissue, facilitating high-throughput testing in drug development and clinical treatment. HoC is primarily used to create a biomimetic microphysiological environment conducive to fostering the maturation of cardiac tissue and to gather information regarding the real-time condition of cardiac tissue. The development of architectural design and advanced manufacturing for these "3S" components, scaffolds, stimulation, and sensors is essential for improving the maturity of cardiac tissue cultivated on-chip, as well as the precision and accuracy of tissue states. In this review, the typical structures and manufacturing technologies of the "3S" components are summarized. The design and manufacturing suggestions for each component are proposed. Furthermore, key challenges and future perspectives of HoC platforms with integrated "3S" components are discussed. Architecture design concepts of scaffolds, stimulation and sensors in chips.
Collapse
Affiliation(s)
- Feng Xu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Hang Jin
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Lingling Liu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Yuanyuan Yang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Jianzheng Cen
- Guangdong Provincial People’s Hospital, Guangzhou, 510080 China
| | - Yaobin Wu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Songyue Chen
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Daoheng Sun
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| |
Collapse
|
4
|
Martín D, Ruano D, Yúfera A, Daza P. Electrical pulse stimulation parameters modulate N2a neuronal differentiation. Cell Death Discov 2024; 10:49. [PMID: 38272891 PMCID: PMC10810886 DOI: 10.1038/s41420-024-01820-y] [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: 08/01/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Electrical pulse stimulation has been used to enhance the differentiation or proliferation of neuronal progenitor cells in tissue engineering and cancer treatment. Therefore, a comprehensive investigation of the effects caused by its parameters is crucial for improvements in those fields. We propose a study of pulse parameters, to allow the control of N2a cell line fate and behavior. We have focused on designing an experimental setup that allows for the knowledge and control over the environment and the stimulation signals applied. To map the effects of the stimulation on N2a cells, their morphology and the cellular and molecular reactions induced by the pulse stimulation have been analyzed. Immunofluorescence, rt-PCR and western blot analysis have been carried out for this purpose, as well as cell counting. Our results show that low-amplitude electrical pulse stimulation promotes proliferation of N2a cells, whilst amplitudes in the range 250 mV/mm-500 mV/mm induce differentiation. Amplitudes higher than 750 mV/mm produce cell damage at low frequencies. For high frequencies, large amplitudes are needed to cause cell death. An inverse relation has been found between cell density and pulse-induced neuronal differentiation. The best condition for neuronal differentiation was found to be 500 mV/mm at 100 Hz. These findings have been confirmed by up-regulation of the Neurod1 gene. Our preliminary study of the molecular effects of electrical pulse stimulation on N2a offers premonitory clues of the PI3K/Akt/GSK-3β pathway implications on the neuronal differentiation process through ES. In general, we have successfully mapped the sensitivity of N2a cells to electrical pulse stimulation parameters.
Collapse
Affiliation(s)
- Daniel Martín
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain.
- Instituto de Microelectrónica de Sevilla (IMSE), Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain.
| | - Diego Ruano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
| | - Alberto Yúfera
- Instituto de Microelectrónica de Sevilla (IMSE), Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
- Departamento de Tecnología Electrónica, ETSII, Universidad de Sevilla, Sevilla, Spain
| | - Paula Daza
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
5
|
Suarato G, Pressi S, Menna E, Ruben M, Petrini EM, Barberis A, Miele D, Sandri G, Salerno M, Schirato A, Alabastri A, Athanassiou A, Proietti Zaccaria R, Papadopoulou EL. Modified Carbon Nanotubes Favor Fibroblast Growth by Tuning the Cell Membrane Potential. ACS APPLIED MATERIALS & INTERFACES 2024; 16:3093-3105. [PMID: 38206310 PMCID: PMC10811621 DOI: 10.1021/acsami.3c14527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024]
Abstract
As is known, carbon nanotubes favor cell growth in vitro, although the underlying mechanisms are not yet fully elucidated. In this study, we explore the hypothesis that electrostatic fields generated at the interface between nonexcitable cells and appropriate scaffold might favor cell growth by tuning their membrane potential. We focused on primary human fibroblasts grown on electrospun polymer fibers (poly(lactic acid)─PLA) with embedded multiwall carbon nanotubes (MWCNTs). The MWCNTs were functionalized with either the p-methoxyphenyl (PhOME) or the p-acetylphenyl (PhCOMe) moiety, both of which allowed uniform dispersion in a solvent, good mixing with PLA and the consequent smooth and homogeneous electrospinning process. The inclusion of the electrically conductive MWCNTs in the insulating PLA matrix resulted in differences in the surface potential of the fibers. Both PLA and PLA/MWCNT fiber samples were found to be biocompatible. The main features of fibroblasts cultured on different substrates were characterized by scanning electron microscopy, immunocytochemistry, Rt-qPCR, and electrophysiology revealing that fibroblasts grown on PLA/MWCNT reached a healthier state as compared to pure PLA. In particular, we observed physiological spreading, attachment, and Vmem of fibroblasts on PLA/MWCNT. Interestingly, the electrical functionalization of the scaffold resulted in a more suitable extracellular environment for the correct biofunctionality of these nonexcitable cells. Finally, numerical simulations were also performed in order to understand the mechanism behind the different cell behavior when grown either on PLA or PLA/MWCNT samples. The results show a clear effect on the cell membrane potential, depending on the underlying substrate.
Collapse
Affiliation(s)
- Giulia Suarato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Samuel Pressi
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Enzo Menna
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Massimo Ruben
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | | | - Andrea Barberis
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Dalila Miele
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Marco Salerno
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Schirato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
- Dipartimento
di Fisica, Politecnico di Milano, Pizza Leonardo da Vinci 32, Milan 20133, Italy
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Alessandro Alabastri
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | | | | | | |
Collapse
|
6
|
Okhovatian S, Shakeri A, Huyer LD, Radisic M. Elastomeric Polyesters in Cardiovascular Tissue Engineering and Organs-on-a-Chip. Biomacromolecules 2023; 24:4511-4531. [PMID: 37639715 PMCID: PMC10915885 DOI: 10.1021/acs.biomac.3c00387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cardiovascular tissue constructs provide unique design requirements due to their functional responses to substrate mechanical properties and cyclic stretching behavior of cardiac tissue that requires the use of durable elastic materials. Given the diversity of polyester synthesis approaches, an opportunity exists to develop a new class of biocompatible, elastic, and immunomodulatory cardiovascular polymers. Furthermore, elastomeric polyester materials have the capability to provide tailored biomechanical synergy with native tissue and hence reduce inflammatory response in vivo and better support tissue maturation in vitro. In this review, we highlight underlying chemistry and design strategies of polyester elastomers optimized for cardiac tissue scaffolds. The major advantages of these materials such as their tunable elasticity, desirable biodegradation, and potential for incorporation of bioactive compounds are further expanded. Unique fabrication methods using polyester materials such as micromolding, 3D stamping, electrospinning, laser ablation, and 3D printing are discussed. Moreover, applications of these biomaterials in cardiovascular organ-on-a-chip devices and patches are analyzed. Finally, we outline unaddressed challenges in the field that need further study to enable the impactful translation of soft polyesters to clinical applications.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Locke Davenport Huyer
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
7
|
Saveh-Shemshaki N, Barajaa MA, Otsuka T, Mirdamadi ES, Nair LS, Laurencin CT. Electroconductivity, a regenerative engineering approach to reverse rotator cuff muscle degeneration. Regen Biomater 2023; 10:rbad099. [PMID: 38020235 PMCID: PMC10676522 DOI: 10.1093/rb/rbad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Muscle degeneration is one the main factors that lead to the high rate of retear after a successful repair of rotator cuff (RC) tears. The current surgical practices have failed to treat patients with chronic massive rotator cuff tears (RCTs). Therefore, regenerative engineering approaches are being studied to address the challenges. Recent studies showed the promising outcomes of electroactive materials (EAMs) on the regeneration of electrically excitable tissues such as skeletal muscle. Here, we review the most important biological mechanism of RC muscle degeneration. Further, the review covers the recent studies on EAMs for muscle regeneration including RC muscle. Finally, we will discuss the future direction toward the application of EAMs for the augmentation of RCTs.
Collapse
Affiliation(s)
- Nikoo Saveh-Shemshaki
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Mohammed A Barajaa
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University, Dammam 31451, Saudi Arabia
| | - Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Elnaz S Mirdamadi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
8
|
English EJ, Samolyk BL, Gaudette GR, Pins GD. Micropatterned fibrin scaffolds increase cardiomyocyte alignment and contractility for the fabrication of engineered myocardial tissue. J Biomed Mater Res A 2023; 111:1309-1321. [PMID: 36932841 PMCID: PMC11128133 DOI: 10.1002/jbm.a.37530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023]
Abstract
Cardiovascular disease is the leading cause of death in the United States, which can result in blockage of a coronary artery, triggering a myocardial infarction (MI), scar tissue formation in the myocardium, and ultimately heart failure. Currently, the gold-standard solution for total heart failure is a heart transplantation. An alternative to total-organ transplantation is surgically remodeling the ventricle with the implantation of a cardiac patch. Acellular cardiac patches have previously been investigated using synthetic or decellularized native materials to improve cardiac function. However, a limitation of this strategy is that acellular cardiac patches only reshape the ventricle and do not increase cardiac contractile function. Toward the development of a cardiac patch, our laboratory previously developed a cell-populated composite fibrin scaffold and aligned microthreads to recapitulate the mechanical properties of native myocardium. In this study, we explore micropatterning the surfaces of fibrin gels to mimic anisotropic native tissue architecture and promote cellular alignment of human induced pluripotent stem cell cardiomyocytes (hiPS-CM), which is crucial for increasing scaffold contractile properties. hiPS-CMs seeded on micropatterned surfaces exhibit cellular elongation, distinct sarcomere alignment, and circumferential connexin-43 staining at 14 days of culture, which are necessary for mature contractile properties. Constructs were also subject to electrical stimulation during culture to promote increased contractile properties. After 7 days of stimulation, contractile strains of micropatterned constructs were significantly higher than unpatterned controls. These results suggest that the use of micropatterned topographic cues on fibrin scaffolds may be a promising strategy for creating engineered cardiac tissue.
Collapse
Affiliation(s)
- Elizabeth J. English
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Tessera Therapeutics, Somerville, Massachusetts, USA
| | - Bryanna L. Samolyk
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Glenn R. Gaudette
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Department of Engineering, Boston College, Newton, Massachusetts, USA
| | - George D. Pins
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
9
|
Rojas-Rodríguez M, Fiaschi T, Mannelli M, Mortati L, Celegato F, Wiersma DS, Parmeggiani C, Martella D. Cellular Contact Guidance on Liquid Crystalline Networks with Anisotropic Roughness. ACS APPLIED MATERIALS & INTERFACES 2023; 15. [PMID: 36791024 PMCID: PMC10037237 DOI: 10.1021/acsami.2c22892] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Cell contact guidance is widely employed to manipulate cell alignment and differentiation in vitro. The use of nano- or micro-patterned substrates allows efficient control of cell organization, thus opening up to biological models that cannot be reproduced spontaneously on standard culture dishes. In this paper, we explore the concept of cell contact guidance by Liquid Crystalline Networks (LCNs) presenting different surface topographies obtained by self-assembly of the monomeric mixture. The materials are prepared by photopolymerization of a low amount of diacrylate monomer dissolved in a liquid crystalline solvent, not participating in the reaction. The alignment of the liquid crystals, obtained before polymerization, determines the scaffold morphology, characterized by a nanometric structure. Such materials are able to drive the organization of different cell lines, e.g., fibroblasts and myoblasts, allowing for the alignment of single cells or high-density cell cultures. These results demonstrate the capabilities of rough surfaces prepared from the spontaneous assembly of liquid crystals to control biological models without the need of lithographic patterning or complex fabrication procedures. Interestingly, during myoblast differentiation, also myotube structuring in linear arrays is observed along the LCN fiber orientation. The implementation of this technology will open up to the formation of muscular tissue with well-aligned fibers in vitro mimicking the structure of native tissues.
Collapse
Affiliation(s)
- Marta Rojas-Rodríguez
- European
Laboratory for Non-linear Spectroscopy, via Nello Carrara 1, 50019 Sesto Fiorentino, Italy
| | - Tania Fiaschi
- Department
of Biomedical, Experimental, and Clinical Sciences “Mario Serio”, University of Florence, viale Morgagni 50, 50143 Florence, Italy
| | - Michele Mannelli
- Department
of Biomedical, Experimental, and Clinical Sciences “Mario Serio”, University of Florence, viale Morgagni 50, 50143 Florence, Italy
| | - Leonardo Mortati
- Istituto
Nazionale di Ricerca Metrologica (INRiM), strada delle Cacce 91, 10135 Turin, Italy
| | - Federica Celegato
- Istituto
Nazionale di Ricerca Metrologica (INRiM), strada delle Cacce 91, 10135 Turin, Italy
| | - Diederik S. Wiersma
- European
Laboratory for Non-linear Spectroscopy, via Nello Carrara 1, 50019 Sesto Fiorentino, Italy
- Istituto
Nazionale di Ricerca Metrologica (INRiM), strada delle Cacce 91, 10135 Turin, Italy
- Department
of Physics and Astronomy, University of
Florence, via Sansone
1, 50019 Sesto Fiorentino, Italy
| | - Camilla Parmeggiani
- European
Laboratory for Non-linear Spectroscopy, via Nello Carrara 1, 50019 Sesto Fiorentino, Italy
- Istituto
Nazionale di Ricerca Metrologica (INRiM), strada delle Cacce 91, 10135 Turin, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, via della Lastruccia 3−13, 50019 Sesto Fiorentino, Italy
| | - Daniele Martella
- European
Laboratory for Non-linear Spectroscopy, via Nello Carrara 1, 50019 Sesto Fiorentino, Italy
- Istituto
Nazionale di Ricerca Metrologica (INRiM), strada delle Cacce 91, 10135 Turin, Italy
| |
Collapse
|
10
|
Mostert D, Groenen B, Klouda L, Passier R, Goumans MJ, Kurniawan NA, Bouten CVC. Human pluripotent stem cell-derived cardiomyocytes align under cyclic strain when guided by cardiac fibroblasts. APL Bioeng 2022; 6:046108. [PMID: 36567768 PMCID: PMC9771596 DOI: 10.1063/5.0108914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The myocardium is a mechanically active tissue typified by anisotropy of the resident cells [cardiomyocytes (CMs) and cardiac fibroblasts (cFBs)] and the extracellular matrix (ECM). Upon ischemic injury, the anisotropic tissue is replaced by disorganized scar tissue, resulting in loss of coordinated contraction. Efforts to re-establish tissue anisotropy in the injured myocardium are hampered by a lack of understanding of how CM and/or cFB structural organization is affected by the two major physical cues inherent in the myocardium: ECM organization and cyclic mechanical strain. Herein, we investigate the singular and combined effect of ECM (dis)organization and cyclic strain in a two-dimensional human in vitro co-culture model of the myocardial microenvironment. We show that (an)isotropic ECM protein patterning can guide the orientation of CMs and cFBs, both in mono- and co-culture. Subsequent application of uniaxial cyclic strain-mimicking the local anisotropic deformation of beating myocardium-causes no effect when applied parallel to the anisotropic ECM. However, when cultured on isotropic substrates, cFBs, but not CMs, orient away from the direction of cyclic uniaxial strain (strain avoidance). In contrast, CMs show strain avoidance via active remodeling of their sarcomeres only when co-cultured with at least 30% cFBs. Paracrine signaling or N-cadherin-mediated communication between CMs and cFBs was no contributing factor. Our findings suggest that the mechanoresponsive cFBs provide structural guidance for CM orientation and elongation. Our study, therefore, highlights a synergistic mechanobiological interplay between CMs and cFBs in shaping tissue organization, which is of relevance for regenerating functionally organized myocardium.
Collapse
Affiliation(s)
| | - Bart Groenen
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Leda Klouda
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Marie-Jose Goumans
- Department of Cell and Chemical Biology and Center for Biomedical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | |
Collapse
|
11
|
High-throughput rhythmic regulation of cardiomyocytes by integrated electrical stimulation and video-based automated analysis biosensing platform. Biosens Bioelectron 2022; 209:114252. [DOI: 10.1016/j.bios.2022.114252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 11/22/2022]
|
12
|
Wang J, Soto F, Ma P, Ahmed R, Yang H, Chen S, Wang J, Liu C, Akin D, Fu K, Cao X, Chen P, Hsu EC, Soh HT, Stoyanova T, Wu JC, Demirci U. Acoustic Fabrication of Living Cardiomyocyte-based Hybrid Biorobots. ACS NANO 2022; 16:10219-10230. [PMID: 35671037 DOI: 10.1021/acsnano.2c01908] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Organized assemblies of cells have demonstrated promise as bioinspired actuators and devices; still, the fabrication of such "biorobots" has predominantly relied on passive assembly methods that reduce design capabilities. To address this, we have developed a strategy for the rapid formation of functional biorobots composed of live cardiomyocytes. We employ tunable acoustic fields to facilitate the efficient aggregation of millions of cells into high-density macroscopic architectures with directed cell orientation and enhanced cell-cell interaction. These biorobots can perform actuation functions both through naturally occurring contraction-relaxation cycles and through external control with chemical and electrical stimuli. We demonstrate that these biorobots can be used to achieve controlled actuation of a soft skeleton and pumping of microparticles. The biocompatible acoustic assembly strategy described here should prove generally useful for cellular manipulation in the context of tissue engineering, soft robotics, and other applications.
Collapse
Affiliation(s)
- Jie Wang
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Fernando Soto
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Peng Ma
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Rajib Ahmed
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California 94304-5427, United States
| | - Sihan Chen
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China
| | - Jibo Wang
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California 94304-5427, United States
| | - Demir Akin
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Kaiyu Fu
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Xu Cao
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California 94304-5427, United States
| | - Pu Chen
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China
| | - En-Chi Hsu
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Hyongsok Tom Soh
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Tanya Stoyanova
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California 94304-5427, United States
| | - Utkan Demirci
- Bio-Acoutic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, United States
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, United States
| |
Collapse
|
13
|
Bjørge IM, de Sousa BM, Patrício SG, Silva AS, Nogueira LP, Santos LF, Vieira SI, Haugen HJ, Correia CR, Mano JF. Bioengineered Hierarchical Bonelike Compartmentalized Microconstructs Using Nanogrooved Microdiscs. ACS APPLIED MATERIALS & INTERFACES 2022; 14:19116-19128. [PMID: 35446549 DOI: 10.1021/acsami.2c01161] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fabrication of vascularized large-scale constructs for regenerative medicine remains elusive since most strategies rely solely on cell self-organization or overly control cell positioning, failing to address nutrient diffusion limitations. We propose a modular and hierarchical tissue-engineering strategy to produce bonelike tissues carrying signals to promote prevascularization. In these 3D systems, disc-shaped microcarriers featuring nanogrooved topographical cues guide cell behavior by harnessing mechanotransduction mechanisms. A sequential seeding strategy of adipose-derived stromal cells and endothelial cells is implemented within compartmentalized, liquefied-core macrocapsules in a self-organizing and dynamic system. Importantly, our system autonomously promotes osteogenesis and construct's mineralization while promoting a favorable environment for prevascular-like endothelial organization. Given its modular and self-organizing nature, our strategy may be applied for the fabrication of larger constructs with a highly controlled starting point to be used for local regeneration upon implantation or as drug-screening platforms.
Collapse
Affiliation(s)
- Isabel M Bjørge
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| | - Bárbara M de Sousa
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Sónia G Patrício
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| | - Ana Sofia Silva
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| | - Liebert P Nogueira
- Oral Research Laboratory, Institute of Clinical Dentistry, University of Oslo, Oslo 0455, Norway
| | - Lúcia F Santos
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| | - Sandra I Vieira
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Håvard J Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo 0455, Norway
| | - Clara R Correia
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| | - João F Mano
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-168, Portugal
| |
Collapse
|
14
|
Najjari A, Mehdinavaz Aghdam R, Ebrahimi SAS, Suresh K S, Krishnan S, Shanthi C, Ramalingam M. Smart piezoelectric biomaterials for tissue engineering and regenerative medicine: a review. BIOMED ENG-BIOMED TE 2022; 67:71-88. [PMID: 35313098 DOI: 10.1515/bmt-2021-0265] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/01/2022] [Indexed: 01/06/2023]
Abstract
Due to the presence of electric fields and piezoelectricity in various living tissues, piezoelectric materials have been incorporated into biomedical applications especially for tissue regeneration. The piezoelectric scaffolds can perfectly mimic the environment of natural tissues. The ability of scaffolds which have been made from piezoelectric materials in promoting cell proliferation and regeneration of damaged tissues has encouraged researchers in biomedical areas to work on various piezoelectric materials for fabricating tissue engineering scaffolds. In this review article, the way that cells of different tissues like cardio, bone, cartilage, bladder, nerve, skin, tendon, and ligament respond to electric fields and the mechanism of tissue regeneration with the help of piezoelectric effect will be discussed. Furthermore, all of the piezoelectric materials are not suitable for biomedical applications even if they have high piezoelectricity since other properties such as biocompatibility are vital. Seen in this light, the proper piezoelectric materials which are approved for biomedical applications are mentioned. Totally, the present review introduces the recent materials and technologies that have been used for tissue engineering besides the role of electric fields in living tissues.
Collapse
Affiliation(s)
- Aryan Najjari
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | | | - S A Seyyed Ebrahimi
- Advanced Magnetic Materials Research Center, College of Engineering, University of Tehran, Tehran, Iran
| | - Shoma Suresh K
- Advanced Magnetic Materials Research Center, College of Engineering, University of Tehran, Tehran, Iran
| | - Sasirekha Krishnan
- Advanced Magnetic Materials Research Center, College of Engineering, University of Tehran, Tehran, Iran
| | - Chittibabu Shanthi
- Biomaterials & Organ Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics, School of Mechanical Engineering, Vellore Institute of Technology, Vellore, India
| | - Murugan Ramalingam
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
15
|
Bjørge IM, Correia CR, Mano JF. Hipster microcarriers: exploring geometrical and topographical cues of non-spherical microcarriers in biomedical applications. MATERIALS HORIZONS 2022; 9:908-933. [PMID: 34908074 DOI: 10.1039/d1mh01694f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Structure and organisation are key aspects of the native tissue environment, which ultimately condition cell fate via a myriad of processes, including the activation of mechanotransduction pathways. By modulating the formation of integrin-mediated adhesions and consequently impacting cell contractility, engineered geometrical and topographical cues may be introduced to activate downstream signalling and ultimately control cell morphology, proliferation, and differentiation. Microcarriers appear as attractive vehicles for cell-based tissue engineering strategies aiming to modulate this 3D environment, but also as vehicles for cell-free applications, given the ease in tuning their chemical and physical properties. In this review, geometry and topography are highlighted as two preponderant features in actively regulating interactions between cells and the extracellular matrix. While most studies focus on the 2D environment, we focus on how the incorporation of these strategies in 3D systems could be beneficial. The techniques applied to design 3D microcarriers with unique geometries and surface topographical cues are covered, as well as specific tissue engineering approaches employing these microcarriers. In fact, successfully achieving a functional histoarchitecture may depend on a combination of fine-tuned geometrically shaped microcarriers presenting intricately tailored topographical cues. Lastly, we pinpoint microcarrier geometry as a key player in cell-free biomaterial-based strategies, and its impact on drug release kinetics, the production of steerable microcarriers to target tumour cells, and as protein or antibody biosensors.
Collapse
Affiliation(s)
- Isabel M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - Clara R Correia
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
16
|
Tan YH, Helms HR, Nakayama KH. Decellularization Strategies for Regenerating Cardiac and Skeletal Muscle Tissues. Front Bioeng Biotechnol 2022; 10:831300. [PMID: 35295645 PMCID: PMC8918733 DOI: 10.3389/fbioe.2022.831300] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is associated with approximately 17.9 million deaths each year. Musculoskeletal conditions affect more than 1.71 billion people globally and are the leading cause of disability. These two areas represent a massive global health burden that is perpetuated by a lack of functionally restorative treatment options. The fields of regenerative medicine and tissue engineering offer great promise for the development of therapies to repair damaged or diseased tissues. Decellularized tissues and extracellular matrices are cornerstones of regenerative biomaterials and have been used clinically for decades and many have received FDA approval. In this review, we first discuss and compare methods used to produce decellularized tissues and ECMs from cardiac and skeletal muscle. We take a focused look at how different biophysical properties such as spatial topography, extracellular matrix composition, and mechanical characteristics influence cell behavior and function in the context of regenerative medicine. Lastly, we describe emerging research and forecast the future high impact applications of decellularized cardiac and skeletal muscle that will drive novel and effective regenerative therapies.
Collapse
Affiliation(s)
| | | | - Karina H. Nakayama
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
17
|
Khaw JS, Xue R, Cassidy NJ, Cartmell SH. Electrical stimulation of titanium to promote stem cell orientation, elongation and osteogenesis. Acta Biomater 2022; 139:204-217. [PMID: 34390847 DOI: 10.1016/j.actbio.2021.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/06/2021] [Accepted: 08/06/2021] [Indexed: 11/29/2022]
Abstract
Electrical stimulation of cells allows exogenous electric signals as stimuli to manipulate cell growth, preferential orientation and bone remodelling. In this study, commercially pure titanium discs were utilised in combination with a custom-built bioreactor to investigate the cellular responses of human mesenchymal stem cells via in-vitro functional assays. Finite element analysis revealed the homogeneous delivery of electric field in the bioreactor chamber with no detection of current density fluctuation in the proposed model. The custom-built bioreactor with capacitive stimulation delivery system features long-term stimulation with homogeneous electric field, biocompatible, sterilisable, scalable design and cost-effective in the manufacturing process. Using a continuous stimulation regime of 100 and 200 mV/mm on cp Ti discs, viability tests revealed up to an approximately 5-fold increase of cell proliferation rate as compared to non-stimulated controls. The human mesenchymal stem cells showed more elongated and differentiated morphology under this regime, with evidence of nuclear elongation and cytoskeletal orientation perpendicular to the direction of electric field. The continuous stimulation did not cause pH fluctuations and hydrogen peroxide production caused by Faradic reactions, signifying the suitability for long-term toxic free stimulation as opposed to the commonly used direct stimulation regime. An approximate of 4-fold increase in alkaline phosphatase production and approximately 9-fold increase of calcium deposition were observed on 200 mV/mm exposed samples relative to non-stimulated controls. It is worth noting that early stem cell differentiation and matrix production were observed under the said electric field even without the presence of chemical inductive growth factors. STATEMENT OF SIGNIFICANCE: This manuscript presents a study on combining pure titanium (primarily preferred as medical implant materials) and electrical stimulation in a purpose-built bioreactor with capacitive stimulation delivery system. A continuous capacitive stimulation regime on titanium disc has resulted in enhanced stem cell orientation, nuclei elongation, proliferation and differentiation as compared to non-stimulated controls. We believe that this manuscript creates a paradigm for future studies on the evolution of healthcare treatments in the area of targeted therapy on implantable and wearable medical devices through tailored innovative electrical stimulation approach, thereby influencing therapeutic conductive and electroactive biomaterials research prospects and development.
Collapse
Affiliation(s)
- Juan Shong Khaw
- The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester M13 9PL, UK
| | - Ruikang Xue
- The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester M13 9PL, UK
| | - Nigel J Cassidy
- Civil Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Sarah H Cartmell
- The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
18
|
Öztürk Ş, Shahbazi R, Zeybek ND, Kurum B, Gultekinoglu M, Aksoy EA, Demircin M, Ulubayram K. Assessment of electromechanically stimulated bone marrow stem cells seeded acellular cardiac patch in a rat myocardial infarct model. Biomed Mater 2021; 16. [PMID: 34330118 DOI: 10.1088/1748-605x/ac199a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
In this study, we evaluated cardiomyogenic differentiation of electromechanically stimulated rat bone marrow-derived stem cells (rt-BMSCs) on an acellular bovine pericardium (aBP) and we looked at the functioning of this engineered patch in a rat myocardial infarct (MI) model. aBP was prepared using a detergent-based decellularization procedure followed by rt-BMSCs seeding, and electrical, mechanical, or electromechanical stimulations (3 millisecond pulses of 5 V cm-1at 1 Hz, 5% stretching) to enhance cardiomyogenic differentiation. Furthermore, the electromechanically stimulated patch was applied to the MI region over 3 weeks. After this period, the retrieved patch and infarct region were evaluated for the presence of calcification, inflammatory reaction (CD68), patch to host tissue cell migration, and structural sarcomere protein expressions. In conjunction with any sign of calcification, a higher number of BrdU-labelled cells, and a low level of CD68 positive cells were observed in the infarct region under electromechanically stimulated conditions compared with static conditions. More importantly, MHC, SAC, Troponin T, and N-cad positive cells were observed in both infarct region, and retrieved engineered patch after 3 weeks. In a clear alignment with other results, our developed acellular patch promoted the expression of cardiomyogenic differentiation factors under electromechanical stimulation. Our engineered patch showed a successful integration with the host tissue followed by the cell migration to the infarct region.
Collapse
Affiliation(s)
- Şükrü Öztürk
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Altındağ, Ankara 06100, Turkey.,Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
| | - Reza Shahbazi
- Hematology/Oncology Division, School of Medicine, Indiana University, Indianapolis, IN, United States of America
| | - Naciye Dilara Zeybek
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Barıs Kurum
- Department of Surgery, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Turkey
| | - Merve Gultekinoglu
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Altındağ, Ankara 06100, Turkey
| | - Eda Ayse Aksoy
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Altındağ, Ankara 06100, Turkey
| | - Metin Demircin
- Departments of Thoracic Surgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Kezban Ulubayram
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Altındağ, Ankara 06100, Turkey.,Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey.,Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
| |
Collapse
|
19
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
20
|
López-Canosa A, Perez-Amodio S, Yanac-Huertas E, Ordoño J, Rodriguez-Trujillo R, Samitier J, Castaño O, Engel E. A microphysiological system combining electrospun fibers and electrical stimulation for the maturation of highly anisotropic cardiac tissue. Biofabrication 2021; 13. [PMID: 33962409 DOI: 10.1088/1758-5090/abff12] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/07/2021] [Indexed: 12/28/2022]
Abstract
The creation of cardiac tissue models for preclinical testing is still a non-solved problem in drug discovery, due to the limitations related to thein vitroreplication of cardiac tissue complexity. Among these limitations, the difficulty of mimicking the functional properties of the myocardium due to the immaturity of the used cells hampers the obtention of reliable results that could be translated into human patients.In vivomodels are the current gold standard to test new treatments, although it is widely acknowledged that the used animals are unable to fully recapitulate human physiology, which often leads to failures during clinical trials. In the present work, we present a microfluidic platform that aims to provide a range of signaling cues to immature cardiac cells to drive them towards an adult phenotype. The device combines topographical electrospun nanofibers with electrical stimulation in a microfabricated system. We validated our platform using a co-culture of neonatal mouse cardiomyocytes and cardiac fibroblasts, showing that it allows us to control the degree of anisotropy of the cardiac tissue inside the microdevice in a cost-effective way. Moreover, a 3D computational model of the electrical field was created and validated to demonstrate that our platform is able to closely match the distribution obtained with the gold standard (planar electrode technology) using inexpensive rod-shaped biocompatible stainless-steel electrodes. The functionality of the electrical stimulation was shown to induce a higher expression of the tight junction protein Cx-43, as well as the upregulation of several key genes involved in conductive and structural cardiac properties. These results validate our platform as a powerful tool for the tissue engineering community due to its low cost, high imaging compatibility, versatility, and high-throughput configuration capabilities.
Collapse
Affiliation(s)
- Adrián López-Canosa
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.,Electronics and Biomedical Engineering, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Soledad Perez-Amodio
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.,IMEM-BRT Group, Department Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| | - Eduardo Yanac-Huertas
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Jesús Ordoño
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Romen Rodriguez-Trujillo
- Electronics and Biomedical Engineering, Universitat de Barcelona (UB), 08028 Barcelona, Spain.,Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri i Reixac 15-21, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Josep Samitier
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.,Electronics and Biomedical Engineering, Universitat de Barcelona (UB), 08028 Barcelona, Spain.,Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri i Reixac 15-21, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Oscar Castaño
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.,Electronics and Biomedical Engineering, Universitat de Barcelona (UB), 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Elisabeth Engel
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.,CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.,IMEM-BRT Group, Department Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| |
Collapse
|
21
|
Batalov I, Jallerat Q, Kim S, Bliley J, Feinberg AW. Engineering aligned human cardiac muscle using developmentally inspired fibronectin micropatterns. Sci Rep 2021; 11:11502. [PMID: 34075068 PMCID: PMC8169656 DOI: 10.1038/s41598-021-87550-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/31/2021] [Indexed: 11/09/2022] Open
Abstract
Cardiac two-dimensional tissues were engineered using biomimetic micropatterns based on the fibronectin-rich extracellular matrix (ECM) of the embryonic heart. The goal of this developmentally-inspired, in vitro approach was to identify cell-cell and cell-ECM interactions in the microenvironment of the early 4-chambered vertebrate heart that drive cardiomyocyte organization and alignment. To test this, biomimetic micropatterns based on confocal imaging of fibronectin in embryonic chick myocardium were created and compared to control micropatterns designed with 2 or 20 µm wide fibronectin lines. Results show that embryonic chick cardiomyocytes have a unique density-dependent alignment on the biomimetic micropattern that is mediated in part by N-cadherin, suggesting that both cell-cell and cell-ECM interactions play an important role in the formation of aligned myocardium. Human induced pluripotent stem cell-derived cardiomyocytes also showed density-dependent alignment on the biomimetic micropattern but were overall less well organized. Interestingly, the addition of human adult cardiac fibroblasts and conditioning with T3 hormone were both shown to increase human cardiomyocyte alignment. In total, these results show that cardiomyocyte maturation state, cardiomyocyte-cardiomyocyte and cardiomyocyte-fibroblast interactions, and cardiomyocyte-ECM interactions can all play a role when engineering anisotropic cardiac tissues in vitro and provides insight as to how these factors may influence cardiogenesis in vivo.
Collapse
Affiliation(s)
- Ivan Batalov
- Department of Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Quentin Jallerat
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Sean Kim
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Jacqueline Bliley
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Adam W Feinberg
- Department of Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA. .,Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA.
| |
Collapse
|
22
|
Xavier Mendes A, Moraes Silva S, O'Connell CD, Duchi S, Quigley AF, Kapsa RMI, Moulton SE. Enhanced Electroactivity, Mechanical Properties, and Printability through the Addition of Graphene Oxide to Photo-Cross-linkable Gelatin Methacryloyl Hydrogel. ACS Biomater Sci Eng 2021; 7:2279-2295. [PMID: 33956434 DOI: 10.1021/acsbiomaterials.0c01734] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human tissues most sensitive to electrical activity such as neural and muscle tissues are relatively soft, and yet traditional conductive materials used to interface with them are typically stiffer by many orders of magnitude. Overcoming this mismatch, by creating both very soft and electroactive materials, is a major challenge in bioelectronics and biomaterials science. One strategy is to imbue soft materials, such as hydrogels, with electroactive properties by adding small amounts of highly conductive nanomaterials. However, electroactive hydrogels reported to date have required relatively large volume fractions (>1%) of added nanomaterial, have shown only modest electroactivity, and have not been processable via additive manufacturing to create 3D architectures. Here, we describe the development and characterization of improved biocompatible photo-cross-linkable soft hybrid electroactive hydrogels based on gelatin methacryloyol (GelMA) and large area graphene oxide (GO) flakes, which resolve each of these three limitations. The addition of very small amounts (less than a 0.07% volume fraction) of GO to a 5% w/v GelMA hydrogel resulted in a dramatic (∼35-fold) decrease in the impedance at 1 Hz compared with GelMA alone. The GelMA/GO coated indium tin oxide (ITO) electrode also showed a considerable reduction in the impedance at 1 kHz (down to 170 Ω compared with 340 Ω for the GelMA-coated ITO), while charge injection capacity increased more than 6-fold. We attribute this enhanced electroactivity to the increased electroactive surface area contributed by the GO. Despite this dramatic change in electroactivity, the GelMA/GO composite hydrogels' mechanical properties were only moderately affected. Mechanical properties increased by ∼2-fold, and therefore, the hydrogels' desired softness of <4 kPa was retained. Also, we demonstrate how light attenuation through the gel can be used to create a stiffness gradient with the exposed surface of the gel having an elastic modulus of <1.5 kPa. GO addition also enhanced the rheological properties of the GelMA composites, thus facilitating 3D extrusion printing. GelMA/GO enhanced filament formation as well as improved printability and the shape fidelity/integrity of 3D printed structures compared with GelMA alone. Additionally, the GelMA/GO 3D printed structures presented a higher electroactive behavior than nonprinted samples containing the same GelMA/GO amount, which can be attributed to the higher electroactive surface area of 3D printed structures. These findings provide new rational choices of electroactive hydrogel (EAH) compositions with broad potential applications in bioelectronics, tissue engineering, and drug delivery.
Collapse
Affiliation(s)
- Alexandre Xavier Mendes
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Saimon Moraes Silva
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Cathal D O'Connell
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Serena Duchi
- The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Anita F Quigley
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Robert M I Kapsa
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Simon E Moulton
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria 3122, Australia
| |
Collapse
|
23
|
Raghunathan V, Edwards SG, Leonard BC, Kim S, Evashenk AT, Song Y, Rewinski E, Marangakis Price A, Hoehn A, Chang C, Reilly CM, Muppala S, Murphy CJ, Thomasy SM. Differential effects of Hsp90 inhibition on corneal cells in vitro and in vivo. Exp Eye Res 2020; 202:108362. [PMID: 33220237 DOI: 10.1016/j.exer.2020.108362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 10/23/2022]
Abstract
The transformation of quiescent keratocytes to activated fibroblasts and myofibroblasts (KFM transformation) largely depends on transforming growth factor beta (TGFβ) signaling. Initiation of the TGFβ signaling cascade results from binding of TGFβ to the labile type I TGFβ receptor (TGFβRI), which is stabilized by the 90 kDa heat shock protein (Hsp90). Since myofibroblast persistence within the corneal stroma can result in stromal haze and corneal fibrosis in patients undergoing keratorefractive therapy, modulation of TGFβ signaling through Hsp90 inhibition would represent a novel approach to prevent myofibroblast persistence. In vitro, rabbit corneal fibroblasts (RCFs) or stratified immortalized human corneal epithelial cells (hTCEpi) were treated with a Hsp90 inhibitor (17AAG) in the presence/absence of TGFβ1. RCFs were cultured either on tissue culture plastic, anisotropically patterned substrates, and hydrogels of varying stiffness. Cellular responses to both cytoactive and variable substrates were assessed by morphologic changes to the cells, and alterations in expression patterns of key keratocyte and myofibroblast proteins using PCR, Western blotting and immunocytochemistry. Transepithelial electrical resistance (TEER) measurements were performed to establish epithelial barrier integrity. In vivo, the corneas of New Zealand White rabbits were wounded by phototherapeutic keratectomy (PTK) and treated with 17AAG (3× or 6× daily) either immediately or 7 days after wounding for 28 days. Rabbits underwent clinical ophthalmic examinations, SPOTS scoring and advanced imaging on days 0, 1, 3, 7, 10, 14, 21 and 28. On day 28, rabbits were euthanized and histopathology/immunohistochemistry was performed. In vitro data demonstrated that 17AAG inhibited KFM transformation with the de-differentiation of spindle shaped myofibroblasts to dendritic keratocyte-like cells accompanied by significant upregulation of corneal crystallins and suppression of myofibroblast markers regardless of TGFβ1 treatment. RCFs cultured on soft hydrogels or patterned substrates exhibited elevated expression of α-smooth muscle actin (αSMA) in the presence of 17AAG. Treatment of hTCEpi cells disrupted zonula occludens 1 (ZO-1) adherens junction formation. In vivo, there were no differences detected in nearly all clinical parameters assessed between treatment groups. However, rabbits treated with 17AAG developed greater stromal haze formation compared with controls, irrespective of frequency of administration. Lastly, there was increased αSMA positive myofibroblasts in the stroma of 17AAG treated animals when compared with controls. Hsp90 inhibition promoted reversion of the myofibroblast to keratocyte phenotype, although this only occurred on rigid substrates. By contrast, in vivo Hsp90 inhibition was detrimental to corneal wound healing likely due to impairment in corneal epithelial closure and barrier function restoration. Collectively, our data demonstrated a strong interplay in vitro between biophysical cues and soluble signaling molecules in determining corneal stromal cell phenotype.
Collapse
Affiliation(s)
- VijayKrishna Raghunathan
- Department of Basic Sciences, United States; The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, United States.
| | - Sydney Garrison Edwards
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Alexander T Evashenk
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Yeonju Song
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Eva Rewinski
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Ariana Marangakis Price
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Alyssa Hoehn
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Connor Chang
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Christopher M Reilly
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Santoshi Muppala
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States; Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, United States; Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States.
| |
Collapse
|
24
|
Sakaguchi K, Takahashi H, Tobe Y, Sasaki D, Matsuura K, Iwasaki K, Shimizu T, Umezu M. Measuring the Contractile Force of Multilayered Human Cardiac Cell Sheets. Tissue Eng Part C Methods 2020; 26:485-492. [PMID: 32799760 DOI: 10.1089/ten.tec.2020.0164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three-dimensional (3D) cardiac tissue reconstruction using tissue engineering technology is a rapidly growing area of regenerative medicine and drug screening development. However, there remains an urgent need for the development of a method capable of accurately measuring the contractile force of physiologically relevant 3D myocardial tissues to facilitate the prediction of human heart tissue drug sensitivity. To this end, our laboratory has developed a novel drug screening model that measures the contractile force of cardiac cell sheets prepared using temperature-responsive culture dishes. To circumvent the difficulties that commonly arise during the stacking of cardiomyocyte sheets, we established a stacking method using centrifugal force, making it possible to measure 3D myocardial tissue. Human induced pluripotent stem cell-derived cardiomyocytes were seeded in a temperature-responsive culture dish and processed into a sheet. The cardiac cell sheets were multilayered to construct 3D cardiac tissue. Measurement of the contractile force and cross-sectional area of the multilayered 3D cardiac tissue were then obtained and used to determine the relationship between the cross-sectional area of the cardiac tissue and its contractile force. The contractile force of the 1-, 3-, and 5-layer tissues increased linearly in proportion to the cross-sectional area. A result of 6.4 mN/mm2, accounting for one-seventh of the contractile force found in adult tissue, was obtained. However, with 7-layer tissues, there was a sudden drop in the contractile force, possibly because of limited oxygen and nutrient supply. In conclusion, we established a method wherein the thickness of the cell sheets was controlled through layering, thus enabling accurate evaluation of the cardiac contractile function. This method may enable comparisons with living heart tissue while providing information applicable to regenerative medicine and drug screening models.
Collapse
Affiliation(s)
- Katsuhisa Sakaguchi
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Hiroaki Takahashi
- Department of Modern Mechanical Engineering, School of Creative Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Yusuke Tobe
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Daisuke Sasaki
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Kiyotaka Iwasaki
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, Tokyo, Japan.,Department of Modern Mechanical Engineering, School of Creative Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Mitsuo Umezu
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, Tokyo, Japan.,Department of Modern Mechanical Engineering, School of Creative Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| |
Collapse
|
25
|
Electrical stimulation induces differentiation of human cardiosphere-derived cells (hCDCs) to committed cardiomyocyte. Mol Cell Biochem 2020; 470:29-39. [PMID: 32388790 DOI: 10.1007/s11010-020-03742-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/02/2020] [Indexed: 10/24/2022]
Abstract
Logistic complexities of heart transplantation embossed the necessity of utilizing novel methods, which enable heart regeneration. Human cardiosphere-derived cells (hCDCs) are taken into consideration as a promising cell resource in cell therapy in recent years. In this study, we designed an electrochemical stimulation system, which sends square pulses to the hCDCs and records their electrical response. Morphology, viability and differentiation of hCDCs are monitored at certain time courses of the treatment. Differentiating hCDCs aligned perpendicularly with respect to the direction of applied electric current, and obtained a spindle-like morphology, while they remained viable. At the same time, specific cardiac marker genes including GATA4, cTnT and α-MHC showed a considerable up-regulation. Our findings confirm that hCDCs differentiate to committed cardiomyocytes when hCDCs receive an electrical energy of 0.06 - 0.12 Wh. This amount of electrical energy could be applied to the stem cells using versatile electrical stimulation patterns via commercially available devices.
Collapse
|
26
|
Walter T, Gruenewald A, Detsch R, Boccaccini AR, Vogel N. Cell Interactions with Size-Controlled Colloidal Monolayers: Toward Improved Coatings in Bone Tissue Engineering. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:1793-1803. [PMID: 32017853 DOI: 10.1021/acs.langmuir.9b03308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The surface structure of biomaterials is of key importance to control its interactions with biological environments. Industrial fabrication and coating processes often introduce particulate nanostructures at implant surfaces. Understanding the cellular interaction with particle-based surface topologies and feature sizes in the colloidal length scale therefore offers the possibility to improve the biological response of synthetic biomaterials. Here, surfaces with controlled topography and regular feature sizes covering the relevant length scale of particulate coatings (100-1000 nm) are fabricated by colloidal templating. Using fluorescent microscopy, WST assay, and morphology analysis, results show that adhesion and attachment of bone-marrow derived murine stromal cells (ST2) are strongly influenced by the surface feature size while geometric details play an insignificant role. Quantitative analysis shows enhanced cell adhesion, spreading, viability, and activity when surface feature size decreases below 200 nm compared to flat surfaces, while larger feature sizes are detrimental to cell adhesion. Kinetic studies reveal that most cells on surfaces with larger features lose contact with the substrate over time. This study identifies colloidal templating as a simple method for creating highly defined model systems to investigate complex cell functions and provides design criteria for the choice of particulate coatings on commercial implant materials.
Collapse
Affiliation(s)
- Teresa Walter
- Institute of Particle Technology , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 4 , 91058 Erlangen , Germany
| | - Alina Gruenewald
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Rainer Detsch
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Nicolas Vogel
- Institute of Particle Technology , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 4 , 91058 Erlangen , Germany
| |
Collapse
|
27
|
Abasi S, Aggas JR, Venkatesh N, Vallavanatt IG, Guiseppi-Elie A. Design, fabrication and testing of an electrical cell stimulation and recording apparatus (ECSARA) for cells in electroculture. Biosens Bioelectron 2020; 147:111793. [DOI: 10.1016/j.bios.2019.111793] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
|
28
|
Yang L, Gao Q, Ge L, Zhou Q, Warszawik EM, Bron R, Lai KWC, van Rijn P. Topography induced stiffness alteration of stem cells influences osteogenic differentiation. Biomater Sci 2020; 8:2638-2652. [DOI: 10.1039/d0bm00264j] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Topography-driven alterations to single cell stiffness rather than alterations in cell morphology, is the underlying driver for influencing cell biological processes, particularly stem cell differentiation.
Collapse
Affiliation(s)
- Liangliang Yang
- Department of Biomedical Engineering-FB40
- University of Groningen
- University Medical Center Groningen
- 9713 AV Groningen
- The Netherlands
| | - Qi Gao
- Department of Biomedical Engineering
- City University of Hong Kong
- Hong Kong
| | - Lu Ge
- Department of Biomedical Engineering-FB40
- University of Groningen
- University Medical Center Groningen
- 9713 AV Groningen
- The Netherlands
| | - Qihui Zhou
- Institute for Translational Medicine
- Department of Stomatology
- The Affiliated Hospital of Qingdao University
- Qingdao University
- Qingdao 266003
| | - Eliza M. Warszawik
- Department of Biomedical Engineering-FB40
- University of Groningen
- University Medical Center Groningen
- 9713 AV Groningen
- The Netherlands
| | - Reinier Bron
- Department of Biomedical Engineering-FB40
- University of Groningen
- University Medical Center Groningen
- 9713 AV Groningen
- The Netherlands
| | - King Wai Chiu Lai
- Department of Biomedical Engineering
- City University of Hong Kong
- Hong Kong
| | - Patrick van Rijn
- Department of Biomedical Engineering-FB40
- University of Groningen
- University Medical Center Groningen
- 9713 AV Groningen
- The Netherlands
| |
Collapse
|
29
|
Song SY, Yoo J, Go S, Hong J, Sohn HS, Lee JR, Kang M, Jeong GJ, Ryu S, Kim SHL, Hwang NS, Char K, Kim BS. Cardiac-mimetic cell-culture system for direct cardiac reprogramming. Theranostics 2019; 9:6734-6744. [PMID: 31660065 PMCID: PMC6815967 DOI: 10.7150/thno.35574] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Rationale: Cardiovascular diseases often cause substantial heart damage and even heart failure due to the limited regenerative capacity of adult cardiomyocytes. The direct cardiac reprogramming of fibroblasts could be a promising therapeutic option for these patients. Although exogenous transcriptional factors can induce direct cardiac reprogramming, the reprogramming efficiency is too low to be used clinically. Herein, we introduce a cardiac-mimetic cell-culture system that resembles the microenvironment in the heart and provides interactions with cardiomyocytes and electrical cues to the cultured fibroblasts for direct cardiac reprogramming. Methods: Nano-thin and nano-porous membranes and heart like electric stimulus were used in the cardiac-mimetic cell-culture system. The human neonatal dermal fibroblasts containing cardiac transcription factors were plated on the membrane and cultured with the murine cardiomyocyte in the presence of the electric stimulus. The reprogramming efficiency was evaluated by qRT-PCR and immunocytochemistry. Results: Nano-thin and nano-porous membranes in the culture system facilitated interactions between fibroblasts and cardiomyocytes in coculture. The cellular interactions and electric stimulation supplied by the culture system dramatically enhanced the cardiac reprogramming efficiency of cardiac-specific transcriptional factor-transfected fibroblasts. Conclusion: The cardiac-mimetic culture system may serve as an effective tool for producing a feasible number of reprogrammed cardiomyocytes from fibroblasts.
Collapse
|
30
|
Villanueva P, Pereira S, Olmo A, Pérez P, Yuste Y, Yúfera A, Portilla F. Electrical pulse stimulation of skeletal myoblasts cell cultures with simulated action potentials. J Tissue Eng Regen Med 2019; 13:1265-1269. [DOI: 10.1002/term.2869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/08/2019] [Accepted: 03/21/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Paula Villanueva
- Instituto de Biomedicina de Sevilla (IBIS)Campus Hospital Universitario Virgen del Rocío Sevilla Spain
| | - Sheila Pereira
- Instituto de Biomedicina de Sevilla (IBIS)Campus Hospital Universitario Virgen del Rocío Sevilla Spain
| | - Alberto Olmo
- Instituto de Microelectrónica de SevillaIMSE, CNM (CSIC, Universidad de Sevilla) Sevilla Spain
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología ElectrónicaUniversidad de Sevilla Sevilla Spain
| | - Pablo Pérez
- Instituto de Microelectrónica de SevillaIMSE, CNM (CSIC, Universidad de Sevilla) Sevilla Spain
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología ElectrónicaUniversidad de Sevilla Sevilla Spain
| | - Yaiza Yuste
- Instituto de Biomedicina de Sevilla (IBIS)Campus Hospital Universitario Virgen del Rocío Sevilla Spain
| | - Alberto Yúfera
- Instituto de Microelectrónica de SevillaIMSE, CNM (CSIC, Universidad de Sevilla) Sevilla Spain
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología ElectrónicaUniversidad de Sevilla Sevilla Spain
| | - Fernando Portilla
- Instituto de Biomedicina de Sevilla (IBIS)Campus Hospital Universitario Virgen del Rocío Sevilla Spain
| |
Collapse
|
31
|
Dwenger M, Kowalski WJ, Ye F, Yuan F, Tinney JP, Setozaki S, Nakane T, Masumoto H, Campbell P, Guido W, Keller BB. Chronic optical pacing conditioning of h-iPSC engineered cardiac tissues. J Tissue Eng 2019; 10:2041731419841748. [PMID: 31024681 PMCID: PMC6472158 DOI: 10.1177/2041731419841748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
The immaturity of human induced pluripotent stem cell derived engineered cardiac
tissues limits their ability to regenerate damaged myocardium and to serve as
robust in vitro models for human disease and drug toxicity
studies. Several chronic biomimetic conditioning protocols, including mechanical
stretch, perfusion, and/or electrical stimulation promote engineered cardiac
tissue maturation but have significant technical limitations. Non-contacting
chronic optical stimulation using heterologously expressed channelrhodopsin
light-gated ion channels, termed optogenetics, may be an advantageous
alternative to chronic invasive electrical stimulation for engineered cardiac
tissue conditioning. We designed proof-of-principle experiments to successfully
transfect human induced pluripotent stem cell derived engineered cardiac tissues
with a desensitization resistant, chimeric channelrhodopsin protein, and then
optically paced engineered cardiac tissues to accelerate maturation. We
transfected human induced pluripotent stem cell engineered cardiac tissues using
an adeno-associated virus packaged chimeric channelrhodopsin and then verified
optically paced by whole cell patch clamp. Engineered cardiac tissues were then
chronically optically paced above their intrinsic beat rates in
vitro from day 7 to 14. Chronically optically paced resulted in
improved engineered cardiac tissue electrophysiological properties and subtle
changes in the expression of some cardiac relevant genes, though active force
generation and histology were unchanged. These results validate the feasibility
of a novel chronically optically paced paradigm to explore non-invasive and
scalable optically paced–induced engineered cardiac tissue maturation
strategies.
Collapse
Affiliation(s)
- Marc Dwenger
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William J Kowalski
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA.,Laboratory of Stem Cell and Neurovascular Biology, Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fei Ye
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Fangping Yuan
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Joseph P Tinney
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Shuji Setozaki
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cardiovascular Surgery, Okamura Memorial Hospital, Shimizu, Japan
| | - Takeichiro Nakane
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,RIKEN Center for Biosystems Dynamics Research (BDR), Wako, Japan
| | - Peter Campbell
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Bradley B Keller
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
32
|
Ashtari K, Nazari H, Ko H, Tebon P, Akhshik M, Akbari M, Alhosseini SN, Mozafari M, Mehravi B, Soleimani M, Ardehali R, Ebrahimi Warkiani M, Ahadian S, Khademhosseini A. Electrically conductive nanomaterials for cardiac tissue engineering. Adv Drug Deliv Rev 2019; 144:162-179. [PMID: 31176755 PMCID: PMC6784829 DOI: 10.1016/j.addr.2019.06.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 01/26/2023]
Abstract
Patient deaths resulting from cardiovascular diseases are increasing across the globe, posing the greatest risk to patients in developed countries. Myocardial infarction, as a result of inadequate blood flow to the myocardium, results in irreversible loss of cardiomyocytes which can lead to heart failure. A sequela of myocardial infarction is scar formation that can alter the normal myocardial architecture and result in arrhythmias. Over the past decade, a myriad of tissue engineering approaches has been developed to fabricate engineered scaffolds for repairing cardiac tissue. This paper highlights the recent application of electrically conductive nanomaterials (carbon and gold-based nanomaterials, and electroactive polymers) to the development of scaffolds for cardiac tissue engineering. Moreover, this work summarizes the effects of these nanomaterials on cardiac cell behavior such as proliferation and migration, as well as cardiomyogenic differentiation in stem cells.
Collapse
Affiliation(s)
- Khadijeh Ashtari
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran; Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hojjatollah Nazari
- Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Stem Cell Technology Research Center, Tehran, Iran
| | - Hyojin Ko
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Masoud Akhshik
- Faculty of Forestry, University of Toronto, Toronto, Canada; Center for Biocomposites and Biomaterials Processing (CBBP), University of Toronto, Toronto, Canada; Shahdad Ronak Commercialization Company, Tehran, Iran
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, Canada; Center for Biomedical Research, University of Victoria, Victoria, Canada; Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, Canada
| | - Sanaz Naghavi Alhosseini
- Biomaterials Group, Department of Biomaterial Engineering, Amirkabir University of Technology, Tehran, Iran; Stem Cell Technology Research Center, Tehran, Iran
| | - Masoud Mozafari
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Bita Mehravi
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran; Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Faculty of Medical Sciences, Department of Hematology and Cell Therapy, Tarbiat Modares University, Tehran, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California - Los Angeles, USA
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia; Institute of Molecular Medicine, Sechenov University, Moscow, Russia
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA; Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, USA; Department of Radiology, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, USA.
| |
Collapse
|
33
|
Bloise N, Petecchia L, Ceccarelli G, Fassina L, Usai C, Bertoglio F, Balli M, Vassalli M, Cusella De Angelis MG, Gavazzo P, Imbriani M, Visai L. The effect of pulsed electromagnetic field exposure on osteoinduction of human mesenchymal stem cells cultured on nano-TiO2 surfaces. PLoS One 2018; 13:e0199046. [PMID: 29902240 PMCID: PMC6002089 DOI: 10.1371/journal.pone.0199046] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) are considered a great promise in the repair and regeneration of bone. Considerable efforts have been oriented towards uncovering the best strategy to promote stem cells osteogenic differentiation. In previous studies, hBM-MSCs exposed to physical stimuli such as pulsed electromagnetic fields (PEMFs) or directly seeded on nanostructured titanium surfaces (TiO2) were shown to improve their differentiation to osteoblasts in osteogenic condition. In the present study, the effect of a daily PEMF-exposure on osteogenic differentiation of hBM-MSCs seeded onto nanostructured TiO2 (with clusters under 100 nm of dimension) was investigated. TiO2-seeded cells were exposed to PEMF (magnetic field intensity: 2 mT; intensity of induced electric field: 5 mV; frequency: 75 Hz) and examined in terms of cell physiology modifications and osteogenic differentiation. Results showed that PEMF exposure affected TiO2-seeded cells osteogenesis by interfering with selective calcium-related osteogenic pathways, and greatly enhanced hBM-MSCs osteogenic features such as the expression of early/late osteogenic genes and protein production (e.g., ALP, COL-I, osteocalcin and osteopontin) and ALP activity. Finally, PEMF-treated cells resulted to secrete into conditioned media higher amounts of BMP-2, DCN and COL-I than untreated cell cultures. These findings confirm once more the osteoinductive potential of PEMF, suggesting that its combination with TiO2 nanostructured surface might be a great option in bone tissue engineering applications.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- * E-mail: (NB); (LV)
| | | | - Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (C.H.T.), University of Pavia, Pavia, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Federico Bertoglio
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
| | - Martina Balli
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Massimo Vassalli
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Maria Gabriella Cusella De Angelis
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Paola Gavazzo
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Marcello Imbriani
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- * E-mail: (NB); (LV)
| |
Collapse
|
34
|
Tijore A, Irvine SA, Sarig U, Mhaisalkar P, Baisane V, Venkatraman S. Contact guidance for cardiac tissue engineering using 3D bioprinted gelatin patterned hydrogel. Biofabrication 2018; 10:025003. [PMID: 29235444 DOI: 10.1088/1758-5090/aaa15d] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Here, we have developed a 3D bioprinted microchanneled gelatin hydrogel that promotes human mesenchymal stem cell (hMSC) myocardial commitment and supports native cardiomyocytes (CMs) contractile functionality. Firstly, we studied the effect of bioprinted microchanneled hydrogel on the alignment, elongation, and differentiation of hMSC. Notably, the cells displayed well defined F-actin anisotropy and elongated morphology on the microchanneled hydrogel, hence showing the effects of topographical control over cell behavior. Furthermore, the aligned stem cells showed myocardial lineage commitment, as detected using mature cardiac markers. The fluorescence-activated cell sorting analysis also confirmed a significant increase in the commitment towards myocardial tissue lineage. Moreover, seeded CMs were found to be more aligned and demonstrated synchronized beating on microchanneled hydrogel as compared to the unpatterned hydrogel. Overall, our study proved that microchanneled hydrogel scaffold produced by 3D bioprinting induces myocardial differentiation of stem cells as well as supports CMs growth and contractility. Applications of this approach may be beneficial for generating in vitro cardiac model systems to physiological and cardiotoxicity studies as well as in vivo generating custom designed cell impregnated constructs for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Ajay Tijore
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore
| | | | | | | | | | | |
Collapse
|
35
|
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: 165] [Impact Index Per Article: 27.5] [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
|
36
|
Ren J, Xu Q, Chen X, Li W, Guo K, Zhao Y, Wang Q, Zhang Z, Peng H, Li YG. Superaligned Carbon Nanotubes Guide Oriented Cell Growth and Promote Electrophysiological Homogeneity for Synthetic Cardiac Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1702713. [PMID: 29024059 DOI: 10.1002/adma.201702713] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/19/2017] [Indexed: 05/21/2023]
Abstract
Cardiac engineering of patches and tissues is a promising option to restore infarcted hearts, by seeding cardiac cells onto scaffolds and nurturing their growth in vitro. However, current patches fail to fully imitate the hierarchically aligned structure in the natural myocardium, the fast electrotonic propagation, and the subsequent synchronized contractions. Here, superaligned carbon-nanotube sheets (SA-CNTs) are explored to culture cardiomyocytes, mimicking the aligned structure and electrical-impulse transmission behavior of the natural myocardium. The SA-CNTs not only induce an elongated and aligned cell morphology of cultured cardiomyocytes, but also provide efficient extracellular signal-transmission pathways required for regular and synchronous cell contractions. Furthermore, the SA-CNTs can reduce the beat-to-beat and cell-to-cell dispersion in repolarization of cultured cells, which is essential for a normal beating rhythm, and potentially reduce the occurrence of arrhythmias. Finally, SA-CNT-based flexible one-piece electrodes demonstrate a multipoint pacing function. These combined high properties make SA-CNTs promising in applications in cardiac resynchronization therapy in patients with heart failure and following myocardial infarctions.
Collapse
Affiliation(s)
- Jing Ren
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Quanfu Xu
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaomeng Chen
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Wei Li
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kai Guo
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yang Zhao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Qian Wang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhitao Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Yi-Gang Li
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
37
|
Unraveling the mechanistic effects of electric field stimulation towards directing stem cell fate and function: A tissue engineering perspective. Biomaterials 2017; 150:60-86. [PMID: 29032331 DOI: 10.1016/j.biomaterials.2017.10.003] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
Electric field (EF) stimulation can play a vital role in eliciting appropriate stem cell response. Such an approach is recently being established to guide stem cell differentiation through osteogenesis/neurogenesis/cardiomyogenesis. Despite significant recent efforts, the biophysical mechanisms by which stem cells sense, interpret and transform electrical cues into biochemical and biological signals still remain unclear. The present review critically analyses the variety of EF stimulation approaches that can be employed to evoke appropriate stem cell response and also makes an attempt to summarize the underlying concepts of this notion, placing special emphasis on stem cell based tissue engineering and regenerative medicine. This review also discusses the major signaling pathways and cellular responses that are elicited by electric stimulation, including the participation of reactive oxygen species and heat shock proteins, modulation of intracellular calcium ion concentration, ATP production and numerous other events involving the clustering or reassembling of cell surface receptors, cytoskeletal remodeling and so on. The specific advantages of using external electric stimulation in different modalities to regulate stem cell fate processes are highlighted with explicit examples, in vitro and in vivo.
Collapse
|
38
|
Zhou Q, Castañeda Ocampo O, Guimarães CF, Kühn PT, van Kooten TG, van Rijn P. Screening Platform for Cell Contact Guidance Based on Inorganic Biomaterial Micro/nanotopographical Gradients. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31433-31445. [PMID: 28825457 PMCID: PMC5609122 DOI: 10.1021/acsami.7b08237] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/21/2017] [Indexed: 05/19/2023]
Abstract
High-throughput screening (HTS) methods based on topography gradients or arrays have been extensively used to investigate cell-material interactions. However, it is a huge technological challenge to cost efficiently prepare topographical gradients of inorganic biomaterials due to their inherent material properties. Here, we developed a novel strategy translating PDMS-based wrinkled topography gradients with amplitudes from 49 to 2561 nm and wavelengths between 464 and 7121 nm to inorganic biomaterials (SiO2, Ti/TiO2, Cr/CrO3, and Al2O3) which are frequently used clinical materials. Optimal substratum conditions promoted human bone-marrow derived mesenchymal stem cell alignment, elongation, cytoskeleton arrangement, filopodia development as well as cell adhesion in vitro, which depended both on topography and interface material. This study displays a positive correlation between cell alignment and the orientation of cytoskeleton, filopodia, and focal adhesions. This platform vastly minimizes the experimental efforts both for inorganic material interface engineering and cell biological assessments in a facile and effective approach. The practical application of the HTS technology is expected to aid in the acceleration of developments of inorganic clinical biomaterials.
Collapse
Affiliation(s)
- Qihui Zhou
- Department of Biomedical
Engineering—FB40, University of Groningen,
University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff
Institute for Biomedical Engineering and Materials Science—FB41, University of Groningen, University Medical Center
Groningen, Groningen,
A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Olga Castañeda Ocampo
- Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
- Stratingh Institute for Chemistry, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Carlos F. Guimarães
- Department of Biomedical
Engineering—FB40, University of Groningen,
University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Philipp T. Kühn
- Department of Biomedical
Engineering—FB40, University of Groningen,
University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff
Institute for Biomedical Engineering and Materials Science—FB41, University of Groningen, University Medical Center
Groningen, Groningen,
A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Theo G. van Kooten
- Department of Biomedical
Engineering—FB40, University of Groningen,
University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff
Institute for Biomedical Engineering and Materials Science—FB41, University of Groningen, University Medical Center
Groningen, Groningen,
A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Patrick van Rijn
- Department of Biomedical
Engineering—FB40, University of Groningen,
University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff
Institute for Biomedical Engineering and Materials Science—FB41, University of Groningen, University Medical Center
Groningen, Groningen,
A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| |
Collapse
|
39
|
Zhu S, Jing W, Hu X, Huang Z, Cai Q, Ao Y, Yang X. Time-dependent effect of electrical stimulation on osteogenic differentiation of bone mesenchymal stromal cells cultured on conductive nanofibers. J Biomed Mater Res A 2017; 105:3369-3383. [PMID: 28795778 DOI: 10.1002/jbm.a.36181] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/28/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022]
Abstract
Bone tissue engineering using bone mesenchymal stromal cells (BMSCs) is a multidisciplinary strategy that requires biodegradable scaffold, cell, various promoting cues to work simultaneously. Electrical stimulation (ES) is known able to promote osteogenic differentiation of BMSCs, but it is interesting to know how can it play the strongest promotion effect. To strengthen local ES on BMSCs, parallel-aligned conductive nanofibers were electrospun from the mixtures of poly(L-lactide) (PLLA) and multi-walled carbon nanotubes (MWCNTs), and used for cell culture. Osteogenic differentiation of BMSCs was conducted by applying ES (direct current, 1.5 V, 1.5 h/day) perpendicular to the fiber direction during the day 1-7, day 8-14, or day 15-21 period of the osteoinductive culture. In comparison with ES-free groups, bone-related markers and genes were found significantly up-regulated when ES was applied on BMSCs growing on nanofibers having higher conductivity. When the ES was applied at the earlier stage of osteoinductive culture, the promotion effect on osteogenic differentiation would be stronger. In the presence of a BMP blocker, the down-regulated expressions of bone-related genes were able to be slightly recovered by ES, especially when the ES was applied at the beginning of osteoinductive culture (i.e. day 1-7). The promotion effect generated by ES in the early stage was found sustainable to later stages of differentiation, while those ES applied at later stages of differentiation should have missed the optimal point. In other words, later ES was not so necessary in inducing the osteogenic differentiation of BMSCs. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3369-3383, 2017.
Collapse
Affiliation(s)
- Siqi Zhu
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Wei Jing
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Zirong Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Xiaoping Yang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
40
|
Park SJ, Kim KH, Jeon WY, Seo J, Han JM, Kim JS, Chung HM, Lee JH, Moon SH, Kim HH. Enzyme catalyzed electrostimulation of human embryonic stem cell-derived cardiomyocytes influence contractility and synchronization. Biochem Eng J 2017. [DOI: 10.1016/j.bej.2017.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
41
|
Sadeghi AH, Shin SR, Deddens JC, Fratta G, Mandla S, Yazdi IK, Prakash G, Antona S, Demarchi D, Buijsrogge MP, Sluijter JPG, Hjortnaes J, Khademhosseini A. Engineered 3D Cardiac Fibrotic Tissue to Study Fibrotic Remodeling. Adv Healthc Mater 2017; 6:10.1002/adhm.201601434. [PMID: 28498548 PMCID: PMC5545804 DOI: 10.1002/adhm.201601434] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/02/2017] [Indexed: 12/19/2022]
Abstract
Activation of cardiac fibroblasts into myofibroblasts is considered to play an essential role in cardiac remodeling and fibrosis. A limiting factor in studying this process is the spontaneous activation of cardiac fibroblasts when cultured on two-dimensional (2D) culture plates. In this study, a simplified three-dimensional (3D) hydrogel platform of contractile cardiac tissue, stimulated by transforming growth factor-β1 (TGF-β1), is presented to recapitulate a fibrogenic microenvironment. It is hypothesized that the quiescent state of cardiac fibroblasts can be maintained by mimicking the mechanical stiffness of native heart tissue. To test this hypothesis, a 3D cell culture model consisting of cardiomyocytes and cardiac fibroblasts encapsulated within a mechanically engineered gelatin methacryloyl hydrogel, is developed. The study shows that cardiac fibroblasts maintain their quiescent phenotype in mechanically tuned hydrogels. Additionally, treatment with a beta-adrenergic agonist increases beating frequency, demonstrating physiologic-like behavior of the heart constructs. Subsequently, quiescent cardiac fibroblasts within the constructs are activated by the exogenous addition of TGF-β1. The expression of fibrotic protein markers (and the functional changes in mechanical stiffness) in the fibrotic-like tissues are analyzed to validate the model. Overall, this 3D engineered culture model of contractile cardiac tissue enables controlled activation of cardiac fibroblasts, demonstrating the usability of this platform to study fibrotic remodeling.
Collapse
Affiliation(s)
- Amir Hossein Sadeghi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Department of Cardiology, University Medical Center Utrecht, 3584, CX, Utrecht, The Netherlands
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, 3584, CX, The Netherlands
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Janine C Deddens
- Department of Cardiology, University Medical Center Utrecht, 3584, CX, Utrecht, The Netherlands
- Netherlands Heart Institute (ICIN), 3584, CX, Utrecht, The Netherlands
| | - Giuseppe Fratta
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129, Torino, Italy
| | - Serena Mandla
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Iman K Yazdi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Gyan Prakash
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Silvia Antona
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129, Torino, Italy
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129, Torino, Italy
| | - Marc P Buijsrogge
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, 3584, CX, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, University Medical Center Utrecht, 3584, CX, Utrecht, The Netherlands
- Netherlands Heart Institute (ICIN), 3584, CX, Utrecht, The Netherlands
- UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, 3584, CX, Utrecht, The Netherlands
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, 3584, CX, The Netherlands
- UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, 3584, CX, Utrecht, The Netherlands
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 65 Landsdowne Street, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, 130-701, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| |
Collapse
|
42
|
Gouveia PJ, Rosa S, Ricotti L, Abecasis B, Almeida HV, Monteiro L, Nunes J, Carvalho FS, Serra M, Luchkin S, Kholkin AL, Alves PM, Oliveira PJ, Carvalho R, Menciassi A, das Neves RP, Ferreira LS. Flexible nanofilms coated with aligned piezoelectric microfibers preserve the contractility of cardiomyocytes. Biomaterials 2017. [PMID: 28622605 DOI: 10.1016/j.biomaterials.2017.05.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The use of engineered cardiac tissue for high-throughput drug screening/toxicology assessment remains largely unexplored. Here we propose a scaffold that mimics aspects of cardiac extracellular matrix while preserving the contractility of cardiomyocytes. The scaffold is based on a poly(caprolactone) (PCL) nanofilm with magnetic properties (MNF, standing for magnetic nanofilm) coated with a layer of piezoelectric (PIEZO) microfibers of poly(vinylidene fluoride-trifluoroethylene) (MNF+PIEZO). The nanofilm creates a flexible support for cell contraction and the aligned PIEZO microfibers deposited on top of the nanofilm creates conditions for cell alignment and electrical stimulation of the seeded cells. Our results indicate that MNF+PIEZO scaffold promotes rat and human cardiac cell attachment and alignment, maintains the ratio of cell populations overtime, promotes cell-cell communication and metabolic maturation, and preserves cardiomyocyte (CM) contractility for at least 12 days. The engineered cardiac construct showed high toxicity against doxorubicin, a cardiotoxic molecule, and responded to compounds that modulate CM contraction such as epinephrine, propranolol and heptanol.
Collapse
Affiliation(s)
- P José Gouveia
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - S Rosa
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - L Ricotti
- The BioRobotics Institute, Scuola Superiore Sant' Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera (PI), Italy
| | - B Abecasis
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - H V Almeida
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - L Monteiro
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - J Nunes
- Center for Mechanical Engineering, University of Coimbra, 3030-788 Coimbra, Portugal
| | - F Sofia Carvalho
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - M Serra
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - S Luchkin
- CICECO - Materials Institute of Aveiro & Physics Department, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - A Leonidovitch Kholkin
- CICECO - Materials Institute of Aveiro & Physics Department, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; School of Natural Sciences and Mathematics, Ural Federal University, 620000 Ekaterinburg, Russia
| | - P Marques Alves
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - P Jorge Oliveira
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - R Carvalho
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - A Menciassi
- The BioRobotics Institute, Scuola Superiore Sant' Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera (PI), Italy
| | - R Pires das Neves
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - L Silva Ferreira
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
43
|
Monteiro LM, Vasques-Nóvoa F, Ferreira L, Pinto-do-Ó P, Nascimento DS. Restoring heart function and electrical integrity: closing the circuit. NPJ Regen Med 2017; 2:9. [PMID: 29302345 PMCID: PMC5665620 DOI: 10.1038/s41536-017-0015-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/19/2017] [Accepted: 03/06/2017] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular diseases are the main cause of death in the world and are often associated with the occurrence of arrhythmias due to disruption of myocardial electrical integrity. Pathologies involving dysfunction of the specialized cardiac excitatory/conductive tissue are also common and constitute an added source of morbidity and mortality since current standard therapies withstand a great number of limitations. As electrical integrity is essential for a well-functioning heart, innovative strategies have been bioengineered to improve heart conduction and/or promote myocardial repair, based on: (1) gene and/or cell delivery; or (2) conductive biomaterials as tools for cardiac tissue engineering. Herein we aim to review the state-of-art in the area, while briefly describing the biological principles underlying the heart electrical/conduction system and how this system can be disrupted in heart disease. Suggestions regarding targets for future studies are also presented.
Collapse
Affiliation(s)
- Luís Miguel Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- CNC—Center for Neuroscience and Cell Biology, Universidade de Coimbra, Coimbra, Portugal
| | - Francisco Vasques-Nóvoa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Departamento de Fisiologia e Cirurgia Cardiotorácica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Lino Ferreira
- CNC—Center for Neuroscience and Cell Biology, Universidade de Coimbra, Coimbra, Portugal
| | - Perpétua Pinto-do-Ó
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Diana Santos Nascimento
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
44
|
Kim MS, Lee B, Kim HN, Bang S, Yang HS, Kang SM, Suh KY, Park SH, Jeon NL. 3D tissue formation by stacking detachable cell sheets formed on nanofiber mesh. Biofabrication 2017; 9:015029. [PMID: 28332479 DOI: 10.1088/1758-5090/aa64a0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We present a novel approach for assembling 3D tissue by layer-by-layer stacking of cell sheets formed on aligned nanofiber mesh. A rigid frame was used to repeatedly collect aligned electrospun PCL (polycaprolactone) nanofiber to form a mesh structure with average distance between fibers 6.4 µm. When human umbilical vein endothelial cells (HUVECs), human foreskin dermal fibroblasts, and skeletal muscle cells (C2C12) were cultured on the nanofiber mesh, they formed confluent monolayers and could be handled as continuous cell sheets with areas 3 × 3 cm2 or larger. Thicker 3D tissues have been formed by stacking multiple cell sheets collected on frames that can be nested (i.e. Matryoshka dolls) without any special tools. When cultured on the nanofiber mesh, skeletal muscle, C2C12 cells oriented along the direction of the nanofibers and differentiated into uniaxially aligned multinucleated myotube. Myotube cell sheets were stacked (upto 3 layers) in alternating or aligned directions to form thicker tissue with ∼50 µm thickness. Sandwiching HUVEC cell sheets with two dermal fibroblast cell sheets resulted in vascularized 3D tissue. HUVECs formed extensive networks and expressed CD31, a marker of endothelial cells. Cell sheets formed on nanofiber mesh have a number of advantages, including manipulation and stacking of multiple cell sheets for constructing 3D tissue and may find applications in a variety of tissue engineering applications.
Collapse
Affiliation(s)
- Min Sung Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kofron CM, Mende U. In vitro models of the cardiac microenvironment to study myocyte and non-myocyte crosstalk: bioinspired approaches beyond the polystyrene dish. J Physiol 2017; 595:3891-3905. [PMID: 28116799 DOI: 10.1113/jp273100] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022] Open
Abstract
The heart is a complex pluricellular organ composed of cardiomyocytes and non-myocytes including fibroblasts, endothelial cells and immune cells. Myocytes are responsible for electrical conduction and contractile force generation, while the other cell types are responsible for matrix deposition, vascularization, and injury response. Myocytes and non-myocytes are known to communicate and exert mutual regulatory effects. In concert, they determine the structural, electrical and mechanical characteristics in the healthy and remodelled myocardium. Dynamic crosstalk between myocytes and non-myocytes plays a crucial role in stress/injury-induced hypertrophy and fibrosis development that can ultimately lead to heart failure and arrhythmias. Investigations of heterocellular communication in the myocardium are hampered by the intricate interspersion of the different cell types and the complexity of the tissue architecture. In vitro models have facilitated investigations of cardiac cells in a direct and controllable manner and have provided important functional and mechanistic insights. However, these cultures often lack regulatory input from the other cell types as well as additional topographical, electrical, mechanical and biochemical cues from the cardiac microenvironment that all contribute to modulating cell differentiation, maturation, alignment, function and survival. Advancements in the development of more complex pluricellular physiological platforms that incorporate diverse cues from the myocardial microenvironment are expected to lead to more physiologically relevant cardiac tissue-like in vitro models for mechanistic biological research, disease modelling, therapeutic target identification, drug testing and regeneration.
Collapse
Affiliation(s)
- Celinda M Kofron
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
46
|
Atmanli A, Domian IJ. Recreating the Cardiac Microenvironment in Pluripotent Stem Cell Models of Human Physiology and Disease. Trends Cell Biol 2016; 27:352-364. [PMID: 28007424 DOI: 10.1016/j.tcb.2016.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/18/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
The advent of human pluripotent stem cell (hPSC) biology has opened unprecedented opportunities for the use of tissue engineering to generate human cardiac tissue for in vitro study. Engineering cardiac constructs that recapitulate human development and disease requires faithful recreation of the cardiac niche in vitro. Here we discuss recent progress in translating the in vivo cardiac microenvironment into PSC models of the human heart. We review three key physiologic features required to recreate the cardiac niche and facilitate normal cardiac differentiation and maturation: the biochemical, biophysical, and bioelectrical signaling cues. Finally, we discuss key barriers that must be overcome to fulfill the promise of stem cell biology in preclinical applications and ultimately in clinical practice.
Collapse
Affiliation(s)
- Ayhan Atmanli
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Ibrahim John Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
47
|
Tallawi M, Dippold D, Rai R, D'Atri D, Roether J, Schubert D, Rosellini E, Engel F, Boccaccini A. Novel PGS/PCL electrospun fiber mats with patterned topographical features for cardiac patch applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:569-76. [DOI: 10.1016/j.msec.2016.06.083] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/24/2016] [Accepted: 06/25/2016] [Indexed: 10/21/2022]
|
48
|
Enhancement of neurite adhesion, alignment and elongation on conductive polypyrrole-poly(lactide acid) fibers with cell-derived extracellular matrix. Colloids Surf B Biointerfaces 2016; 149:217-225. [PMID: 27768911 DOI: 10.1016/j.colsurfb.2016.10.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 01/11/2023]
Abstract
Extracellular matrix (ECM) can promote peripheral nerve repair. In this study, a conductive fiber-film (CFF) with core-sheath structure and conductivity of ∼10Scm-1 was prepared by electrospinning of aligned poly(l-lactide acid) (PLLA) fibers and electrochemical deposition of polypyrole (PPy) nanoparticles. Then the multiple components of ECM, including laminin, fibronectin and collagen, were coated on the surface of CFF by culturing and lysing L929 cells to fabricate the bioactive scaffold of ECM-linked CFF (ECM-CFF). The electrical stimulation (ES) of 100mV/cm for 14days and 2h per day did not significantly decrease the conductivity of ECM-CFF. The results of PC12 cells test indicated that, cells adhesion rate, neurite-bearing cell rate and neurite alignment rate on ECM-CFF were ∼95%, ∼77%, ∼70%, respectively, significantly larger than the corresponding values on bare CFF (17%, 29% and 14%, respectively). The neurites length on ECM-CFF (∼79mm) was also larger than that on bare CFF (∼25mm). ES of 100mV/cm onto PC12 cells through ECM-CFF could significantly promote neurite extension in first 3days of the neurite growth. These results indicated that, the combination of ECM-CFF with ES could improve the nerve regeneration by encouraging neural-cell adhesion, neurite growth and extension.
Collapse
|
49
|
Hosseini V, Gantenbein S, Avalos Vizcarra I, Schoen I, Vogel V. Stretchable Silver Nanowire Microelectrodes for Combined Mechanical and Electrical Stimulation of Cells. Adv Healthc Mater 2016; 5:2045-54. [PMID: 27245368 DOI: 10.1002/adhm.201600045] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/09/2016] [Indexed: 01/09/2023]
Abstract
The use of stretchable electrodes interfaced with the human body has enabled a new frontier in biomedical engineering, and the miniaturization of such electrodes can allow for a more precise spatial control to monitor or stimulate tissues. The understanding of the response of cells or tissues to combined electromechanical stimulation, as made possible by stretchable electrodes, is essential to improve medical devices and therapies. Cheap to produce and easy to use platforms for in vitro cell studies are thus urgently needed. This study reports the successful implementation of silver nanowires (AgNWs) into an integrated miniaturized electromechanical stimulator, which is compatible with cell culture. The innovative steps include a lithography-based lift-off method to micropattern AgNWs onto an elastic silicone membrane. These stretchable microelectrodes are then integrated into a microfluidic device for cell culture, which enables the synchronous electromechanical stimulation of cells. In a proof-of-concept study, it is furthermore shown that fibroblasts respond uniquely to mechanical stretching, electrical stimulation, and combined electromechanical stimulations in terms of cell alignment and morphology, as well as by producing the extracellular matrix protein collagen. This proof-of-concept study illustrates the functionality and usability of these stretchable AgNWs microelectrodes for either basic research or future biomedical applications.
Collapse
Affiliation(s)
- Vahid Hosseini
- Laboratory of Applied Mechanobiology; Department of Health Sciences and Technology; ETH Zurich; Vladimir-Prelog-Weg 1-5 CH-8093 Zurich Switzerland
| | - Silvan Gantenbein
- Laboratory of Applied Mechanobiology; Department of Health Sciences and Technology; ETH Zurich; Vladimir-Prelog-Weg 1-5 CH-8093 Zurich Switzerland
| | - Ima Avalos Vizcarra
- Laboratory of Applied Mechanobiology; Department of Health Sciences and Technology; ETH Zurich; Vladimir-Prelog-Weg 1-5 CH-8093 Zurich Switzerland
| | - Ingmar Schoen
- Laboratory of Applied Mechanobiology; Department of Health Sciences and Technology; ETH Zurich; Vladimir-Prelog-Weg 1-5 CH-8093 Zurich Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology; Department of Health Sciences and Technology; ETH Zurich; Vladimir-Prelog-Weg 1-5 CH-8093 Zurich Switzerland
| |
Collapse
|
50
|
Richards DJ, Tan Y, Coyle R, Li Y, Xu R, Yeung N, Parker A, Menick DR, Tian B, Mei Y. Nanowires and Electrical Stimulation Synergistically Improve Functions of hiPSC Cardiac Spheroids. NANO LETTERS 2016; 16:4670-8. [PMID: 27328393 PMCID: PMC4994528 DOI: 10.1021/acs.nanolett.6b02093] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The advancement of human induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology has shown promising potential to provide a patient-specific, regenerative cell therapy strategy to treat cardiovascular disease. Despite the progress, the unspecific, underdeveloped phenotype of hiPSC-CMs has shown arrhythmogenic risk and limited functional improvements after transplantation. To address this, tissue engineering strategies have utilized both exogenous and endogenous stimuli to accelerate the development of hiPSC-CMs. Exogenous electrical stimulation provides a biomimetic pacemaker-like stimuli that has been shown to advance the electrical properties of tissue engineered cardiac constructs. Recently, we demonstrated that the incorporation of electrically conductive silicon nanowires to hiPSC cardiac spheroids led to advanced structural and functional development of hiPSC-CMs by improving the endogenous electrical microenvironment. Here, we reasoned that the enhanced endogenous electrical microenvironment of nanowired hiPSC cardiac spheroids would synergize with exogenous electrical stimulation to further advance the functional development of nanowired hiPSC cardiac spheroids. For the first time, we report that the combination of nanowires and electrical stimulation enhanced cell-cell junction formation, improved development of contractile machinery, and led to a significant decrease in the spontaneous beat rate of hiPSC cardiac spheroids. The advancements made here address critical challenges for the use of hiPSC-CMs in cardiac developmental and translational research and provide an advanced cell delivery vehicle for the next generation of cardiac repair.
Collapse
Affiliation(s)
- Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yang Li
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ruoyu Xu
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Nelson Yeung
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Arran Parker
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|