1
|
Esmaeili H, Zhang Y, Ravi K, Neff K, Zhu W, Migrino RQ, Park JG, Nikkhah M. Development of an electroconductive Heart-on-a-chip model to investigate cellular and molecular response of human cardiac tissue to gold nanomaterials. Biomaterials 2025; 320:123275. [PMID: 40138961 DOI: 10.1016/j.biomaterials.2025.123275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
To date, various strategies have been developed to construct biomimetic and functional in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs). Among these approaches, microfluidic-based Heart-on-a-chip (HOC) models are promising, as they enable the engineering of miniaturized, physiologically relevant in vitro cardiac tissues with precise control over cellular constituents and tissue architecture. Despite significant advancements, previously reported HOC models often lack the electroconductivity features of the native human myocardium. In this study, we developed a 3D electroconductive HOC (referred to as eHOC) model through the co-culture of isogenic hiPSC-derived cardiomyocytes (hiCMs) and cardiac fibroblasts (hiCFs), embedded within an electroconductive hydrogel scaffold in a microfluidic-based chip system. Functional and gene expression analyses demonstrated that, compared to non-conductive HOC, the eHOC model exhibited enhanced contractile functionality, improved calcium transients, and increased expression of structural and calcium handling genes. The eHOC model was further leveraged to investigate the underlying electroconduction-induced pathway(s) associated with cardiac tissue development through single-cell RNA sequencing (scRNA-seq). Notably, scRNA-seq analyses revealed a significant downregulation of a set of cardiac genes, associated with the fetal stage of heart development, as well as upregulation of sarcomere- and conduction-related genes within the eHOC model. Additionally, upregulation of the cardiac muscle contraction and motor protein pathways were observed in the eHOC model, consistent with enhanced contractile functionality of the engineered cardiac tissues. Comparison of scRNA-seq data from the 3D eHOC model with published datasets of adult human hearts demonstrated a similar expression pattern of fetal- and adult-like cardiac genes. Overall, this study provides a unique eHOC model with improved biomimcry and organotypic features, which could be potentially used for drug testing and discovery, as well as disease modeling applications.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Yining Zhang
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Kalpana Ravi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Keagan Neff
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85022, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
2
|
Van Tran V, Phung VD, Do HH. Morphological advances and innovations in conjugated polymer films for high-performance gas sensors. Talanta 2025; 292:127904. [PMID: 40073824 DOI: 10.1016/j.talanta.2025.127904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Conjugated polymers (CPs) are considered one of the most important gas-sensing materials due to their unique features, combining the benefits of both metals and semiconductors, along with their outstanding mechanical properties and excellent processability. However, CPs with conventional morphological structures, such as largely amorphous and bulky matrices, face limitations in practical applications because of their inferior charge transport characteristics, low surface area, and insufficient sensitivity. Therefore, the design and development of novel morphological nanostructures in CPs have attracted significant attention as a promising strategy for improving morphological and electrical characteristics, thereby enabling a considerable increase in the sensing performance of corresponding gas sensors. Numerous CP nanostructures have been developed and implemented for high-performance gas sensors. Highlighting the morphological advances and bottlenecks of these nanostructures is crucial for providing an overview of developing trends, potential strategies, and emerging areas for the future development of CP nanostructures in the field. In this regard, this study describes state-of-the-art CP nanostructures, emphasizing their attractive morphological and electrical characteristics to help readers and researchers better understand emerging trends, promising future directions, and key obstacles for the application of CP nanostructure-based gas sensors. The most crucial aspects of CP nanostructures, including advanced preparation techniques, morphological properties, and sensing characteristics, are discussed and assessed in detail. Moreover, development strategies and perspectives for achieving high sensing efficiency in CP nanostructure-based flexible and wearable sensors are summarized and emphasized.
Collapse
Affiliation(s)
- Vinh Van Tran
- Laboratory for Advanced Nanomaterials and Sustainable Energy Technologies, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, Viet Nam; Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City, Viet Nam.
| | - Viet-Duc Phung
- Institute of Fundamental and Applied Sciences, Duy Tan University, Ho Chi Minh City, Viet Nam; Faculty of Environmental and Chemical Engineering, Duy Tan University, Da Nang City, Viet Nam
| | - Ha Huu Do
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
3
|
Zhao J, Chen Y, Qin Y, Li Y, Lu X, Xie C. Adhesive and Conductive Hydrogels for the Treatment of Myocardial Infarction. Macromol Rapid Commun 2025; 46:e2400835. [PMID: 39803789 DOI: 10.1002/marc.202400835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/27/2024] [Indexed: 05/02/2025]
Abstract
Myocardial infarction (MI) is a leading cause of mortality among cardiovascular diseases. Following MI, the damaged myocardium is progressively being replaced by fibrous scar tissue, which exhibits poor electrical conductivity, ultimately resulting in arrhythmias and adverse cardiac remodeling. Due to their extracellular matrix-like structure and excellent biocompatibility, hydrogels are emerging as a focal point in cardiac tissue engineering. However, traditional hydrogels lack the necessary conductivity to restore electrical signal transmission in the infarcted regions. Imparting conductivity to hydrogels while also enhancing their adhesive properties enables them to adhere closely to myocardial tissue, establish stable electrical connections, and facilitate synchronized contraction and myocardial tissue repair within the infarcted area. This paper reviews the strategies for constructing conductive and adhesive hydrogels, focusing on their application in MI repair. Furthermore, the challenges and future directions in developing adhesive and conductive hydrogels for MI repair are discussed.
Collapse
Affiliation(s)
- Jialiang Zhao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Ying Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yuanyuan Qin
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yongqi Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
4
|
Shin W, Choi HJ, Kang B, Lee K, Choi YS, Choi JH, Kim HA, Choi MK, Chung K. Materials design and characterization of injectable and degradable oxidized alginate PANI:PSS hydrogels for photothermal therapy. Carbohydr Polym 2025; 347:122652. [PMID: 39486920 DOI: 10.1016/j.carbpol.2024.122652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 11/04/2024]
Abstract
Photothermal therapy has gained great attention as an alternative candidate for radiation therapy or chemotherapy for cancers. However, photothermal agents for photothermal therapy are generally in the form of nanoparticles that are too small to remain in the target tissue, and therefore, the agents are rather quickly removed from the targeted site. Furthermore, conventional photothermal agents are generally expensive or complicated to synthesize. As an approach to these issues, here we present new hydrogels with oxidized alginate ionically crosslinked with Ca2+, bearing polyaniline:poly(sodium 4-styrenesulfonate) (PANI:PSS) nanoparticles in the polymer network. The presented oxidized alginate PANI:PSS hydrogels exhibited excellent injectability as well as a gradual degradation rate from several days to several months depending on the oxidation degree of alginate chains. The presented oxidized alginate PANI:PSS hydrogels showed an excellent photothermal effect even under a neutral pH environment by showing temperature increased to 53 °C in 5 min upon NIR irradiation, which provide strong potential as a candidate for photothermal agent in photothermal therapy.
Collapse
Affiliation(s)
- Woohyeon Shin
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hui Ju Choi
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Bongkyun Kang
- KNU Institute of Basic Sciences and KNU G-LAMP Project Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyueui Lee
- KNU Institute of Basic Sciences and KNU G-LAMP Project Group, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea; Biomedical Research Institute, Kyungpook National University Hospital, Daegu 41940, Republic of Korea
| | - Yoon-Seong Choi
- Department of Carbon and Fiber Composite, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jin Hyun Choi
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Carbon and Fiber Composite, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyeon Ah Kim
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Carbon and Fiber Composite, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Moon Kee Choi
- Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Kyeongwoon Chung
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Carbon and Fiber Composite, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
5
|
Mierke CT. Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues. Cells 2024; 13:1638. [PMID: 39404401 PMCID: PMC11476109 DOI: 10.3390/cells13191638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The 3D bioprinting technique has made enormous progress in tissue engineering, regenerative medicine and research into diseases such as cancer. Apart from individual cells, a collection of cells, such as organoids, can be printed in combination with various hydrogels. It can be hypothesized that 3D bioprinting will even become a promising tool for mechanobiological analyses of cells, organoids and their matrix environments in highly defined and precisely structured 3D environments, in which the mechanical properties of the cell environment can be individually adjusted. Mechanical obstacles or bead markers can be integrated into bioprinted samples to analyze mechanical deformations and forces within these bioprinted constructs, such as 3D organoids, and to perform biophysical analysis in complex 3D systems, which are still not standard techniques. The review highlights the advances of 3D and 4D printing technologies in integrating mechanobiological cues so that the next step will be a detailed analysis of key future biophysical research directions in organoid generation for the development of disease model systems, tissue regeneration and drug testing from a biophysical perspective. Finally, the review highlights the combination of bioprinted hydrogels, such as pure natural or synthetic hydrogels and mixtures, with organoids, organoid-cell co-cultures, organ-on-a-chip systems and organoid-organ-on-a chip combinations and introduces the use of assembloids to determine the mutual interactions of different cell types and cell-matrix interferences in specific biological and mechanical environments.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
6
|
Wei X, Jiang X, Li H. Fundamental characteristics of ultrasonic green formulations using Avena sativa L. extract-mediated gold nanoparticles and electroconductive nanofibers for cardiovascular nursing care. Heliyon 2024; 10:e35018. [PMID: 39170527 PMCID: PMC11336310 DOI: 10.1016/j.heliyon.2024.e35018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
In the pursuit of novel approaches to address chronic heart failure and enhance cardiovascular nursing care, environmentally sustainable nanomaterials have taken center stage. Recent progress in regenerative medicine has opened doors for the use of biocompatible biomaterials that provide mechanical support to damaged heart tissue and facilitate electrical signaling. This study was dedicated to developing advanced electroconductive nanofibers by incorporating eco-friendly Avena sativa L. extract-mediated gold nanoparticles (AuNPs) into polyaniline to create an intricate cardiac patch. The AuNPs were synthesized through an environmentally friendly chemical process aided by ultrasonic conditions. Comprehensive physicochemical analyses, such as UV-Vis spectroscopy, SEM, TEM, DPPH assay, and XRD, were carried out to characterize the AuNPs. These AuNPs were then blended with a polycaprolactone/gelatin polymeric solution and electrospun to fabricate cardiac patches, which underwent thorough evaluation using various techniques. The resulting cardiac patch demonstrated excellent hemocompatibility, antioxidant properties, and cytocompatibility, offering a promising therapeutic approach for myocardial infarctions and the advancement of cardiovascular nursing care.
Collapse
Affiliation(s)
- Xinfang Wei
- Department of Cardiovascular Medicine CCU, Zhongshan People's Hospital, No. 2 Sunwendong Road, Zhongshan City, Guangdong, 528403, China
| | - Xiaoshan Jiang
- Department of Geriatrics, Qingdao Chengyang District People's Hospital, No. 600, Changcheng Road, Chengyang District, Qingdao, 266109, Shandong Province, China
| | - Hongzan Li
- School of Nursing, Guangdong Medical University, No. 1 Xincheng Road, Songshan Lake Science and Technology Park, Dongguan, Guangdong, 523808, China
| |
Collapse
|
7
|
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
|
8
|
Elkhoury K, Kodeih S, Enciso‐Martínez E, Maziz A, Bergaud C. Advancing Cardiomyocyte Maturation: Current Strategies and Promising Conductive Polymer-Based Approaches. Adv Healthc Mater 2024; 13:e2303288. [PMID: 38349615 PMCID: PMC11468390 DOI: 10.1002/adhm.202303288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/31/2024] [Indexed: 02/21/2024]
Abstract
Cardiovascular diseases are a leading cause of mortality and pose a significant burden on healthcare systems worldwide. Despite remarkable progress in medical research, the development of effective cardiovascular drugs has been hindered by high failure rates and escalating costs. One contributing factor is the limited availability of mature cardiomyocytes (CMs) for accurate disease modeling and drug screening. Human induced pluripotent stem cell-derived CMs offer a promising source of CMs; however, their immature phenotype presents challenges in translational applications. This review focuses on the road to achieving mature CMs by summarizing the major differences between immature and mature CMs, discussing the importance of adult-like CMs for drug discovery, highlighting the limitations of current strategies, and exploring potential solutions using electro-mechano active polymer-based scaffolds based on conductive polymers. However, critical considerations such as the trade-off between 3D systems and nutrient exchange, biocompatibility, degradation, cell adhesion, longevity, and integration into wider systems must be carefully evaluated. Continued advancements in these areas will contribute to a better understanding of cardiac diseases, improved drug discovery, and the development of personalized treatment strategies for patients with cardiovascular disorders.
Collapse
Affiliation(s)
- Kamil Elkhoury
- LAAS‐CNRS, Université de Toulouse, CNRSToulouseF‐31400France
| | - Sacha Kodeih
- Faculty of Medicine and Medical SciencesUniversity of BalamandTripoliP.O. Box 100Lebanon
| | | | - Ali Maziz
- LAAS‐CNRS, Université de Toulouse, CNRSToulouseF‐31400France
| | | |
Collapse
|
9
|
Yao ZF, Kuang Y, Wu HT, Lundqvist E, Fu X, Celt N, Pei J, Yee A, Ardoña HAM. Selective Induction of Molecular Assembly to Tissue-Level Anisotropy on Peptide-Based Optoelectronic Cardiac Biointerfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312231. [PMID: 38335948 PMCID: PMC11126358 DOI: 10.1002/adma.202312231] [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: 11/15/2023] [Revised: 01/19/2024] [Indexed: 02/12/2024]
Abstract
The conduction efficiency of ions in excitable tissues and of charged species in organic conjugated materials both benefit from having ordered domains and anisotropic pathways. In this study, a photocurrent-generating cardiac biointerface is presented, particularly for investigating the sensitivity of cardiomyocytes to geometrically comply to biomacromolecular cues differentially assembled on a conductive nanogrooved substrate. Through a polymeric surface-templated approach, photoconductive substrates with symmetric peptide-quaterthiophene (4T)-peptide units assembled as 1D nanostructures on nanoimprinted polyalkylthiophene (P3HT) surface are developed. The 4T-based peptides studied here can form 1D nanostructures on prepatterned polyalkylthiophene substrates, as directed by hydrogen bonding, aromatic interactions between 4T and P3HT, and physical confinement on the nanogrooves. It is observed that smaller 4T-peptide units that can achieve a higher degree of assembly order within the polymeric templates serve as a more efficient driver of cardiac cytoskeletal anisotropy than merely presenting aligned -RGD bioadhesive epitopes on a nanotopographic surface. These results unravel some insights on how cardiomyocytes perceive submicrometer dimensionality, local molecular order, and characteristics of surface cues in their immediate environment. Overall, the work offers a cardiac patterning platform that presents the possibility of a gene modification-free cardiac photostimulation approach while controlling the conduction directionality of the biotic and abiotic components.
Collapse
Affiliation(s)
- Ze-Fan Yao
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
| | - Yuyao Kuang
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Hao-Tian Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Emil Lundqvist
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Xin Fu
- Department of Materials Science and Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Natalie Celt
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Jian Pei
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Albert Yee
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Herdeline Ann M. Ardoña
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
10
|
Esmaeili H, Patino-Guerrero A, Nelson RA, Karamanova N, M Fisher T, Zhu W, Perreault F, Migrino RQ, Nikkhah M. Engineered Gold and Silica Nanoparticle-Incorporated Hydrogel Scaffolds for Human Stem Cell-Derived Cardiac Tissue Engineering. ACS Biomater Sci Eng 2024; 10:2351-2366. [PMID: 38323834 PMCID: PMC11075803 DOI: 10.1021/acsbiomaterials.3c01256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Electrically conductive biomaterials and nanomaterials have demonstrated great potential in the development of functional and mature cardiac tissues. In particular, gold nanomaterials have emerged as promising candidates due to their biocompatibility and ease of fabrication for cardiac tissue engineering utilizing rat- or stem cell-derived cardiomyocytes (CMs). However, despite significant advancements, it is still not clear whether the enhancement in cardiac tissue function is primarily due to the electroconductivity features of gold nanoparticles or the structural changes of the scaffold resulting from the addition of these nanoparticles. To address this question, we developed nanoengineered hydrogel scaffolds comprising gelatin methacrylate (GelMA) embedded with either electrically conductive gold nanorods (GNRs) or nonconductive silica nanoparticles (SNPs). This enabled us to simultaneously assess the roles of electrically conductive and nonconductive nanomaterials in the functionality and fate of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Our studies revealed that both GNR- and SNP-incorporated hydrogel scaffolds exhibited excellent biocompatibility and similar cardiac cell attachment. Although the expression of sarcomere alpha-actinin did not significantly differ among the conditions, a more organized sarcomere structure was observed within the GNR-embedded hydrogels compared to the nonconductive nanoengineered scaffolds. Furthermore, electrical coupling was notably improved in GNR-embedded scaffolds, as evidenced by the synchronous calcium flux and enhanced calcium transient intensity. While we did not observe a significant difference in the gene expression profile of human cardiac tissues formed on the conductive GNR- and nonconductive SNP-incorporated hydrogels, we noticed marginal improvements in the expression of some calcium and structural genes in the nanomaterial-embedded hydrogel groups as compared to the control condition. Given that the cardiac tissues formed atop the nonconductive SNP-based scaffolds (used as the control for conductivity) also displayed similar levels of gene expression as compared to the conductive hydrogels, it suggests that the electrical conductivity of nanomaterials (i.e., GNRs) may not be the sole factor influencing the function and fate of hiPSC-derived cardiac tissues when cells are cultured atop the scaffolds. Overall, our findings provide additional insights into the role of electrically conductive gold nanoparticles in regulating the functionalities of hiPSC-CMs.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - Ronald A Nelson
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Nina Karamanova
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
| | - Taylor M Fisher
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - François Perreault
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
- University of Arizona College of Medicine, Phoenix, Arizona 85004, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
11
|
Gu B, Han K, Cao H, Huang X, Li X, Mao M, Zhu H, Cai H, Li D, He J. Heart-on-a-chip systems with tissue-specific functionalities for physiological, pathological, and pharmacological studies. Mater Today Bio 2024; 24:100914. [PMID: 38179431 PMCID: PMC10765251 DOI: 10.1016/j.mtbio.2023.100914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in heart-on-a-chip systems hold great promise to facilitate cardiac physiological, pathological, and pharmacological studies. This review focuses on the development of heart-on-a-chip systems with tissue-specific functionalities. For one thing, the strategies for developing cardiac microtissues on heart-on-a-chip systems that closely mimic the structures and behaviors of the native heart are analyzed, including the imitation of cardiac structural and functional characteristics. For another, the development of techniques for real-time monitoring of biophysical and biochemical signals from cardiac microtissues on heart-on-a-chip systems is introduced, incorporating cardiac electrophysiological signals, contractile activity, and biomarkers. Furthermore, the applications of heart-on-a-chip systems in intelligent cardiac studies are discussed regarding physiological/pathological research and pharmacological assessment. Finally, the future development of heart-on-a-chip toward a higher level of systematization, integration, and maturation is proposed.
Collapse
Affiliation(s)
- Bingsong Gu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Kang Han
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hanbo Cao
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Xinxin Huang
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Xiao Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Mao Mao
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hui Zhu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hu Cai
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Jiankang He
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| |
Collapse
|
12
|
Wu Z, Li Q, Wang L, Zhang Y, Liu W, Zhao S, Geng X, Fan Y. A novel biomimetic nanofibrous cardiac tissue engineering scaffold with adjustable mechanical and electrical properties based on poly(glycerol sebacate) and polyaniline. Mater Today Bio 2023; 23:100798. [PMID: 37753375 PMCID: PMC10518490 DOI: 10.1016/j.mtbio.2023.100798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/02/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Biomaterial tissue engineering scaffolds play a critical role in providing mechanical support, promoting cells growth and proliferation. However, due to the insulation and inappropriate stiffness of most biomaterials, there is an unmet need to engineer a biomimetic nanofibrous cardiac tissue engineering scaffold with tailorable mechanical and electrical properties. Here, we demonstrate for the first time the feasibility to generate a novel type of biocompatible fibrous scaffolds by blending elastic poly(glycerol sebacate) (PGS) and conductive polyaniline (PANI) with the help of a nontoxic carrier polymer, poly (vinyl alcohol) (PVA). Aligned and random PGS/PANI scaffolds are successfully obtained after electrospinning, cross-linking, water and ethanol wash. Incorporating of different concentrations of PANI into PGS fibers, the fibrous sheets show enhanced conductivity and slower degradation rates while maintaining the favorable hemocompatibility. The elastic modulus of the PGS/PANI scaffolds is in the range of 0.65-2.18 MPa under wet conditions, which is similar to that of natural myocardium. All of these fibrous mats show good cell viability and were able to promote adhesion and proliferation of H9c2 cells. Furthermore, the in vivo host responses of both random and aligned scaffolds confirm their good biocompatibility. Therefore, these PGS/PANI scaffolds have great potential for cardiac tissue engineering.
Collapse
Affiliation(s)
- Zebin Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Qiao Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yang Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Wei Liu
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Shudong Zhao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xuezheng Geng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
13
|
Yi H, Patel R, Patel KD, Bouchard LS, Jha A, Perriman AW, Patel M. Conducting polymer-based scaffolds for neuronal tissue engineering. J Mater Chem B 2023; 11:11006-11023. [PMID: 37953707 DOI: 10.1039/d3tb01838e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Neuronal tissue engineering has immense potential for treating neurological disorders and facilitating nerve regeneration. Conducting polymers (CPs) have emerged as a promising class of materials owing to their unique electrical conductivity and biocompatibility. CPs, such as poly(3,4-ethylenedioxythiophene) (PEDOT), poly(3-hexylthiophene) (P3HT), polypyrrole (PPy), and polyaniline (PANi), have been extensively explored for their ability to provide electrical cues to neural cells. These polymers are widely used in various forms, including porous scaffolds, hydrogels, and nanofibers, and offer an ideal platform for promoting cell adhesion, differentiation, and axonal outgrowth. CP-based scaffolds can also serve as drug delivery systems, enabling localized and controlled release of neurotrophic factors and therapeutic agents to enhance neural regeneration and repair. CP-based scaffolds have demonstrated improved neural regeneration, both in vitro and in vivo, for treating spinal cord and peripheral nerve injuries. In this review, we discuss synthesis and scaffold processing methods for CPs and their applications in neuronal tissue regeneration. We focused on a detailed literature review of the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Hagje Yi
- Bio-Convergence (BC), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, Songdogwahak-ro, Yeonsu-gu, Incheon 21983, South Korea
| | - Rajkumar Patel
- Energy & Environmental Science and Engineering (EESE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdogwahak-ro, Yeonsugu, Incheon, 21938, South Korea
| | - Kapil D Patel
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Research School of Chemistry (RSC), Australian National University, Canberra, ACT 2601, Australia
- John Curtin School of Medical Research (JCSMR), Australian National University, Canberra, ACT 2601, Australia
| | | | - Amitabh Jha
- Department of Chemistry, Acadia University, Wolfville, NS, Canada
| | - Adam Willis Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Research School of Chemistry (RSC), Australian National University, Canberra, ACT 2601, Australia
- John Curtin School of Medical Research (JCSMR), Australian National University, Canberra, ACT 2601, Australia
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea.
| |
Collapse
|
14
|
Mehrotra S, Dey S, Sachdeva K, Mohanty S, Mandal BB. Recent advances in tailoring stimuli-responsive hybrid scaffolds for cardiac tissue engineering and allied applications. J Mater Chem B 2023; 11:10297-10331. [PMID: 37905467 DOI: 10.1039/d3tb00450c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
To recapitulate bio-physical properties and functional behaviour of native heart tissues, recent tissue engineering-based approaches are focused on developing smart/stimuli-responsive materials for interfacing cardiac cells. Overcoming the drawbacks of the traditionally used biomaterials, these smart materials portray outstanding mechanical and conductive properties while promoting cell-cell interaction and cell-matrix transduction cues in such excitable tissues. To date, a large number of stimuli-responsive materials have been employed for interfacing cardiac tissues alone or in combination with natural/synthetic materials for cardiac tissue engineering. However, their comprehensive classification and a comparative analysis of the role played by these materials in regulating cardiac cell behaviour and in vivo metabolism are much less discussed. In an attempt to cover the recent advances in fabricating stimuli-responsive biomaterials for engineering cardiac tissues, this review details the role of these materials in modulating cardiomyocyte behaviour, functionality and surrounding matrix properties. Furthermore, concerns and challenges regarding the clinical translation of these materials and the possibility of using such materials for the fabrication of bio-actuators and bioelectronic devices are discussed.
Collapse
Affiliation(s)
- Shreya Mehrotra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahti-781039, Assam, India. biman.mandal@iitg,ac.in
| | - Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahti-781039, Assam, India
| | - Kunj Sachdeva
- DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Sujata Mohanty
- DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahti-781039, Assam, India. biman.mandal@iitg,ac.in
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahti-781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| |
Collapse
|
15
|
Kyriakou S, Acosta S, El Maachi I, Rütten S, Jockenhoevel S. A Dexamethasone-Loaded Polymeric Electrospun Construct as a Tubular Cardiovascular Implant. Polymers (Basel) 2023; 15:4332. [PMID: 37960012 PMCID: PMC10649717 DOI: 10.3390/polym15214332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular tissue engineering is providing many solutions to cardiovascular diseases. The complex disease demands necessitating tissue-engineered constructs with enhanced functionality. In this study, we are presenting the production of a dexamethasone (DEX)-loaded electrospun tubular polymeric poly(l-lactide) (PLA) or poly(d,l-lactide-co-glycolide) (PLGA) construct which contains iPSC-CMs (induced pluripotent stem cell cardiomyocytes), HUVSMCs (human umbilical vein smooth muscle cells), and HUVECs (human umbilical vein endothelial cells) embedded in fibrin gel. The electrospun tube diameter was calculated, as well as the DEX release for 50 days for 2 different DEX concentrations. Furthermore, we investigated the influence of the polymer composition and concentration on the function of the fibrin gels by imaging and quantification of CD31, alpha-smooth muscle actin (αSMA), collagen I (col I), sarcomeric alpha actinin (SAA), and Connexin 43 (Cx43). We evaluated the cytotoxicity and cell proliferation of HUVECs and HUVSMCs cultivated in PLA and PLGA polymeric sheets. The immunohistochemistry results showed efficient iPSC-CM marker expression, while the HUVEC toxicity was higher than the respective HUVSMC value. In total, our study emphasizes the combination of fibrin gel and electrospinning in a functionalized construct, which includes three cell types and provides useful insights of the DEX release and cytotoxicity in a tissue engineering perspective.
Collapse
Affiliation(s)
- Stavroula Kyriakou
- Department of Biohybrid & Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (S.A.); (I.E.M.)
| | - Sergio Acosta
- Department of Biohybrid & Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (S.A.); (I.E.M.)
| | - Ikram El Maachi
- Department of Biohybrid & Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (S.A.); (I.E.M.)
| | - Stephan Rütten
- Electron Microscopy Facility, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (S.A.); (I.E.M.)
- AMIBM—Aachen-Maastricht-Institute for Biobased Materials, Maastricht University, 6167 RD Geleen, The Netherlands
| |
Collapse
|
16
|
Gonzalez G, Nelson AC, Holman AR, Whitehead AJ, LaMontagne E, Lian R, Vatsyayan R, Dayeh SA, Engler AJ. Conductive electrospun polymer improves stem cell-derived cardiomyocyte function and maturation. Biomaterials 2023; 302:122363. [PMID: 37898021 PMCID: PMC10841997 DOI: 10.1016/j.biomaterials.2023.122363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 10/30/2023]
Abstract
Despite numerous efforts to generate mature human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), cells often remain immature, electrically isolated, and may not reflect adult biology. Conductive polymers are attractive candidates to facilitate electrical communication between hPSC-CMs, especially at sub-confluent cell densities or diseased cells lacking cell-cell junctions. Here we electrospun conductive polymers to create a conductive fiber mesh and assess if electrical signal propagation is improved in hPSC-CMs seeded on the mesh network. Matrix characterization indicated fiber structure remained stable over weeks in buffer, scaffold stiffness remained near in vivo cardiac stiffness, and electrical conductivity scaled with conductive polymer concentration. Cells remained adherent and viable on the scaffolds for at least 5 days. Transcriptomic profiling of hPSC-CMs cultured on conductive substrates for 3 days showed upregulation of cardiac and muscle-related genes versus non-conductive fibers. Structural proteins were more organized and calcium handling was improved on conductive substrates, even at sub-confluent cell densities; prolonged culture on conductive scaffolds improved membrane depolarization compared to non-conductive substrates. Taken together, these data suggest that blended, conductive scaffolds are stable, supportive of electrical coupling in hPSC-CMs, and promote maturation, which may improve our ability to model cardiac diseases and develop targeted therapies.
Collapse
Affiliation(s)
- Gisselle Gonzalez
- Shu Chien-Gene Lay Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Aileena C Nelson
- Shu Chien-Gene Lay Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Alyssa R Holman
- Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | | | - Erin LaMontagne
- Shu Chien-Gene Lay Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Rachel Lian
- Shu Chien-Gene Lay Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Ritwik Vatsyayan
- Department of Electrical and Computer Engineering, University California San Diego, La Jolla, CA, 92093, USA
| | - Shadi A Dayeh
- Department of Electrical and Computer Engineering, University California San Diego, La Jolla, CA, 92093, USA
| | - Adam J Engler
- Shu Chien-Gene Lay Department of Bioengineering, La Jolla, CA, 92093, USA; Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
17
|
Shokrollahi P, Omidi Y, Cubeddu LX, Omidian H. Conductive polymers for cardiac tissue engineering and regeneration. J Biomed Mater Res B Appl Biomater 2023; 111:1979-1995. [PMID: 37306139 DOI: 10.1002/jbm.b.35293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/17/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023]
Abstract
Cardiovascular diseases, such as myocardial infarction, are considered a significant global burden and the leading cause of death. Given the inability of damaged cardiac tissue to self-repair, cell-based tissue engineering and regeneration may be the only viable option for restoring normal heart function. To maintain the normal excitation-contraction coupling function of cardiac tissue, uniform electronic and ionic conductance properties are required. To transport cells to damaged cardiac tissues, several techniques, including the incorporation of cells into conductive polymers (CPs) and biomaterials, have been utilized. Due to the complexity of cardiac tissues, the success of tissue engineering for the damaged heart is highly dependent on several variables, such as the cell source, growth factors, and scaffolds. In this review, we sought to provide a comprehensive overview of the electro CPs and biomaterials used in the engineering and regeneration of heart tissue.
Collapse
Affiliation(s)
- Parvin Shokrollahi
- Centre for Ocular Research & Education, School of Optometry & Vision Science, University of Waterloo, Waterloo, Canada
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Luigi X Cubeddu
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Hossein Omidian
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|
18
|
Benko A, Webster TJ. How to fix a broken heart-designing biofunctional cues for effective, environmentally-friendly cardiac tissue engineering. Front Chem 2023; 11:1267018. [PMID: 37901157 PMCID: PMC10602933 DOI: 10.3389/fchem.2023.1267018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023] Open
Abstract
Cardiovascular diseases bear strong socioeconomic and ecological impact on the worldwide healthcare system. A large consumption of goods, use of polymer-based cardiovascular biomaterials, and long hospitalization times add up to an extensive carbon footprint on the environment often turning out to be ineffective at healing such cardiovascular diseases. On the other hand, cardiac cell toxicity is among the most severe but common side effect of drugs used to treat numerous diseases from COVID-19 to diabetes, often resulting in the withdrawal of such pharmaceuticals from the market. Currently, most patients that have suffered from cardiovascular disease will never fully recover. All of these factors further contribute to the extensive negative toll pharmaceutical, biotechnological, and biomedical companies have on the environment. Hence, there is a dire need to develop new environmentally-friendly strategies that on the one hand would promise cardiac tissue regeneration after damage and on the other hand would offer solutions for the fast screening of drugs to ensure that they do not cause cardiovascular toxicity. Importantly, both require one thing-a mature, functioning cardiac tissue that can be fabricated in a fast, reliable, and repeatable manner from environmentally friendly biomaterials in the lab. This is not an easy task to complete as numerous approaches have been undertaken, separately and combined, to achieve it. This review gathers such strategies and provides insights into which succeed or fail and what is needed for the field of environmentally-friendly cardiac tissue engineering to prosper.
Collapse
Affiliation(s)
| | - Thomas J. Webster
- Department of Biomedical Engineering, Hebei University of Technology, Tianjin, China
- School of Engineering, Saveetha University, Chennai, India
- Program in Materials Science, UFPI, Teresina, Brazil
| |
Collapse
|
19
|
Kistamás K, Müller A, Muenthaisong S, Lamberto F, Zana M, Dulac M, Leal F, Maziz A, Costa P, Bernotiene E, Bergaud C, Dinnyés A. Multifactorial approaches to enhance maturation of human iPSC-derived cardiomyocytes. J Mol Liq 2023; 387:122668. [DOI: 10.1016/j.molliq.2023.122668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Montes A, Valor D, Penabad Y, Domínguez M, Pereyra C, de la Ossa EM. Formation of PLGA-PEDOT: PSS Conductive Scaffolds by Supercritical Foaming. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2441. [PMID: 36984321 PMCID: PMC10057315 DOI: 10.3390/ma16062441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
The usage of conjugated materials for the fabrication of foams intended to be used as therapeutic scaffolds is gaining relevance these days, as they hold certain properties that are not exhibited by other polymer types that have been regularly used until the present. Hence, this work aims to design a specific supercritical CO2 foaming process that would allow the production of porous polymeric devices with improved conductive properties, which would better simulate matrix extracellular conditions when used as therapeutic scaffolds (PLGA-PEDOT:PSS) systems. The effects of pressure, temperature, and contact time on the expansion factor, porosity, mechanical properties, and conductivity of the foam have been evaluated. The foams have been characterized by scanning electron and atomic force microscopies, liquid displacement, PBS degradation test, compression, and resistance to conductivity techniques. Values close to 40% porosity were obtained, with a uniform distribution of polymers on the surface and in the interior, expansion factors of up to 10 orders, and a wide range of conductivity values (2.2 × 10-7 to 1.0 × 10-5 S/cm) and mechanical properties (0.8 to 13.6 MPa Young's modulus in compression test). The conductive and porous scaffolds that have been produced by supercritical CO2 in this study show an interesting potential for tissue engineering and for neural or cardiac tissue regeneration purposes due to the fact that electrical conductivity is a crucial factor for proper cell function and tissue development.
Collapse
Affiliation(s)
- Antonio Montes
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Diego Valor
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Yaiza Penabad
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Manuel Domínguez
- Department Condensed Matter Physics and Institute of Electron Microscopy and Materials, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Clara Pereyra
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Enrique Martínez de la Ossa
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| |
Collapse
|
21
|
Serrano-Garcia W, Bonadies I, Thomas SW, Guarino V. New Insights to Design Electrospun Fibers with Tunable Electrical Conductive-Semiconductive Properties. SENSORS (BASEL, SWITZERLAND) 2023; 23:1606. [PMID: 36772646 PMCID: PMC9919353 DOI: 10.3390/s23031606] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 05/14/2023]
Abstract
Fiber electronics, such as those produced by the electrospinning technique, have an extensive range of applications including electrode surfaces for batteries and sensors, energy storage, electromagnetic interference shielding, antistatic coatings, catalysts, drug delivery, tissue engineering, and smart textiles. New composite materials and blends from conductive-semiconductive polymers (C-SPs) offer high surface area-to-volume ratios with electrical tunability, making them suitable for use in fields including electronics, biofiltration, tissue engineering, biosensors, and "green polymers". These materials and structures show great potential for embedded-electronics tissue engineering, active drug delivery, and smart biosensing due to their electronic transport behavior and mechanical flexibility with effective biocompatibility. Doping, processing methods, and morphologies can significantly impact the properties and performance of C-SPs and their composites. This review provides an overview of the current literature on the processing of C-SPs as nanomaterials and nanofibrous structures, mainly emphasizing the electroactive properties that make these structures suitable for various applications.
Collapse
Affiliation(s)
- William Serrano-Garcia
- Advanced Materials Bio & Integration Research (AMBIR) Laboratory, Department of Electrical Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Irene Bonadies
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Sylvia W Thomas
- Advanced Materials Bio & Integration Research (AMBIR) Laboratory, Department of Electrical Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Vincenzo Guarino
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Mostra d'Oltremare, Pad.20, 80125 Naples, Italy
| |
Collapse
|
22
|
Jalilinejad N, Rabiee M, Baheiraei N, Ghahremanzadeh R, Salarian R, Rabiee N, Akhavan O, Zarrintaj P, Hejna A, Saeb MR, Zarrabi A, Sharifi E, Yousefiasl S, Zare EN. Electrically conductive carbon-based (bio)-nanomaterials for cardiac tissue engineering. Bioeng Transl Med 2023; 8:e10347. [PMID: 36684103 PMCID: PMC9842069 DOI: 10.1002/btm2.10347] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
A proper self-regenerating capability is lacking in human cardiac tissue which along with the alarming rate of deaths associated with cardiovascular disorders makes tissue engineering critical. Novel approaches are now being investigated in order to speedily overcome the challenges in this path. Tissue engineering has been revolutionized by the advent of nanomaterials, and later by the application of carbon-based nanomaterials because of their exceptional variable functionality, conductivity, and mechanical properties. Electrically conductive biomaterials used as cell bearers provide the tissue with an appropriate microenvironment for the specific seeded cells as substrates for the sake of protecting cells in biological media against attacking mechanisms. Nevertheless, their advantages and shortcoming in view of cellular behavior, toxicity, and targeted delivery depend on the tissue in which they are implanted or being used as a scaffold. This review seeks to address, summarize, classify, conceptualize, and discuss the use of carbon-based nanoparticles in cardiac tissue engineering emphasizing their conductivity. We considered electrical conductivity as a key affecting the regeneration of cells. Correspondingly, we reviewed conductive polymers used in tissue engineering and specifically in cardiac repair as key biomaterials with high efficiency. We comprehensively classified and discussed the advantages of using conductive biomaterials in cardiac tissue engineering. An overall review of the open literature on electroactive substrates including carbon-based biomaterials over the last decade was provided, tabulated, and thoroughly discussed. The most commonly used conductive substrates comprising graphene, graphene oxide, carbon nanotubes, and carbon nanofibers in cardiac repair were studied.
Collapse
Affiliation(s)
- Negin Jalilinejad
- Biomaterial Group, Department of Biomedical EngineeringAmirkabir University of TechnologyTehranIran
| | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical EngineeringAmirkabir University of TechnologyTehranIran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | | | - Reza Salarian
- Biomedical Engineering DepartmentMaziar UniversityRoyanMazandaranIran
| | - Navid Rabiee
- Department of PhysicsSharif University of TechnologyTehranIran
- School of EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- Department of Materials Science and EngineeringPohang University of Science and Technology (POSTECH), 77 Cheongam‐ro, Nam‐guPohangGyeongbukSouth Korea
| | - Omid Akhavan
- Department of PhysicsSharif University of TechnologyTehranIran
| | - Payam Zarrintaj
- School of Chemical EngineeringOklahoma State UniversityStillwaterOklahomaUSA
| | - Aleksander Hejna
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural SciencesIstinye UniversityIstanbulTurkey
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and TechnologiesHamadan University of Medical SciencesHamadanIran
| | - Satar Yousefiasl
- School of DentistryHamadan University of Medical SciencesHamadanIran
| | | |
Collapse
|
23
|
Acosta M, Santiago MD, Irvin JA. Electrospun Conducting Polymers: Approaches and Applications. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15248820. [PMID: 36556626 PMCID: PMC9782039 DOI: 10.3390/ma15248820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 05/14/2023]
Abstract
Inherently conductive polymers (CPs) can generally be switched between two or more stable oxidation states, giving rise to changes in properties including conductivity, color, and volume. The ability to prepare CP nanofibers could lead to applications including water purification, sensors, separations, nerve regeneration, wound healing, wearable electronic devices, and flexible energy storage. Electrospinning is a relatively inexpensive, simple process that is used to produce polymer nanofibers from solution. The nanofibers have many desirable qualities including high surface area per unit mass, high porosity, and low weight. Unfortunately, the low molecular weight and rigid rod nature of most CPs cannot yield enough chain entanglement for electrospinning, instead yielding polymer nanoparticles via an electrospraying process. Common workarounds include co-extruding with an insulating carrier polymer, coaxial electrospinning, and coating insulating electrospun polymer nanofibers with CPs. This review explores the benefits and drawbacks of these methods, as well as the use of these materials in sensing, biomedical, electronic, separation, purification, and energy conversion and storage applications.
Collapse
Affiliation(s)
- Mariana Acosta
- Materials Science, Engineering and Commercialization Program, Texas State University, San Marcos, TX 78666, USA
| | - Marvin D. Santiago
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA
| | - Jennifer A. Irvin
- Materials Science, Engineering and Commercialization Program, Texas State University, San Marcos, TX 78666, USA
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA
- Correspondence:
| |
Collapse
|
24
|
Meng J, Xiao B, Wu F, Sun L, Li B, Guo W, Hu X, Xu X, Wen T, Liu J, Xu H. Co-axial fibrous scaffolds integrating with carbon fiber promote cardiac tissue regeneration post myocardial infarction. Mater Today Bio 2022; 16:100415. [PMID: 36105673 PMCID: PMC9465342 DOI: 10.1016/j.mtbio.2022.100415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Myocardium is an excitable tissue with electrical conductivity and mechanical strength. In this work, carbon fibers (CFs) and co-axial fibrous mesh were integrated which combined the high modulus and excellent electrical conductivity of CFs and the fibrous and porous structures of the electrospun fibers. The scaffold was fabricated by simply integrating coaxial electrospun fibers and carbon fibers through a freeze-drying procedure. It was shown that the integration of carbon fibers have the conductivity and Young's modulus of the fibrous mesh increased significantly, meanwhile, upregulated the expression of CX43, α-actinin, RhoA of the neonatal rat primary cardiomyocytes and primary human umbilical vein endothelial cells (HUVECs), and promoted the secretion of VEGF of HUVECs. Moreover, the cardiomyocytes grown on the scaffolds increased the ability of HUVECs migration. When implanted to the injury area post myocardial infraction, the scaffolds were able to effectively enhance the tissue regeneration and new vessel formation, which rescued the heart dysfunction induced by the myocardial infraction, evidenced by the results of echocardiography and histochemical analysis. In conclusion, the composite scaffolds could promote the myocardium regeneration and function's recovery by enhancing cardiomyocytes maturation and angiogenesis and establishing the crosstalk between the cardiomyocytes and the vascular endothelial cells.
Collapse
Affiliation(s)
- Jie Meng
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Bo Xiao
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Fengxin Wu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Lihong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Bo Li
- Peking Union Medical College, Beijing, 100730, China
| | - Wen Guo
- Peking Union Medical College, Beijing, 100730, China
| | - Xuechun Hu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Xuegai Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Tao Wen
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jian Liu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
25
|
Tambrchi P, DaliriJoupari M, Mahdavi AH, Soltani L. Adipose-Derived Mesenchymal Stem Cells Differentiation Toward Cardiomyocyte-Like Cells on the PCL/PANI Nanofibrous Scaffold: An Experimental Study. IRANIAN JOURNAL OF BIOTECHNOLOGY 2022; 20:e3205. [PMID: 38344322 PMCID: PMC10858366 DOI: 10.30498/ijb.2022.316370.3205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2024]
Abstract
BACKGROUND Owing to the fact that the heart tissue is not able to repair itself. Biomaterial-based scaffolds are important cues in tissue engineering (TE) applications. Recent advances in TE have led to the development of suitable scaffold architecture for various tissue defects. OBJECTIVE Given the importance of cellular therapy, it was the aim of the present study to differentiate cardio myocyte cells from human adipose-derived mesenchymal stem cells (Ad-MSCs) using suitable induction reagents (namely, 5-azacytidine and transforming growth factor beta (TGF-β)) on poly-caprolactone (PCL)/Poly aniline (PANI) Nano fibrous scaffolds prepared by electrospinning. MATERIALS AND METHODS For this purpose, the adipose-derived mesenchymal stem cells (Ad-MSCs) were initially isolated and characterized before cultivation on the PCL/PANI Nano fibrous scaffold to be treated for 21 days with 5-azacytidine either singly or in combination with TGF-β in medium. The scaffold's morphological and cell attachment properties were investigated using electron microscopy (SEM). Finally, the cardio myocyte differentiation of Ad-MSCs on the scaffold was studied using both quantitative Real-time PCR (qPCR) and flow-cytometry while the expression rates of the cardio myocytes' specific genes (Gata4, NKX2.5, MYH-7, and Troponin I) were also determined. RESULTS The results of Ad-MSCs culture, MTT assay, and SEM indicated that the cells had well proliferated on the PCL/PANI scaffolds, showing the biocompatibility of the nanofibers for cellular growth and adhesion. After 21 days of induced cardio myocyte differentiation by both agents, Real-time PCR revealed increases in the expressions of Gata4, Troponin I, MYH-7, and NKX2.5 genes in the cells cultured on the PCL/PANI scaffolds while the flow-cytometry test approved the expression of troponin I. CONCLUSION The data obtained showed that the PCL/PANI Nano fibrous scaffolds were able to promote and support mesenchymal stem cell transformation to cardio myocyte cells. Generally speaking, the results of the study might be exploited in future in vitro and in vivo experimental model studies of cardio myocyte differentiation using co-polymer scaffolds.
Collapse
Affiliation(s)
- Parastoo Tambrchi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Morteza DaliriJoupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Leila Soltani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Razi University, Kermanshah, Iran
| |
Collapse
|
26
|
Kerignard E, Bethry A, Falcoz C, Nottelet B, Pinese C. Design of Hybrid Polymer Nanofiber/Collagen Patches Releasing IGF and HGF to Promote Cardiac Regeneration. Pharmaceutics 2022; 14:1854. [PMID: 36145603 PMCID: PMC9502465 DOI: 10.3390/pharmaceutics14091854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Myocardial infarction in particular leads to a high rate of mortality, and in the case of survival, to a loss of myocardial functionality due to post-infarction necrosis. This functionality can be restored by cell therapy or biomaterial implantation, and the need for a rapid regeneration has led to the development of bioactive patches, in particular through the incorporation of growth factors (GF). In this work, we designed hybrid patches composed of polymer nanofibers loaded with HGF and IGF and associated with a collagen membrane. Among the different copolymers studied, the polymers and their porogens PLA-Pluronic-PLA + PEG and PCL + Pluronic were selected to encapsulate HGF and IGF. While 89 and 92% of IGF were released in 2 days, HGF was released up to 58% and 50% in 35 days from PLA-Pluronic-PLA + PEG and PCL + Pluronic nanofibers, respectively. We also compared two ways of association for the loaded nanofibers and the collagen membrane, namely a direct deposition of the nanofibers on a moisturized collagen membrane (wet association), or entrapment between collagen layers (sandwich association). The interfacial cohesion and the degradation properties of the patches were evaluated. We also show that the sandwich association decreases the burst release of HGF while increasing the release efficiency. Finally, we show that the patches are cytocompatible and that the presence of collagen and IGF promotes the proliferation of C2C12 myoblast cells for 11 days. Taken together, these results show that these hybrid patches are of interest for cardiac muscle regeneration.
Collapse
|
27
|
Scott L, Elídóttir K, Jeevaratnam K, Jurewicz I, Lewis R. Electrical stimulation through conductive scaffolds for cardiomyocyte tissue engineering: Systematic review and narrative synthesis. Ann N Y Acad Sci 2022; 1515:105-119. [PMID: 35676231 PMCID: PMC9796457 DOI: 10.1111/nyas.14812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Electrical conductivity is of great significance to cardiac tissue engineering and permits the use of electrical stimulation in mimicking cardiac pacing. The development of biomaterials for tissue engineering can incorporate physical properties that are uncommon to standard cell culture and can facilitate improved cardiomyocyte function. In this review, the PICOT question asks, "How has the application of external electrical stimulation in conductive scaffolds for tissue engineering affected cardiomyocyte behavior in in vitro cell culture?" The Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines, with predetermined inclusion and quality appraisal criteria, were used to assess publications from PubMed, Web of Science, and Scopus. Results revealed carbon nanotubes to be the most common conductive agent in biomaterials and rodent-sourced cell types as the most common cardiomyocytes used. To assess cardiomyocytes, immunofluorescence was used most often, utilizing proteins, such as connexin 43, cardiac α-actinin, and cardiac troponins. It was determined that the modal average stimulation protocol comprised 1-3 V square biphasic 50-ms pulses at 1 Hz, applied toward the end of cell culture. The addition of electrical stimulation to in vitro culture has exemplified it as a powerful tool for cardiac tissue engineering and brings researchers closer to creating optimal artificial cardiac tissue constructs.
Collapse
Affiliation(s)
- Louie Scott
- School of Veterinary MedicineUniversity of SurreyGuildfordUK
| | | | | | | | - Rebecca Lewis
- School of Veterinary MedicineUniversity of SurreyGuildfordUK
| |
Collapse
|
28
|
Munawar MA, Schubert DW. Thermal-Induced Percolation Phenomena and Elasticity of Highly Oriented Electrospun Conductive Nanofibrous Biocomposites for Tissue Engineering. Int J Mol Sci 2022; 23:ijms23158451. [PMID: 35955588 PMCID: PMC9369359 DOI: 10.3390/ijms23158451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Highly oriented electrospun conductive nanofibrous biocomposites (CNBs) of polylactic acid (PLA) and polyaniline (PANi) are fabricated using electrospinning. At the percolation threshold (φc), the growth of continuous paths between PANi particles leads to a steep increase in the electrical conductivity of fibers, and the McLachlan equation is fitted to identify φc. Annealing generates additional conductive channels, which lead to higher conductivity for dynamic percolation. For the first time, dynamic percolation is investigated for revealing time-temperature superposition in oriented conductive nanofibrous biocomposites. The crystallinity (χc) displays a linear dependence on annealing temperature within the confined fiber of CNBs. The increase in crystallinity due to annealing also increases the Young’s modulus E of CNBs. The present study outlines a reliable approach to determining the conductivity and elasticity of nanofibers that are highly desirable for a wide range of biological tissue applications.
Collapse
Affiliation(s)
- Muhammad A. Munawar
- Institute of Polymer Materials, Department of Material Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
- KeyLab Advanced Fiber Technology, Bavarian Polymer Institute, Dr.-Mack-Strasse 77, 90762 Fürth, Germany
- Correspondence: (M.A.M.); (D.W.S.)
| | - Dirk W. Schubert
- Institute of Polymer Materials, Department of Material Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
- KeyLab Advanced Fiber Technology, Bavarian Polymer Institute, Dr.-Mack-Strasse 77, 90762 Fürth, Germany
- Correspondence: (M.A.M.); (D.W.S.)
| |
Collapse
|
29
|
Ghovvati M, Kharaziha M, Ardehali R, Annabi N. Recent Advances in Designing Electroconductive Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200055. [PMID: 35368150 PMCID: PMC9262872 DOI: 10.1002/adhm.202200055] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/12/2022] [Indexed: 12/19/2022]
Abstract
Implantable cardiac patches and injectable hydrogels are among the most promising therapies for cardiac tissue regeneration following myocardial infarction. Incorporating electrical conductivity into these patches and hydrogels is found to be an efficient method to improve cardiac tissue function. Conductive nanomaterials such as carbon nanotube, graphene oxide, gold nanorod, as well as conductive polymers such as polyaniline, polypyrrole, and poly(3,4-ethylenedioxythiophene):polystyrene sulfonate are appealing because they possess the electroconductive properties of semiconductors with ease of processing and have potential to restore electrical signaling propagation through the infarct area. Numerous studies have utilized these materials for regeneration of biological tissues that possess electrical activities, such as cardiac tissue. In this review, recent studies on the use of electroconductive materials for cardiac tissue engineering and their fabrication methods are summarized. Moreover, recent advances in developing electroconductive materials for delivering therapeutic agents as one of emerging approaches for treating heart diseases and regenerating damaged cardiac tissues are highlighted.
Collapse
Affiliation(s)
- Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahshid Kharaziha
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
30
|
Mousa HM, Ali MG, Rezk AI, Nasr EA, Hussein KH. Development of conductive polymeric nanofiber patches for cardiac tissue engineering application. J Appl Polym Sci 2022. [DOI: 10.1002/app.52757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Hamouda M. Mousa
- Department of Mechanical Engineering, Faculty of Engineering South Valley University Qena Egypt
| | - Mustafa Ghazali Ali
- Department of Mechanical Engineering, Faculty of Engineering South Valley University Qena Egypt
| | - Abdelrahman I. Rezk
- Department of Bionanosystem Engineering Jeonbuk National University Jeonju Jeonbuk Republic of Korea
- Department of Bionanotechnology and Bioconvergence Engineering, Graduate School Jeonbuk National University Jeonju Republic of Korea
| | - Emad Abouel Nasr
- Department of Industrial Engineering, College of Engineering King Saud University Riyadh Saudi Arabia
| | - Kamal Hany Hussein
- Center for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering Loughborough University Loughborough Leicestershire UK
| |
Collapse
|
31
|
Suh T, Twiddy J, Mahmood N, Ali KM, Lubna MM, Bradford PD, Daniele MA, Gluck JM. Electrospun Carbon Nanotube-Based Scaffolds Exhibit High Conductivity and Cytocompatibility for Tissue Engineering Applications. ACS OMEGA 2022; 7:20006-20019. [PMID: 35721944 PMCID: PMC9202252 DOI: 10.1021/acsomega.2c01807] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/17/2022] [Indexed: 06/01/2023]
Abstract
Carbon nanotubes (CNTs) are known for their excellent conductive properties. Here, we present two novel methods, "sandwich" (sCNT) and dual deposition (DD CNT), for incorporating CNTs into electrospun polycaprolactone (PCL) and gelatin scaffolds to increase their conductance. Based on CNT percentage, the DD CNT scaffolds contain significantly higher quantities of CNTs than the sCNT scaffolds. The inclusion of CNTs increased the electrical conductance of scaffolds from 0.0 ± 0.00 kS (non-CNT) to 0.54 ± 0.10 kS (sCNT) and 5.22 ± 0.49 kS (DD CNT) when measured parallel to CNT arrays and to 0.25 ± 0.003 kS (sCNT) and 2.85 ± 1.12 (DD CNT) when measured orthogonally to CNT arrays. The inclusion of CNTs increased fiber diameter and pore size, promoting cellular migration into the scaffolds. CNT inclusion also decreased the degradation rate and increased hydrophobicity of scaffolds. Additionally, CNT inclusion increased Young's modulus and failure load of scaffolds, increasing their mechanical robustness. Murine fibroblasts were maintained on the scaffolds for 30 days, demonstrating high cytocompatibility. The increased conductivity and high cytocompatibility of the CNT-incorporated scaffolds make them appropriate candidates for future use in cardiac and neural tissue engineering.
Collapse
Affiliation(s)
- Taylor
C. Suh
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Jack Twiddy
- Joint
Department of Biomedical Engineering, North
Carolina State University and The University of North Carolina at
Chapel Hill, Raleigh, North Carolina 27606, United States
| | - Nasif Mahmood
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Kiran M. Ali
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Mostakima M. Lubna
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Philip D. Bradford
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Michael A. Daniele
- Joint
Department of Biomedical Engineering, North
Carolina State University and The University of North Carolina at
Chapel Hill, Raleigh, North Carolina 27606, United States
- Department
of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Jessica M. Gluck
- Department
of Textile Engineering, Chemistry, and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| |
Collapse
|
32
|
Zhao G, Zhou H, Jin G, Jin B, Geng S, Luo Z, Ge Z, Xu F. Rational Design of Electrically Conductive Biomaterials toward Excitable Tissues Regeneration. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
33
|
Saghebasl S, Akbarzadeh A, Gorabi AM, Nikzamir N, SeyedSadjadi M, Mostafavi E. Biodegradable functional macromolecules as promising scaffolds for cardiac tissue engineering. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5669] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Solmaz Saghebasl
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Abolfazl Akbarzadeh
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Universal Scientific Education and Research Network (USERN) Tabriz Iran
| | - Armita Mahdavi Gorabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Nasrin Nikzamir
- Department of Chemistry, Science and Research Branch Islamic Azad University Tehran Iran
| | | | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford California USA
- Department of Medicine Stanford University School of Medicine Stanford California USA
| |
Collapse
|
34
|
Zhang Y, Wang W, Zhang F, Dai K, Li C, Fan Y, Chen G, Zheng Q. Soft Organic Thermoelectric Materials: Principles, Current State of the Art and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104922. [PMID: 34921579 DOI: 10.1002/smll.202104922] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/25/2021] [Indexed: 06/14/2023]
Abstract
The enormous demand for waste heat utilization and burgeoning eco-friendly wearable materials has triggered huge interest in the development of thermoelectric materials that can harvest low-cost energy resources by converting waste heat to electricity efficiently. In particular, due to their high flexibility, nontoxicity, cost-effectivity, and promising applicability in various fields, organic thermoelectric materials are drawing more attention compared with their toxic, expensive, heavy, and brittle inorganic counterparts. Organic thermoelectric materials are approaching the figure of merit of the inorganic ones via the construction and optimization of unique transport pathways and device geometries. This review presents the recent development of the interdependence and decoupling principles of the thermoelectric efficiency parameters as well as the new achievements of high performance organic thermoelectric materials. Moreover, this review also discusses the advances in the thermoelectric devices with emphasis on their energy-related applications. It is believed that organic thermoelectric materials are emerging as green energy alternatives rivaling their conventional inorganic counterparts in the efficient and pure electricity harvesting from waste heat and solar thermal energy.
Collapse
Affiliation(s)
- Yinhang Zhang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
- Department of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Wei Wang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
| | - Fei Zhang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
- Department of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Kun Dai
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
| | - Chuanbing Li
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
| | - Yuan Fan
- College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, P. R. China
| | - Guangming Chen
- College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, P. R. China
| | - Qingbin Zheng
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
| |
Collapse
|
35
|
Li Y, Wei L, Lan L, Gao Y, Zhang Q, Dawit H, Mao J, Guo L, Shen L, Wang L. Conductive biomaterials for cardiac repair: A review. Acta Biomater 2022; 139:157-178. [PMID: 33887448 DOI: 10.1016/j.actbio.2021.04.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) is one of the fatal diseases in humans. Its incidence is constantly increasing annually all over the world. The problem is accompanied by the limited regenerative capacity of cardiomyocytes, yielding fibrous scar tissue formation. The propagation of electrical impulses in such tissue is severely hampered, negatively influencing the normal heart pumping function. Thus, reconstruction of the internal cardiac electrical connection is currently a major concern of myocardial repair. Conductive biomaterials with or without cell loading were extensively investigated to address this problem. This article introduces a detailed overview of the recent progress in conductive biomaterials and fabrication methods of conductive scaffolds for cardiac repair. After that, the advances in myocardial tissue construction in vitro by the restoration of intercellular communication and simulation of the dynamic electrophysiological environment are systematically reviewed. Furthermore, the latest trend in the study of cardiac repair in vivo using various conductive patches is summarized. Finally, we discuss the achievements and shortcomings of the existing conductive biomaterials and the properties of an ideal conductive patch for myocardial repair. We hope this review will help readers understand the importance and usefulness of conductive biomaterials in cardiac repair and inspire researchers to design and develop new conductive patches to meet the clinical requirements. STATEMENT OF SIGNIFICANCE: After myocardial infarction, the infarcted myocardial area is gradually replaced by heterogeneous fibrous tissue with inferior conduction properties, resulting in arrhythmia and heart remodeling. Conductive biomaterials have been extensively adopted to solve the problem. Summarizing the relevant literature, this review presents an overview of the types and fabrication methods of conductive biomaterials, and focally discusses the recent advances in myocardial tissue construction in vitro and myocardial repair in vivo, which is rarely covered in previous reviews. As well, the deficiencies of the existing conductive patches and their construction strategies for myocardial repair are discussed as well as the improving directions. Confidently, the readers of this review would appreciate advantages and current limitations of conductive biomaterials/patches in cardiac repair.
Collapse
Affiliation(s)
- Yimeng Li
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Leqian Wei
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Lizhen Lan
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Yaya Gao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Qian Zhang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Hewan Dawit
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China.
| | - Lamei Guo
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China
| | - Li Shen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Lu Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
36
|
Esmaeili H, Patino-Guerrero A, Hasany M, Ansari MO, Memic A, Dolatshahi-Pirouz A, Nikkhah M. Electroconductive biomaterials for cardiac tissue engineering. Acta Biomater 2022; 139:118-140. [PMID: 34455109 PMCID: PMC8935982 DOI: 10.1016/j.actbio.2021.08.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022]
Abstract
Myocardial infarction (MI) is still the leading cause of mortality worldwide. The success of cell-based therapies and tissue engineering strategies for treatment of injured myocardium have been notably hindered due to the limitations associated with the selection of a proper cell source, lack of engraftment of engineered tissues and biomaterials with the host myocardium, limited vascularity, as well as immaturity of the injected cells. The first-generation approaches in cardiac tissue engineering (cTE) have mainly relied on the use of desired cells (e.g., stem cells) along with non-conductive natural or synthetic biomaterials for in vitro construction and maturation of functional cardiac tissues, followed by testing the efficacy of the engineered tissues in vivo. However, to better recapitulate the native characteristics and conductivity of the cardiac muscle, recent approaches have utilized electroconductive biomaterials or nanomaterial components within engineered cardiac tissues. This review article will cover the recent advancements in the use of electrically conductive biomaterials in cTE. The specific emphasis will be placed on the use of different types of nanomaterials such as gold nanoparticles (GNPs), silicon-derived nanomaterials, carbon-based nanomaterials (CBNs), as well as electroconductive polymers (ECPs) for engineering of functional and electrically conductive cardiac tissues. We will also cover the recent progress in the use of engineered electroconductive tissues for in vivo cardiac regeneration applications. We will discuss the opportunities and challenges of each approach and provide our perspectives on potential avenues for enhanced cTE. STATEMENT OF SIGNIFICANCE: Myocardial infarction (MI) is still the primary cause of death worldwide. Over the past decade, electroconductive biomaterials have increasingly been applied in the field of cardiac tissue engineering. This review article provides the readers with the leading advances in the in vitro applications of electroconductive biomaterials for cTE along with an in-depth discussion of injectable/transplantable electroconductive biomaterials and their delivery methods for in vivo MI treatment. The article also discusses the knowledge gaps in the field and offers possible novel avenues for improved cardiac tissue engineering.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | | | - Masoud Hasany
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | | | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Alireza Dolatshahi-Pirouz
- Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark; Department of Health Technology, Technical University of Denmark, Center for Intestinal Absorption and Transport of Biopharmaceuticals, 2800 Kgs, Lyngby, Denmark
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA; Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
37
|
Electro‐conductive nanofibrous structure based on
PGS
/
PCL
coated with
PPy
by in situ chemical polymerization applicable as cardiac patch: Fabrication and optimization. J Appl Polym Sci 2022. [DOI: 10.1002/app.52136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
Hull SM, Brunel LG, Heilshorn SC. 3D Bioprinting of Cell-Laden Hydrogels for Improved Biological Functionality. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2103691. [PMID: 34672027 PMCID: PMC8988886 DOI: 10.1002/adma.202103691] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/15/2021] [Indexed: 05/03/2023]
Abstract
The encapsulation of cells within gel-phase materials to form bioinks offers distinct advantages for next-generation 3D bioprinting. 3D bioprinting has emerged as a promising tool for patterning cells, but the technology remains limited in its ability to produce biofunctional, tissue-like constructs due to a dearth of materials suitable for bioinks. While early demonstrations commonly used viscous polymers optimized for printability, these materials often lacked cell compatibility and biological functionality. In response, advanced materials that exist in the gel phase during the entire printing process are being developed, since hydrogels are uniquely positioned to both protect cells during extrusion and provide biological signals to embedded cells as the construct matures during culture. Here, an overview of the design considerations for gel-phase materials as bioinks is presented, with a focus on their mechanical, biochemical, and dynamic gel properties. Current challenges and opportunities that arise due to the fact that bioprinted constructs are active, living hydrogels composed of both acellular and cellular components are also evaluated. Engineering hydrogels with consideration of cells as an intrinsic component of the printed bioink will enable control over the evolution of the living construct after printing to achieve greater biofunctionality.
Collapse
Affiliation(s)
- Sarah M Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
39
|
Pitsalidis C, Pappa AM, Boys AJ, Fu Y, Moysidou CM, van Niekerk D, Saez J, Savva A, Iandolo D, Owens RM. Organic Bioelectronics for In Vitro Systems. Chem Rev 2021; 122:4700-4790. [PMID: 34910876 DOI: 10.1021/acs.chemrev.1c00539] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bioelectronics have made strides in improving clinical diagnostics and precision medicine. The potential of bioelectronics for bidirectional interfacing with biology through continuous, label-free monitoring on one side and precise control of biological activity on the other has extended their application scope to in vitro systems. The advent of microfluidics and the considerable advances in reliability and complexity of in vitro models promise to eventually significantly reduce or replace animal studies, currently the gold standard in drug discovery and toxicology testing. Bioelectronics are anticipated to play a major role in this transition offering a much needed technology to push forward the drug discovery paradigm. Organic electronic materials, notably conjugated polymers, having demonstrated technological maturity in fields such as solar cells and light emitting diodes given their outstanding characteristics and versatility in processing, are the obvious route forward for bioelectronics due to their biomimetic nature, among other merits. This review highlights the advances in conjugated polymers for interfacing with biological tissue in vitro, aiming ultimately to develop next generation in vitro systems. We showcase in vitro interfacing across multiple length scales, involving biological models of varying complexity, from cell components to complex 3D cell cultures. The state of the art, the possibilities, and the challenges of conjugated polymers toward clinical translation of in vitro systems are also discussed throughout.
Collapse
Affiliation(s)
- Charalampos Pitsalidis
- Department of Physics, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE.,Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Anna-Maria Pappa
- Department of Biomedical Engineering, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE
| | - Alexander J Boys
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Ying Fu
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, U.K
| | - Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Douglas van Niekerk
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Janire Saez
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006 Vitoria-Gasteiz, Spain.,Ikerbasque, Basque Foundation for Science, E-48011 Bilbao, Spain
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Donata Iandolo
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, 42023 Saint-Étienne, France
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| |
Collapse
|
40
|
Farahani A, Zarei-Hanzaki A, Abedi HR, Tayebi L, Mostafavi E. Polylactic Acid Piezo-Biopolymers: Chemistry, Structural Evolution, Fabrication Methods, and Tissue Engineering Applications. J Funct Biomater 2021; 12:71. [PMID: 34940550 PMCID: PMC8704870 DOI: 10.3390/jfb12040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 01/11/2023] Open
Abstract
Polylactide acid (PLA), as an FDA-approved biomaterial, has been widely applied due to its unique merits, such as its biocompatibility, biodegradability, and piezoelectricity. Numerous utilizations, including sensors, actuators, and bio-application-its most exciting application to promote cell migration, differentiation, growth, and protein-surface interaction-originate from the piezoelectricity effect. Since PLA exhibits piezoelectricity in both crystalline structure and an amorphous state, it is crucial to study it closely to understand the source of such a phenomenon. In this respect, in the current study, we first reviewed the methods promoting piezoelectricity. The present work is a comprehensive review that was conducted to promote the low piezoelectric constant of PLA in numerous procedures. In this respect, its chemistry and structural origins have been explored in detail. Combining any other variables to induce a specific application or to improve any PLA barriers, namely, its hydrophobicity, poor electrical conductivity, or the tuning of its mechanical properties, especially in the application of cardiovascular tissue engineering, is also discussed wherever relevant.
Collapse
Affiliation(s)
- Amirhossein Farahani
- Hot Deformation & Thermomechanical Processing Laboratory of High Performance Engineering Materials, School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | - Abbas Zarei-Hanzaki
- Hot Deformation & Thermomechanical Processing Laboratory of High Performance Engineering Materials, School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | - Hamid Reza Abedi
- School of Metallurgy & Materials Engineering, Iran University of Science and Technology (IUST), Tehran 16846-13114, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI 53233, USA;
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
41
|
Munawar MA, Schubert DW. Revealing Electrical and Mechanical Performances of Highly Oriented Electrospun Conductive Nanofibers of Biopolymers with Tunable Diameter. Int J Mol Sci 2021; 22:ijms221910295. [PMID: 34638631 PMCID: PMC8509057 DOI: 10.3390/ijms221910295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 11/24/2022] Open
Abstract
The present study outlines a reliable approach to determining the electrical conductivity and elasticity of highly oriented electrospun conductive nanofibers of biopolymers. The highly oriented conductive fibers are fabricated by blending a high molar mass polyethylene oxide (PEO), polycaprolactone (PCL), and polylactic acid (PLA) with polyaniline (PANi) filler. The filler-matrix interaction and molar mass (M) of host polymer are among governing factors for variable fiber diameter. The conductivity as a function of filler fraction (φ) is shown and described using a McLachlan equation to reveal the electrical percolation thresholds (φc) of the nanofibers. The molar mass of biopolymer, storage time, and annealing temperature are significant factors for φc. The Young’s modulus (E) of conductive fibers is dependent on filler fraction, molar mass, and post-annealing process. The combination of high orientation, tunable diameter, tunable conductivity, tunable elasticity, and biodegradability makes the presented nanofibers superior to the fibers described in previous literature and highly desirable for various biomedical and technical applications.
Collapse
Affiliation(s)
- Muhammad A. Munawar
- Institute of Polymer Materials, Department of Material Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
- KeyLab Advanced Fiber Technology, Bavarian Polymer Institute, Dr.-Mack-Strasse 77, 90762 Fürth, Germany
- Correspondence: (M.A.M.); (D.W.S.)
| | - Dirk W. Schubert
- Institute of Polymer Materials, Department of Material Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
- KeyLab Advanced Fiber Technology, Bavarian Polymer Institute, Dr.-Mack-Strasse 77, 90762 Fürth, Germany
- Correspondence: (M.A.M.); (D.W.S.)
| |
Collapse
|
42
|
Ul Haq A, Carotenuto F, De Matteis F, Prosposito P, Francini R, Teodori L, Pasquo A, Di Nardo P. Intrinsically Conductive Polymers for Striated Cardiac Muscle Repair. Int J Mol Sci 2021; 22:8550. [PMID: 34445255 PMCID: PMC8395236 DOI: 10.3390/ijms22168550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
One of the most important features of striated cardiac muscle is the excitability that turns on the excitation-contraction coupling cycle, resulting in the heart blood pumping function. The function of the heart pump may be impaired by events such as myocardial infarction, the consequence of coronary artery thrombosis due to blood clots or plaques. This results in the death of billions of cardiomyocytes, the formation of scar tissue, and consequently impaired contractility. A whole heart transplant remains the gold standard so far and the current pharmacological approaches tend to stop further myocardium deterioration, but this is not a long-term solution. Electrically conductive, scaffold-based cardiac tissue engineering provides a promising solution to repair the injured myocardium. The non-conductive component of the scaffold provides a biocompatible microenvironment to the cultured cells while the conductive component improves intercellular coupling as well as electrical signal propagation through the scar tissue when implanted at the infarcted site. The in vivo electrical coupling of the cells leads to a better regeneration of the infarcted myocardium, reducing arrhythmias, QRS/QT intervals, and scar size and promoting cardiac cell maturation. This review presents the emerging applications of intrinsically conductive polymers in cardiac tissue engineering to repair post-ischemic myocardial insult.
Collapse
Affiliation(s)
- Arsalan Ul Haq
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
| | - Felicia Carotenuto
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Fabio De Matteis
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Paolo Prosposito
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Roberto Francini
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Laura Teodori
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Alessandra Pasquo
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Paolo Di Nardo
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- L.L. Levshin Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
43
|
Liu Z, Wan X, Wang ZL, Li L. Electroactive Biomaterials and Systems for Cell Fate Determination and Tissue Regeneration: Design and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007429. [PMID: 34117803 DOI: 10.1002/adma.202007429] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/19/2020] [Indexed: 06/12/2023]
Abstract
During natural tissue regeneration, tissue microenvironment and stem cell niche including cell-cell interaction, soluble factors, and extracellular matrix (ECM) provide a train of biochemical and biophysical cues for modulation of cell behaviors and tissue functions. Design of functional biomaterials to mimic the tissue/cell microenvironment have great potentials for tissue regeneration applications. Recently, electroactive biomaterials have drawn increasing attentions not only as scaffolds for cell adhesion and structural support, but also as modulators to regulate cell/tissue behaviors and function, especially for electrically excitable cells and tissues. More importantly, electrostimulation can further modulate a myriad of biological processes, from cell cycle, migration, proliferation and differentiation to neural conduction, muscle contraction, embryogenesis, and tissue regeneration. In this review, endogenous bioelectricity and piezoelectricity are introduced. Then, design rationale of electroactive biomaterials is discussed for imitating dynamic cell microenvironment, as well as their mediated electrostimulation and the applying pathways. Recent advances in electroactive biomaterials are systematically overviewed for modulation of stem cell fate and tissue regeneration, mainly including nerve regeneration, bone tissue engineering, and cardiac tissue engineering. Finally, the significance for simulating the native tissue microenvironment is emphasized and the open challenges and future perspectives of electroactive biomaterials are concluded.
Collapse
Affiliation(s)
- Zhirong Liu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0245, USA
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
44
|
Rocha I, Cerqueira G, Varella Penteado F, Córdoba de Torresi SI. Electrical Stimulation and Conductive Polymers as a Powerful Toolbox for Tailoring Cell Behaviour in vitro. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:670274. [PMID: 35047926 PMCID: PMC8757900 DOI: 10.3389/fmedt.2021.670274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/17/2021] [Indexed: 12/26/2022] Open
Abstract
Electrical stimulation (ES) is a well-known method for guiding the behaviour of nerve cells in in vitro systems based on the response of these cells to an electric field. From this perspective, understanding how the electrochemical stimulus can be tuned for the design of a desired cell response is of great importance. Most biomedical studies propose the application of an electrical potential to cell culture arrays while examining the cell response regarding viability, morphology, and gene expression. Conversely, various studies failed to evaluate how the fine physicochemical properties of the materials used for cell culture influence the observed behaviours. Among the various materials used for culturing cells under ES, conductive polymers (CPs) are widely used either in pristine form or in addition to other polymers. CPs themselves do not possess the optimal surface for cell compatibility because of their hydrophobic nature, which leads to poor protein adhesion and, hence, poor bioactivity. Therefore, understanding how to tailor the chemical properties on the material surface will determine the obtention of improved ES platforms. Moreover, the structure of the material, either in a thin film or in porous electrospun scaffolds, also affects the biochemical response and needs to be considered. In this review, we examine how materials based on CPs influence cell behaviour under ES, and we compile the various ES setups and physicochemical properties that affect cell behaviour. This review concerns the culture of various cell types, such as neurons, fibroblasts, osteoblasts, and Schwann cells, and it also covers studies on stem cells prone to ES. To understand the mechanistic behaviour of these devices, we also examine studies presenting a more detailed biomolecular level of interaction. This review aims to guide the design of future ES setups regarding the influence of material properties and electrochemical conditions on the behaviour of in vitro cell studies.
Collapse
Affiliation(s)
- Igor Rocha
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
45
|
Basurto IM, Mora MT, Gardner GM, Christ GJ, Caliari SR. Aligned and electrically conductive 3D collagen scaffolds for skeletal muscle tissue engineering. Biomater Sci 2021; 9:4040-4053. [PMID: 33899845 DOI: 10.1039/d1bm00147g] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal muscle is characterized by its three-dimensional (3D) anisotropic architecture composed of highly aligned and electrically-excitable muscle fibers that enable normal movement. Biomaterial-based tissue engineering approaches to repair skeletal muscle are limited due to difficulties combining 3D structural alignment (to guide cell/matrix organization) and electrical conductivity (to enable electrically-excitable myotube assembly and maturation). In this work we successfully produced aligned and electrically conductive 3D collagen scaffolds using a freeze-drying approach. Conductive polypyrrole (PPy) nanoparticles were synthesized and directly mixed into a suspension of type I collagen and chondroitin sulfate followed by directional lyophilization. Scanning electron microscopy (SEM), energy-dispersive spectroscopy (EDS), and confocal microscopy showed that directional solidification resulted in scaffolds with longitudinally aligned pores with homogeneously-distributed PPy content. Chronopotentiometry verified that PPy incorporation resulted in a five-fold increase in conductivity compared to non-PPy-containing collagen scaffolds without detrimentally affecting myoblast metabolic activity. Furthermore, the aligned scaffold microstructure provided contact guidance cues that directed myoblast growth and organization. Incorporation of PPy also promoted enhanced myotube formation and maturation as measured by myosin heavy chain (MHC) expression and number of nuclei per myotube. Together these data suggest that aligned and electrically conductive 3D collagen scaffolds could be useful for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | | | | | - George J Christ
- Department of Biomedical Engineering, USA. and Department of Orthopedic Surgery, University of Virginia, USA
| | - Steven R Caliari
- Department of Biomedical Engineering, USA. and Department of Chemical Engineering, USA
| |
Collapse
|
46
|
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: 14] [Impact Index Per Article: 3.5] [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
|
47
|
Pushp P, Nogueira DES, Rodrigues CAV, Ferreira FC, Cabral JMS, Gupta MK. A Concise Review on Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Personalized Regenerative Medicine. Stem Cell Rev Rep 2021; 17:748-776. [PMID: 33098306 DOI: 10.1007/s12015-020-10061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
The induced pluripotent stem cells (iPSCs) are derived from somatic cells by using reprogramming factors such as Oct4, Sox2, Klf4, and c-Myc (OSKM) or Oct4, Sox2, Nanog and Lin28 (OSNL). They resemble embryonic stem cells (ESCs) and have the ability to differentiate into cell lineage of all three germ-layer, including cardiomyocytes (CMs). The CMs can be generated from iPSCs by inducing embryoid bodies (EBs) formation and treatment with activin A, bone morphogenic protein 4 (BMP4), and inhibitors of Wnt signaling. However, these iPSC-derived CMs are a heterogeneous population of cells and require purification and maturation to mimic the in vivo CMs. The matured CMs can be used for various therapeutic purposes in regenerative medicine by cardiomyoplasty or through the development of tissue-engineered cardiac patches. In recent years, significant advancements have been made in the isolation of iPSC and their differentiation, purification, and maturation into clinically usable CMs. Newer small molecules have also been identified to substitute the reprogramming factors for iPSC generation as well as for direct differentiation of somatic cells into CMs without an intermediary pluripotent state. This review provides a concise update on the generation of iPSC-derived CMs and their application in personalized cardiac regenerative medicine. It also discusses the current limitations and challenges in the application of iPSC-derived CMs. Graphical abstract.
Collapse
Affiliation(s)
- Pallavi Pushp
- Department of Biotechnology, Institute of Engineering and Technology (IET), Bundelkhand University, Jhansi, Uttar Pradesh, 284128, India
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India
| | - Diogo E S Nogueira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frederico C Ferreira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Mukesh Kumar Gupta
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India.
| |
Collapse
|
48
|
Molino BZ, Fukuda J, Molino PJ, Wallace GG. Redox Polymers for Tissue Engineering. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:669763. [PMID: 35047925 PMCID: PMC8757887 DOI: 10.3389/fmedt.2021.669763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/22/2021] [Indexed: 01/23/2023] Open
Abstract
This review will focus on the targeted design, synthesis and application of redox polymers for use in regenerative medicine and tissue engineering. We define redox polymers to encompass a variety of polymeric materials, from the multifunctional conjugated conducting polymers to graphene and its derivatives, and have been adopted for use in the engineering of several types of stimulus responsive tissues. We will review the fundamental properties of organic conducting polymers (OCPs) and graphene, and how their properties are being tailored to enhance material - biological interfacing. We will highlight the recent development of high-resolution 3D fabrication processes suitable for biomaterials, and how the fabrication of intricate scaffolds at biologically relevant scales is providing exciting opportunities for the application of redox polymers for both in-vitro and in-vivo tissue engineering. We will discuss the application of OCPs in the controlled delivery of bioactive compounds, and the electrical and mechanical stimulation of cells to drive behaviour and processes towards the generation of specific functional tissue. We will highlight the relatively recent advances in the use of graphene and the exploitation of its physicochemical and electrical properties in tissue engineering. Finally, we will look forward at the future of organic conductors in tissue engineering applications, and where the combination of materials development and fabrication processes will next unite to provide future breakthroughs.
Collapse
Affiliation(s)
- Binbin Z. Molino
- Faculty of Engineering, Yokohama National University, Yokohama, Japan
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, Yokohama, Japan
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Paul J. Molino
- Australian Research Council (ARC) Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Gordon G. Wallace
- Australian Research Council (ARC) Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
49
|
Fang W, Sun F, Tang J, Zhao Q, Chen J, Lei X, Zhang J, Zhang Y, Zuo Y, Li J, Li Y. Porous Electroactive and Biodegradable Polyurethane Membrane through Self-Doping Organogel. Macromol Rapid Commun 2021; 42:e2100125. [PMID: 33904219 DOI: 10.1002/marc.202100125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/02/2021] [Indexed: 12/15/2022]
Abstract
In order to improve the processability of conductive polyurethane (CPU) containing aniline oligomers, a new CPU containing aniline trimer (AT) and l-lysine (PUAT) are designed and synthesized. Further, the 3D porous PUAT membranes have been prepared by a simple gel cooperated with freeze-drying method. Chemical testings and conductive properties testify a self- doping model of PUAT based on the rich electronic l-lysine and electroaffinity AT moities. The self-doping behavior further endows the PUAT copolymers specific characteristics such as high electrical conductivity and the formation of the polaron lattice like-structure in good solvent dimethyl sulfoxide. The combination of organogel and freeze-drying could prevent the collapse of pore structure when the copolymers are molded as membranes. The synergistic effect of l-lysine and AT components has a strong influence on the dissolution, degradation, thermal stability, and mechanical properties of PUAT. The excellent properties of PUAT would broad the application of conductive polymers in biomedicine field.
Collapse
Affiliation(s)
- Wei Fang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Fuhua Sun
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, P. R. China
| | - Jiajing Tang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Qing Zhao
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Jie Chen
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiaoyu Lei
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Jinzheng Zhang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Yinglong Zhang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Yi Zuo
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Jidong Li
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Yubao Li
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
50
|
Chandika P, Heo SY, Kim TH, Oh GW, Kim GH, Kim MS, Jung WK. Recent advances in biological macromolecule based tissue-engineered composite scaffolds for cardiac tissue regeneration applications. Int J Biol Macromol 2020; 164:2329-2357. [DOI: 10.1016/j.ijbiomac.2020.08.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
|