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
|
Lv Q, Zhou D, He Y, Xu T, Qiu X, Zeng J. Engineering functional electroconductive hydrogels for targeted therapy in myocardial infarction repair. Bioact Mater 2025; 49:172-192. [PMID: 40124599 PMCID: PMC11929901 DOI: 10.1016/j.bioactmat.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 03/25/2025] Open
Abstract
Myocardial infarction (MI) is characterized by a paucity of cardiomyocyte regeneration, leading to significant morbidity and mortality. Contemporary therapeutic modalities, while mitigating ischemic effects, fail to reconstitute the impaired electromechanical coupling within the infracted myocardium. Emerging evidence supports the utility of electroconductive hydrogels (ECHs) in facilitating post-MI cardiac function recovery by restoring the conductive microenvironment of the infarcted tissue. This comprehensive review delineates the taxonomy of ECHs predicated on their constituent conductive materials. It also encapsulates prevailing research trends in ECH-mediated MI repair, encompassing innovative design paradigms and microenvironment-sensitive strategies. The review also provides a critical appraisal of various implantation techniques, underscored by a thorough examination of the attendant considerations. It elucidates the mechanistic underpinnings by which hydrogels exert salutary effects on myocardial repair, namely by augmenting mechanical and electrical integrity, exerting anti-inflammatory actions, fostering angiogenesis, and curtailing adverse remodeling processes. Furthermore, the review engages with the pressing challenge of optimizing ECH functionality to achieve superior reparative outcomes post-MI. The discourse concludes with an anticipatory perspective on the evolution of ECH scaffolds, advocating for a tailored approach that integrates multifaceted physicochemical properties to cater to the nuances of personalized medicine.
Collapse
Affiliation(s)
- Qianqian Lv
- Department of Physiology, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
- International Center for Translational Medicine, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528300, PR China
| | - Dandan Zhou
- Department of Physiology, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
- International Center for Translational Medicine, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528300, PR China
| | - Yutong He
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, PR China
| | - Tao Xu
- Department of Physiology, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Xiaozhong Qiu
- International Center for Translational Medicine, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528300, PR China
| | - Junwei Zeng
- Department of Physiology, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| |
Collapse
|
3
|
Park J, Kim J, Choe G, Jung Y, Lee JY. Conductive hydrogel luminal filler for peripheral nerve regeneration. Biomaterials 2025; 317:123103. [PMID: 39827510 DOI: 10.1016/j.biomaterials.2025.123103] [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: 04/08/2024] [Revised: 12/06/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Peripheral nerve injuries impair quality of life due to pain and loss of sensory and motor functions. Current treatments like autografts and nerve guidance conduits (NGCs) have limitations in functional restoration. Luminal fillers can enhance the effectiveness of NGCs by providing beneficial intraneural environments. In this study, we devised a novel injectable conductive luminal filler that allows for electrically active environments and efficient electrical stimulation of nerves. We developed injectable conductive hydrogel as a luminal filler for NGCs, composed of pluronic-coated reduced graphene oxide (rGO) and gelatin-based polymers, that gels spontaneously under physiological conditions. This filler combines nerve-like softness (0.31 ± 0.02 kPa), appropriate conductivity (2.7 ± 0.3 mS/cm), quick gelation (<5 min), and in vivo degradability. In a rat peripheral nerve defect model, the conductive hydrogel filler with electrical stimulation showed promising results in nerve regrowth, myelination, and functional recovery, performing comparably to autografts. This study underscores the potential of conductive fillers in enhancing nerve regeneration therapies.
Collapse
Affiliation(s)
- Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Junghyun Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Goeun Choe
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
4
|
Han Q, Yu J, Gao Z, Li S, Wang Z, Wu J, Huang F. Polypyrrole-modified gelatin-based hydrogel: A dressing for intestinal perforation treatment with enhanced wound healing and anti-adhesion properties. Int J Biol Macromol 2025; 309:142738. [PMID: 40180106 DOI: 10.1016/j.ijbiomac.2025.142738] [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: 05/18/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Intestinal perforation is a serious medical emergency, and traditional surgery often causes adhesion and other complications. Innovative hydrogels improve postoperative care and rehabilitation with their anti-adhesion, antibacterial, and hemostatic properties. We have developed an advanced anti-adhesion hydrogel, AA-A30, composed of polypyrrole-modified gelatin (PPy-GelMA), carboxymethyl chitosan (CMCS), and NHS-functionalized polyethylene glycol (PEG-NHS). This hydrogel is specifically tailored for intestinal perforations. Upon hydrolysis, PEG-NHS forms a protective barrier that effectively prevents adhesion to surrounding normal tissues. Furthermore, the integration of PPy-GelMA significantly extends the degradation duration of the hydrogel, from 24 to 48 h. In a mouse model of intestinal perforation, the AA-A30 hydrogel demonstrated remarkable efficacy in inhibiting inflammation and preventing tissue adhesion by modulating the expression of both inflammatory and tissue adhesion-related factors, such as IL-1β, TNF-α, and the ratio of tPA to PAI-1. These findings underscore the considerable potential of AA-A30 for the therapeutic management of intestinal perforations.
Collapse
Affiliation(s)
- Qingyue Han
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Jingrong Yu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Zhengkun Gao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Sitong Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Zi Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Jie Wu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Fengjie Huang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China.
| |
Collapse
|
5
|
Lyu X, Liang T, Zheng J, He C, Xu D, Han H, Zou L, Fang J, Hu N. High-Efficiency ICG Molecular Vibration Therapy for Bradyarrhythmia Using Cardiomyocyte-Based Biosensing. ACS Sens 2025; 10:3061-3071. [PMID: 40175296 DOI: 10.1021/acssensors.5c00196] [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] [Indexed: 04/04/2025]
Abstract
Bradyarrhythmia is a major cause of cardiovascular disease morbidity and mortality. Currently, medication and/or surgery are the conventional clinical therapeutic strategies for bradyarrhythmia, whereas drug side effects, invasive surgery, or potential complications limit their extensive application. Therefore, the development of alternative therapies for bradyarrhythmia is urgently needed. Herein, we propose a universal and efficient drug-mimicking strategy to treat bradyarrhythmia, which relies on the photothermal properties of near-infrared-triggered indocyanine green (ICG). An in situ integrated cell-based biosensing-regulating platform was developed to assess treatment efficacy by dynamically analyzing the cardiomyocyte electrophysiology activities. These findings indicate that the thermal vibration of ICG can efficiently enhance the electrophysiology of cardiomyocytes with bradyarrhythmia and maintain a rhythmic state for a long time, which is superior to that of Au nanorod plasmonic localized heating. Moreover, qualitative investigations confirmed that thermal stimulation is a pivotal factor in enhancing cardiomyocyte electrophysiological activity during photothermal treatment. This study provides a noninvasive drug-mimicking treatment strategy for bradyarrhythmia and establishes a reliable cell-based biosensing-regulating platform for electrophysiological assessment and drug screening, contributing to the further development of bradyarrhythmia therapies.
Collapse
Affiliation(s)
- Xuelian Lyu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tao Liang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Jilin Zheng
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chengwen He
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Dongxin Xu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Haote Han
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Ling Zou
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiaru Fang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Neurology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| |
Collapse
|
6
|
Fontana-Escartín A, Rosa E, Diaferia C, Lanzalaco S, Accardo A, Alemán C. Evaluation of the electrochemical response of aromatic peptides for biodetection of dopamine. J Colloid Interface Sci 2025; 679:441-454. [PMID: 39368163 DOI: 10.1016/j.jcis.2024.09.234] [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: 07/27/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
Biologically inspired aromatic peptide-based materials are gaining increasing interest as novel charge transport materials for bioelectronics due to their remarkable electrical response and inherent biocompatibility. In this work, the electrochemical response of ten aromatic amino acids and eleven aromatic peptides has been evaluated to assess the potential of incorporating peptides into electrochemical sensors not as biorecognition elements but as biocompatible electronic materials. While the electrochemical response of amino acids is null in all cases, the hexapeptide of phenylalanine (Phe) capped with eight polyethylene glycol units at the N-terminus and, especially, the cyclic dipeptide formed by two dehydro-phenylalanine residues (cyclo(ΔPhe2)), which organize in fibrillary self-assembled structures of nano- and submicrometric size, respectively, are the most electroactive peptides. Electrodes to electrochemically detect the oxidation of dopamine have been prepared using a plasma-activated polyethylene terephthalate glycol substrate covered with a poly(3,4-ethylenedioxythiophene) layer and a peptide coating deposited at the surface. The highest analytical sensitivity and the lowest limits of detection and quantifications have been obtained for the electrode coated with cyclo(ΔPhe2), which shows much better results than that without peptide. These results, on the one hand, confirm the significant role of electron transport through π-stacking interactions in the electrochemical response of peptides and, on the other hand, demonstrate that peptides can be directly used as electronic materials rather than as simple recognition elements in electrochemical biosensors.
Collapse
Affiliation(s)
- Adrián Fontana-Escartín
- Departament d'Enginyeria Química and Barcelona Research Center in Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain; Departament de Física EETAC, Universitat Politècnica de Catalunya, c/ Esteve Terrades, 7, 08860 Castelldefels, Spain
| | - Elisabetta Rosa
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Via T. De Amicis 95, Naples 80145, Italy
| | - Carlo Diaferia
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Via T. De Amicis 95, Naples 80145, Italy
| | - Sonia Lanzalaco
- Departament d'Enginyeria Química and Barcelona Research Center in Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain
| | - Antonella Accardo
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Via T. De Amicis 95, Naples 80145, Italy.
| | - Carlos Alemán
- Departament d'Enginyeria Química and Barcelona Research Center in Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona Spain.
| |
Collapse
|
7
|
Wang X, Tian H, Pan W, Du B, Chen Z, Zhang R, Zhou P. Applications of carbon dot-mediated cardiomyocyte maturation in regenerative medicine: a review. Nanomedicine (Lond) 2025; 20:319-328. [PMID: 39719674 PMCID: PMC11792849 DOI: 10.1080/17435889.2024.2443378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
The maturation of cardiomyocytes (CMs) plays key roles in regenerative medicine and the treatment of cardiovascular diseases via stem cell-derived CMs. Carbon dots (CDs) have good biocompatibility, optical properties, and electrophysical properties and have been widely applied in bioimaging, biosensors, and biotherapy. In this review, we comprehensively summarize recent advances in promoting the maturation of CMs, mainly human pluripotent stem cell-derived CMs, and related regenerative medicine. Moreover, we explore the innovative application of CDs to enhance the maturation of these CMs. Finally, we look forward to the future design and application of CDs in the maturation of CMs in terms of cell therapies.
Collapse
Affiliation(s)
- Xinyuan Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Hao Tian
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Wen Pan
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Binhong Du
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhen Chen
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Rongzhi Zhang
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu ProvinceChina
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
8
|
Li S, Yin W, Liu Y, Yang C, Zhai Z, Xie M, Ye Z, Song X. Anisotropic conductive scaffolds for post-infarction cardiac repair. Biomater Sci 2025; 13:542-567. [PMID: 39688676 DOI: 10.1039/d4bm01109k] [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: 12/18/2024]
Abstract
Myocardial infarction (MI) remains one of the most common and lethal cardiovascular diseases (CVDs), leading to the deterioration of cardiac function due to myocardial cell necrosis and fibrous scar tissue formation. Myocardial infarction (MI) remains one of the most common and lethal cardiovascular diseases (CVDs), leading to the deterioration of cardiac function due to myocardial cell necrosis and fibrous scar tissue formation. After MI, the anisotropic structural properties of myocardial tissue are destroyed, and its mechanical and electrical microenvironment also undergoes a series of pathological changes, such as ventricular wall stiffness, abnormal contraction, conduction network disruption, and irregular electrical signal propagation, which may further induce myocardial remodeling and even lead to heart failure. Therefore, bionic reconstruction of the anisotropic structural-mechanical-electrical microenvironment of the infarct area is key to repairing damaged myocardium. This article first summarizes the pathological changes in muscle fibre structure and conductive microenvironment after cardiac injury, and focuses on the classification and preparation methods of anisotropic conductive materials. In addition, the effects of these anisotropic conductive materials on the behavior of cardiac resident cells after myocardial infarction, such as directional growth, maturation, proliferation and migration, and the differentiation fate of stem cells and the possible molecular mechanisms involved are summarized. The design strategies for anisotropic conductive scaffolds for myocardial repair in future clinical research are also discussed, with the aim of providing new insights for researchers in related fields.
Collapse
Affiliation(s)
- Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
9
|
Li L, Shanmugasundaram A, Kim J, Oyunbaatar NE, Kanade PP, Cha SE, Lim D, Lee CH, Kim YB, Lee BK, Kim ES, Lee DW. Graphene SU-8 Platform for Enhanced Cardiomyocyte Maturation and Intercellular Communication in Cardiac Drug Screening. ACS NANO 2024; 18:33293-33309. [PMID: 39591586 DOI: 10.1021/acsnano.4c05365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Cell culture substrates designed for myocardial applications are pivotal in promoting the maturation and functional integration of cardiomyocytes. However, traditional in vitro models often inadequately mimic the diverse biochemical signals and electrophysiological properties of mature cardiomyocytes. Herein, we propose the application of monolayer graphene, transferred onto SU-8 cantilevers integrated with a microelectrode array, to evaluate its influence on the structural, functional, and electro-mechano-physiological properties of cardiomyocytes. The monolayer graphene, prepared using chemical vapor deposition, is adeptly transferred to the target substrates via thermal release tape. The electrical conductivity of these graphene-enhanced SU-8 substrates is about 1600 S/cm, markedly surpassing that of previously reported cell culture substrates. Immunofluorescence staining and Western blot analyses reveal that the electrically conductive graphene significantly enhances cardiomyocyte maturation and cardiac marker expression compared to bare SU-8 substrates. Cardiomyocytes cultured on graphene-transferred substrates exhibit conduction velocity approximately 3.4 times greater than that of the control group. Such improvements in cardiac marker expression, mechano-electrophysiological performance lead to better responsiveness to cardiovascular drugs, such as Verapamil and Isoproterenol. While the graphene monolayer does not fully replicate the complex environment found in native cardiac tissue, its use on SU-8 substrates offers a feasible approach for accelerating cardiomyocyte maturation and facilitating drug screening applications.
Collapse
Affiliation(s)
- Longlong Li
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Arunkumar Shanmugasundaram
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jongyun Kim
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Pooja P Kanade
- Centre for Quantum Materials and Technology, School of Mathematics and Physics, Queen's University Belfast, Northern Ireland, Belfast BT7 1NN, U.K
| | - Seong-Eung Cha
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Daeyun Lim
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Chil-Hyoung Lee
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Young-Baek Kim
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Bong-Kee Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
10
|
Kong C, Guo Z, Teng T, Yao Q, Yu J, Wang M, Ma Y, Wang P, Tang Q. Electroactive Nanomaterials for the Prevention and Treatment of Heart Failure: From Materials and Mechanisms to Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406206. [PMID: 39268781 DOI: 10.1002/smll.202406206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Heart failure (HF) represents a cardiovascular disease that significantly threatens global well-being and quality of life. Electroactive nanomaterials, characterized by their distinctive physical and chemical properties, emerge as promising candidates for HF prevention and management. This review comprehensively examines electroactive nanomaterials and their applications in HF intervention. It presents the definition, classification, and intrinsic characteristics of conductive, piezoelectric, and triboelectric nanomaterials, emphasizing their mechanical robustness, electrical conductivity, and piezoelectric coefficients. The review elucidates their applications and mechanisms: 1) early detection and diagnosis, employing nanomaterial-based sensors for real-time cardiac health monitoring; 2) cardiac tissue repair and regeneration, providing mechanical, chemical, and electrical stimuli for tissue restoration; 3) localized administration of bioactive biomolecules, genes, or pharmacotherapeutic agents, using nanomaterials as advanced drug delivery systems; and 4) electrical stimulation therapies, leveraging their properties for innovative pacemaker and neurostimulation technologies. Challenges in clinical translation, such as biocompatibility, stability, and scalability, are discussed, along with future prospects and potential innovations, including multifunctional and stimuli-responsive nanomaterials for precise HF therapies. This review encapsulates current research and future directions concerning the use of electroactive nanomaterials in HF prevention and management, highlighting their potential to innovating in cardiovascular medicine.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| |
Collapse
|
11
|
Vahidi M, Rizkalla AS, Mequanint K. Extracellular Matrix-Surrogate Advanced Functional Composite Biomaterials for Tissue Repair and Regeneration. Adv Healthc Mater 2024; 13:e2401218. [PMID: 39036851 DOI: 10.1002/adhm.202401218] [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: 04/01/2024] [Revised: 06/13/2024] [Indexed: 07/23/2024]
Abstract
Native tissues, comprising multiple cell types and extracellular matrix components, are inherently composites. Mimicking the intricate structure, functionality, and dynamic properties of native composite tissues represents a significant frontier in biomaterials science and tissue engineering research. Biomimetic composite biomaterials combine the benefits of different components, such as polymers, ceramics, metals, and biomolecules, to create tissue-template materials that closely simulate the structure and functionality of native tissues. While the design of composite biomaterials and their in vitro testing are frequently reviewed, there is a considerable gap in whole animal studies that provides insight into the progress toward clinical translation. Herein, we provide an insightful critical review of advanced composite biomaterials applicable in several tissues. The incorporation of bioactive cues and signaling molecules into composite biomaterials to mimic the native microenvironment is discussed. Strategies for the spatiotemporal release of growth factors, cytokines, and extracellular matrix proteins are elucidated, highlighting their role in guiding cellular behavior, promoting tissue regeneration, and modulating immune responses. Advanced composite biomaterials design challenges, such as achieving optimal mechanical properties, improving long-term stability, and integrating multifunctionality into composite biomaterials and future directions, are discussed. We believe that this manuscript provides the reader with a timely perspective on composite biomaterials.
Collapse
Affiliation(s)
- Milad Vahidi
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| | - Amin S Rizkalla
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, N6A5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, N6A5B9, Canada
| |
Collapse
|
12
|
Memarian P, Bagher Z, Asghari S, Aleemardani M, Seifalian A. Emergence of graphene as a novel nanomaterial for cardiovascular applications. NANOSCALE 2024; 16:12793-12819. [PMID: 38919053 DOI: 10.1039/d4nr00018h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CDs) are the foremost cause of death worldwide. Several promising therapeutic methods have been developed for this approach, including pharmacological, surgical intervention, cell therapy, or biomaterial implantation since heart tissue is incapable of regenerating and healing on its own. The best treatment for heart failure to date is heart transplantation and invasive surgical intervention, despite their invasiveness, donor limitations, and the possibility of being rejected by the patient's immune system. To address these challenges, research is being conducted on less invasive and efficient methods. Consequently, graphene-based materials (GBMs) have attracted a great deal of interest in the last decade because of their exceptional mechanical, electrical, chemical, antibacterial, and biocompatibility properties. An overview of GBMs' applications in the cardiovascular system has been presented in this article. Following a brief explanation of graphene and its derivatives' properties, the potential of GBMs to improve and restore cardiovascular system function by using them as cardiac tissue engineering, stents, vascular bypass grafts,and heart valve has been discussed.
Collapse
Affiliation(s)
- Paniz Memarian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sheida Asghari
- Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, S3 7HQ, UK.
- Department of Translational Health Science, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK.
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
| |
Collapse
|
13
|
Li Y, Cheng S, Shi H, Yuan R, Gao C, Wang Y, Zhang Z, Deng Z, Huang J. 3D embedded bioprinting of large-scale intestine with complex structural organization and blood capillaries. Biofabrication 2024; 16:045001. [PMID: 38914075 DOI: 10.1088/1758-5090/ad5b1b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024]
Abstract
Accurate reproduction of human intestinal structure and functionin vitrois of great significance for understanding the development and disease occurrence of the gut. However, mostin vitrostudies are often confined to 2D models, 2.5D organ chips or 3D organoids, which cannot fully recapitulate the tissue architecture, microenvironment and cell compartmentalization foundin vivo. Herein, a centimeter-scale intestine tissue that contains intestinal features, such as hollow tubular structure, capillaries and tightly connected epithelium with invivo-likering folds, crypt-villi, and microvilli is constructed by 3D embedding bioprinting. In our strategy, a novel photocurable bioink composed of methacrylated gelatin, methacrylated sodium alginate and poly (ethylene glycol) diacrylate is developed for the fabrication of intestinal model. The Caco-2 cells implanted in the lumen are induced by the topological structures of the model to derive microvilli, crypt-villi, and tight junctions, simulating the intestinal epithelial barrier. The human umbilical vein endothelial cells encapsulated within the model gradually form microvessels, mimicking the dense capillary network in the intestine. This intestine-like tissue, which closely resembles the structure and cell arrangement of the human gut, can act as a platform to predict the therapeutic and toxic side effects of new drugs on the intestine.
Collapse
Affiliation(s)
- Yuxuan Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Shengnan Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Haihua Shi
- Department of Gastrointestinal surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215001, People's Republic of China
| | - Renshun Yuan
- Department of Gastrointestinal surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215001, People's Republic of China
| | - Chen Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Yuhan Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zhijun Zhang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zongwu Deng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Jie Huang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| |
Collapse
|
14
|
Rezaei Z, Navarro Torres A, Ge D, Wang T, Méndez Terán EC, García Vera SE, Bassous NJ, Soria OYP, Ávila Ramírez AE, Flores Campos LM, Azuela Rosas DA, Hassan S, Khorsandi D, Jucaud V, Hussain MA, Khateeb A, Zhang YS, Lee H, Kim DH, Khademhosseini A, Dokmeci MR, Shin SR. Noninvasive and Continuous Monitoring of On-Chip Stem Cell Osteogenesis Using a Reusable Electrochemical Immunobiosensor. ACS Sens 2024; 9:2334-2345. [PMID: 38639453 DOI: 10.1021/acssensors.3c02165] [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: 04/20/2024]
Abstract
Noninvasive monitoring of biofabricated tissues during the biomanufacturing process is needed to obtain reproducible, healthy, and functional tissues. Measuring the levels of biomarkers secreted from tissues is a promising strategy to understand the status of tissues during biofabrication. Continuous and real-time information from cultivated tissues enables users to achieve scalable manufacturing. Label-free biosensors are promising candidates for detecting cell secretomes since they can be noninvasive and do not require labor-intensive processes such as cell lysing. Moreover, most conventional monitoring techniques are single-use, conducted at the end of the fabrication process, and, challengingly, are not permissive to in-line and continual detection. To address these challenges, we developed a noninvasive and continual monitoring platform to evaluate the status of cells during the biofabrication process, with a particular focus on monitoring the transient processes that stem cells go through during in vitro differentiation over extended periods. We designed and evaluated a reusable electrochemical immunosensor with the capacity for detecting trace amounts of secreted osteogenic markers, such as osteopontin (OPN). The sensor has a low limit of detection (LOD), high sensitivity, and outstanding selectivity in complex biological media. We used this OPN immunosensor to continuously monitor on-chip osteogenesis of human mesenchymal stem cells (hMSCs) cultured 2D and 3D hydrogel constructs inside a microfluidic bioreactor for more than a month and were able to observe changing levels of OPN secretion during culture. The proposed platform can potentially be adopted for monitoring a variety of biological applications and further developed into a fully automated system for applications in advanced cellular biomanufacturing.
Collapse
Affiliation(s)
- Zahra Rezaei
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Sharif University of Technology, Azadi Avenue, Tehran 11365-11155, Iran
| | - Andrea Navarro Torres
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Ting Wang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Eloísa Carolina Méndez Terán
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Stefany Elizabeth García Vera
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Nicole Joy Bassous
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Oscar Yael Perez Soria
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Alan Eduardo Ávila Ramírez
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
- Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Luis Mario Flores Campos
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Diego Arnoldo Azuela Rosas
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi 127788, United Arab Emirates
- Functional Biomaterials Group, Khalifa University, SAN Campus, Abu Dhabi 127788, United Arab Emirates
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Mohammad Asif Hussain
- Electrical and Computer Engineering Department, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulhameed Khateeb
- Electrical and Computer Engineering Department, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - HeaYeon Lee
- Mara Nanotech Inc., Hanmir Hall, Yongdang Campus, Pukyong National University, 365 Sinseon-ro, Nam-gu 48548, Republic of Korea
- MARA Nanotech New York INC., NY Designs, 29-10 Thomson Ave, Rm. C760, L.I.C., New York 11101, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Tang G, Li Z, Ding C, Zhao J, Xing X, Sun Y, Qiu X, Wang L. A cigarette filter-derived biomimetic cardiac niche for myocardial infarction repair. Bioact Mater 2024; 35:362-381. [PMID: 38379697 PMCID: PMC10876615 DOI: 10.1016/j.bioactmat.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Cell implantation offers an appealing avenue for heart repair after myocardial infarction (MI). Nevertheless, the implanted cells are subjected to the aberrant myocardial niche, which inhibits cell survival and maturation, posing significant challenges to the ultimate therapeutic outcome. The functional cardiac patches (CPs) have been proved to construct an elastic conductive, antioxidative, and angiogenic microenvironment for rectifying the aberrant microenvironment of the infarcted myocardium. More importantly, inducing implanted cardiomyocytes (CMs) adapted to the anisotropic arrangement of myocardial tissue by bioengineered structural cues within CPs are more conducive to MI repair. Herein, a functional Cig/(TA-Cu) CP served as biomimetic cardiac niche was fabricated based on structural anisotropic cigarette filter by modifying with tannic acid (TA)-chelated Cu2+ (TA-Cu complex) via a green method. This CP possessed microstructural anisotropy, electrical conductivity and mechanical properties similar to natural myocardium, which could promote elongation, orientation, maturation, and functionalization of CMs. Besides, the Cig/(TA-Cu) CP could efficiently scavenge reactive oxygen species, reduce CM apoptosis, ultimately facilitating myocardial electrical integration, promoting vascular regeneration and improving cardiac function. Together, our study introduces a functional CP that integrates multimodal cues to create a biomimetic cardiac niche and provides an effective strategy for cardiac repair.
Collapse
Affiliation(s)
- Guofeng Tang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Zhentao Li
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Thoracic and Cardiovascular Surgery, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, PR China
| | - Chengbin Ding
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Jiang Zhao
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Xianglong Xing
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Yan Sun
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Leyu Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| |
Collapse
|
16
|
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
|
17
|
Das S, Jegadeesan JT, Basu B. Gelatin Methacryloyl (GelMA)-Based Biomaterial Inks: Process Science for 3D/4D Printing and Current Status. Biomacromolecules 2024; 25:2156-2221. [PMID: 38507816 DOI: 10.1021/acs.biomac.3c01271] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tissue engineering for injured tissue replacement and regeneration has been a subject of investigation over the last 30 years, and there has been considerable interest in using additive manufacturing to achieve these goals. Despite such efforts, many key questions remain unanswered, particularly in the area of biomaterial selection for these applications as well as quantitative understanding of the process science. The strategic utilization of biological macromolecules provides a versatile approach to meet diverse requirements in 3D printing, such as printability, buildability, and biocompatibility. These molecules play a pivotal role in both physical and chemical cross-linking processes throughout the biofabrication, contributing significantly to the overall success of the 3D printing process. Among the several bioprintable materials, gelatin methacryloyl (GelMA) has been widely utilized for diverse tissue engineering applications, with some degree of success. In this context, this review will discuss the key bioengineering approaches to identify the gelation and cross-linking strategies that are appropriate to control the rheology, printability, and buildability of biomaterial inks. This review will focus on the GelMA as the structural (scaffold) biomaterial for different tissues and as a potential carrier vehicle for the transport of living cells as well as their maintenance and viability in the physiological system. Recognizing the importance of printability toward shape fidelity and biophysical properties, a major focus in this review has been to discuss the qualitative and quantitative impact of the key factors, including microrheological, viscoelastic, gelation, shear thinning properties of biomaterial inks, and printing parameters, in particular, reference to 3D extrusion printing of GelMA-based biomaterial inks. Specifically, we emphasize the different possibilities to regulate mechanical, swelling, biodegradation, and cellular functionalities of GelMA-based bio(material) inks, by hybridization techniques, including different synthetic and natural biopolymers, inorganic nanofillers, and microcarriers. At the close, the potential possibility of the integration of experimental data sets and artificial intelligence/machine learning approaches is emphasized to predict the printability, shape fidelity, or biophysical properties of GelMA bio(material) inks for clinically relevant tissues.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| | | | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
18
|
Kobeissi H, Jilberto J, Karakan MÇ, Gao X, DePalma SJ, Das SL, Quach L, Urquia J, Baker BM, Chen CS, Nordsletten D, Lejeune E. MicroBundleCompute: Automated segmentation, tracking, and analysis of subdomain deformation in cardiac microbundles. PLoS One 2024; 19:e0298863. [PMID: 38530829 PMCID: PMC10965069 DOI: 10.1371/journal.pone.0298863] [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] [Received: 08/31/2023] [Accepted: 02/01/2024] [Indexed: 03/28/2024] Open
Abstract
Advancing human induced pluripotent stem cell derived cardiomyocyte (hiPSC-CM) technology will lead to significant progress ranging from disease modeling, to drug discovery, to regenerative tissue engineering. Yet, alongside these potential opportunities comes a critical challenge: attaining mature hiPSC-CM tissues. At present, there are multiple techniques to promote maturity of hiPSC-CMs including physical platforms and cell culture protocols. However, when it comes to making quantitative comparisons of functional behavior, there are limited options for reliably and reproducibly computing functional metrics that are suitable for direct cross-system comparison. In addition, the current standard functional metrics obtained from time-lapse images of cardiac microbundle contraction reported in the field (i.e., post forces, average tissue stress) do not take full advantage of the available information present in these data (i.e., full-field tissue displacements and strains). Thus, we present "MicroBundleCompute," a computational framework for automatic quantification of morphology-based mechanical metrics from movies of cardiac microbundles. Briefly, this computational framework offers tools for automatic tissue segmentation, tracking, and analysis of brightfield and phase contrast movies of beating cardiac microbundles. It is straightforward to implement, runs without user intervention, requires minimal input parameter setting selection, and is computationally inexpensive. In this paper, we describe the methods underlying this computational framework, show the results of our extensive validation studies, and demonstrate the utility of exploring heterogeneous tissue deformations and strains as functional metrics. With this manuscript, we disseminate "MicroBundleCompute" as an open-source computational tool with the aim of making automated quantitative analysis of beating cardiac microbundles more accessible to the community.
Collapse
Affiliation(s)
- Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States of America
| | - Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - M. Çağatay Karakan
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Photonics Center, Boston University, Boston, MA, United States of America
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Xining Gao
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Shoshana L. Das
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Lani Quach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Jonathan Urquia
- Department of Electrical and Computer Engineering, New York Institute of Technology, New York, NY, United States of America
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - David Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, United States of America
- Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King’s Health Partners, King’s College London, King’s Health Partners, London, United Kingdom
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States of America
| |
Collapse
|
19
|
Wu Q, Xue R, Zhao Y, Ramsay K, Wang EY, Savoji H, Veres T, Cartmell SH, Radisic M. Automated fabrication of a scalable heart-on-a-chip device by 3D printing of thermoplastic elastomer nanocomposite and hot embossing. Bioact Mater 2024; 33:46-60. [PMID: 38024233 PMCID: PMC10654006 DOI: 10.1016/j.bioactmat.2023.10.019] [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: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
The successful translation of organ-on-a-chip devices requires the development of an automated workflow for device fabrication, which is challenged by the need for precise deposition of multiple classes of materials in micro-meter scaled configurations. Many current heart-on-a-chip devices are produced manually, requiring the expertise and dexterity of skilled operators. Here, we devised an automated and scalable fabrication method to engineer a Biowire II multiwell platform to generate human iPSC-derived cardiac tissues. This high-throughput heart-on-a-chip platform incorporated fluorescent nanocomposite microwires as force sensors, produced from quantum dots and thermoplastic elastomer, and 3D printed on top of a polystyrene tissue culture base patterned by hot embossing. An array of built-in carbon electrodes was embedded in a single step into the base, flanking the microwells on both sides. The facile and rapid 3D printing approach efficiently and seamlessly scaled up the Biowire II system from an 8-well chip to a 24-well and a 96-well format, resulting in an increase of platform fabrication efficiency by 17,5000-69,000% per well. The device's compatibility with long-term electrical stimulation in each well facilitated the targeted generation of mature human iPSC-derived cardiac tissues, evident through a positive force-frequency relationship, post-rest potentiation, and well-aligned sarcomeric apparatus. This system's ease of use and its capacity to gauge drug responses in matured cardiac tissue make it a powerful and reliable platform for rapid preclinical drug screening and development.
Collapse
Affiliation(s)
- Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Ruikang Xue
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering and The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Yimu Zhao
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Kaitlyn Ramsay
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Erika Yan Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Houman Savoji
- Institute of Biomedical Engineering and Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, Quebec, H3T 1J4, Canada
| | - Teodor Veres
- National Research Council of Canada, Boucherville, QC, J4B 6Y4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, M5S 3G8, Canada
| | - Sarah H. Cartmell
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering and The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| |
Collapse
|
20
|
Sacchi M, Sauter-Starace F, Mailley P, Texier I. Resorbable conductive materials for optimally interfacing medical devices with the living. Front Bioeng Biotechnol 2024; 12:1294238. [PMID: 38449676 PMCID: PMC10916519 DOI: 10.3389/fbioe.2024.1294238] [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: 09/14/2023] [Accepted: 01/02/2024] [Indexed: 03/08/2024] Open
Abstract
Implantable and wearable bioelectronic systems are arising growing interest in the medical field. Linking the microelectronic (electronic conductivity) and biological (ionic conductivity) worlds, the biocompatible conductive materials at the electrode/tissue interface are key components in these systems. We herein focus more particularly on resorbable bioelectronic systems, which can safely degrade in the biological environment once they have completed their purpose, namely, stimulating or sensing biological activity in the tissues. Resorbable conductive materials are also explored in the fields of tissue engineering and 3D cell culture. After a short description of polymer-based substrates and scaffolds, and resorbable electrical conductors, we review how they can be combined to design resorbable conductive materials. Although these materials are still emerging, various medical and biomedical applications are already taking shape that can profoundly modify post-operative and wound healing follow-up. Future challenges and perspectives in the field are proposed.
Collapse
Affiliation(s)
- Marta Sacchi
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
- Université Paris-Saclay, CEA, JACOB-SEPIA, Fontenay-aux-Roses, France
| | - Fabien Sauter-Starace
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| | - Pascal Mailley
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| | - Isabelle Texier
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| |
Collapse
|
21
|
Shin M, Lim J, Park Y, Lee JY, Yoon J, Choi JW. Carbon-based nanocomposites for biomedical applications. RSC Adv 2024; 14:7142-7156. [PMID: 38419681 PMCID: PMC10900039 DOI: 10.1039/d3ra08946k] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
Carbon nanomaterials have attracted significant attention in the biomedical field, including for biosensing, drug delivery, and tissue engineering applications. Based on their inherent properties such as their unique structure and high conductivity, carbon nanomaterials can overcome the current limitations in biomedical research such as poor stability of biomolecules, low sensitivity and selectivity of biosensors, and difficulty in precise drug delivery. In addition, recently, several novel nanomaterials have been integrated with carbon nanomaterials to develop carbon-based nanocomposites for application in biomedical research. In this review, we discuss recent studies on carbon-based nanocomposites and their biomedical applications. First, we discuss the representative carbon nanomaterials and nanocomposites composed of carbon and other novel nanomaterials. Next, applications of carbon nanomaterials and nanocomposites in the biomedical field are discussed according to topics in the biomedical field. We have discussed the recent studies on biosensors, drug delivery, and tissue engineering. In conclusion, we believe that this review provides the potential and applicability of carbon nanomaterials and their nanocomposites and suggests future directions of the application of carbon-based nanocomposites in biomedical applications.
Collapse
Affiliation(s)
- Minkyu Shin
- Department of Chemical & Biomolecular Engineering, Sogang University 35 Baekbeom-ro, Mapo-gu Seoul 04107 Republic of Korea
| | - Joungpyo Lim
- Department of Chemical & Biomolecular Engineering, Sogang University 35 Baekbeom-ro, Mapo-gu Seoul 04107 Republic of Korea
| | - Yongseon Park
- Department of Chemical & Biomolecular Engineering, Sogang University 35 Baekbeom-ro, Mapo-gu Seoul 04107 Republic of Korea
| | - Ji-Young Lee
- Department of Chemical & Biomolecular Engineering, Sogang University 35 Baekbeom-ro, Mapo-gu Seoul 04107 Republic of Korea
| | - Jinho Yoon
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea 43 Jibong-ro, Wonmi-gu Bucheon-si Gyeonggi-do 14662 Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University 35 Baekbeom-ro, Mapo-gu Seoul 04107 Republic of Korea
| |
Collapse
|
22
|
Wang Y, Yang S, Cai H, Hu H, Hu K, Sun Z, Liu R, Wei Y, Han L. A dual-crosslinking electroactive hydrogel based on gelatin methacrylate and dibenzaldehyde-terminated telechelic polyethylene glycol for 3D bio-printing. Sci Rep 2024; 14:4118. [PMID: 38374394 PMCID: PMC10876640 DOI: 10.1038/s41598-024-54853-9] [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: 10/23/2023] [Accepted: 02/17/2024] [Indexed: 02/21/2024] Open
Abstract
Gelatin was widely used as scaffold materials in 3D bio-printing due to its excellent bioactivity and availability and especially that their arginine-glycine-aspartic acid (RGD) sequences could efficiently promote cell adhesion and proliferation. In this study, an electroactive and 3D bio-printable hydrogel was prepared through a two-step chemical cross-linking process. Specifically, residual free amino groups of methacrylated gelatin (GelMA) were cross-linked with the aldehyde groups of dibenzaldehyde-terminated telechelic polyethylene glycol (DF-PEG) via Schiff base bonds, forming a gel at 37 °C. During the subsequent 3D bio-printing process, GelMA underwent UV curing, forming a secondary cross-linked network to the mechanical strength and stability of the printed structure. The uniform dispersion of carbon nanotubes (CNTs) in the GelMA/DF-PEG composite hydrogel significantly increased its conductivity. The optimized GelMA/DF-PEG composite hydrogel, i.e., 30% GelMA and 25% DF-PEG (G30D25-CNTs), exhibited superior bio-printability. When the content of CNTs was above 4%, the conductivity of G30D25-CNTs hydrogel exceeded 10-2 S/m, which satisfied the needs of cells for micro-current stimulation. Furthermore, the pore microstructures, swelling behavior, degradation ability and cell toxicity of G30D25-CNTs electroactive hydrogels were thoroughly evaluated. Thus, the G30D25-CNTs hydrogel with 4% MWCNTs could be considered for further application in electrical stimulation of tissue regeneration such as muscle and cardiac nerve tissue repair.
Collapse
Affiliation(s)
- Yulong Wang
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
- School of Physical Science and Technology, Guangxi University, Nanning, 530004, China
| | - Songsong Yang
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Heqing Cai
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Hailong Hu
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Kun Hu
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Zhicheng Sun
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Ruping Liu
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Yen Wei
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China.
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| | - Lu Han
- The Engineering Research Center of 3D Printing and Bio-Fabrication, Beijing Institute of Graphic Communication, Beijing, 102600, China.
| |
Collapse
|
23
|
Lee IC, Li YCE, Thomas JL, Lee MH, Lin HY. Recent advances using MXenes in biomedical applications. MATERIALS HORIZONS 2024; 11:876-902. [PMID: 38175543 DOI: 10.1039/d3mh01588b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
An MXene is a novel two-dimensional transition metal carbide or nitride, with a typical formula of Mn+1XnTx (M = transition metals, X = carbon or nitrogen, and T = functional groups). MXenes have found wide application in biomedicine and biosensing, owing to their high biocompatibility, abundant reactive surface groups, good conductivity, and photothermal properties. Applications include photo- and electrochemical sensors, energy storage, and electronics. This review will highlight recent applications of MXene and MXene-derived materials in drug delivery, tissue engineering, antimicrobial activity, and biosensors (optical and electrochemical). We further elaborate on recent developments in utilizing MXenes for photothermal cancer therapy, and we explore multimodal treatments, including the integration of chemotherapeutic agents or magnetic nanoparticles for enhanced therapeutic efficacy. The high surface area and reactivity of MXenes provide an interface to respond to the changes in the environment, allowing MXene-based drug carriers to respond to changes in pH, reactive oxygen species (ROS), and electrical signals for controlled release applications. Furthermore, the conductivity of MXene enables it to provide electrical stimulation for cultured cells and endows it with photocatalytic capabilities that can be used in antibiotic applications. Wearable and in situ sensors incorporating MXenes are also included. Major challenges and future development directions of MXenes in biomedical applications are also discussed. The remarkable properties of MXenes will undoubtedly lead to their increasing use in the applications discussed here, as well as many others.
Collapse
Affiliation(s)
- I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung, 40724, Taiwan
| | - James L Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, 700, Kaohsiung University Rd., Nan-Tzu District, Kaohsiung 81148, Taiwan.
| |
Collapse
|
24
|
He W, Deng J, Ma B, Tao K, Zhang Z, Ramakrishna S, Yuan W, Ye T. Recent Advancements of Bioinks for 3D Bioprinting of Human Tissues and Organs. ACS APPLIED BIO MATERIALS 2024; 7:17-43. [PMID: 38091514 DOI: 10.1021/acsabm.3c00806] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
3D bioprinting is recognized as a promising biomanufacturing technology that enables the reproducible and high-throughput production of tissues and organs through the deposition of different bioinks. Especially, bioinks based on loaded cells allow for immediate cellularity upon printing, providing opportunities for enhanced cell differentiation for organ manufacturing and regeneration. Thus, extensive applications have been found in the field of tissue engineering. The performance of the bioinks determines the functionality of the entire printed construct throughout the bioprinting process. It is generally expected that bioinks should support the encapsulated cells to achieve their respective cellular functions and withstand normal physiological pressure exerted on the printed constructs. The bioinks should also exhibit a suitable printability for precise deposition of the constructs. These characteristics are essential for the functional development of tissues and organs in bioprinting and are often achieved through the combination of different biomaterials. In this review, we have discussed the cutting-edge outstanding performance of different bioinks for printing various human tissues and organs in recent years. We have also examined the current status of 3D bioprinting and discussed its future prospects in relieving or curing human health problems.
Collapse
Affiliation(s)
- Wen He
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jinjun Deng
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Binghe Ma
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Tao
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhi Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, National University of Singapore, Singapore 117576, Singapore
| | - Weizheng Yuan
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Ye
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
25
|
Lisboa ES, Serafim C, Santana W, Dos Santos VLS, de Albuquerque-Junior RLC, Chaud MV, Cardoso JC, Jain S, Severino P, Souto EB. Nanomaterials-combined methacrylated gelatin hydrogels (GelMA) for cardiac tissue constructs. J Control Release 2024; 365:617-639. [PMID: 38043727 DOI: 10.1016/j.jconrel.2023.11.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Among non-communicable diseases, cardiovascular diseases are the most prevalent, accounting for approximately 17 million deaths per year. Despite conventional treatment, cardiac tissue engineering emerges as a potential alternative for the advancement and treatment of these patients, using biomaterials to replace or repair cardiac tissues. Among these materials, gelatin in its methacrylated form (GelMA) is a biodegradable and biocompatible polymer with adjustable biophysical properties. Furthermore, gelatin has the ability to replace and perform collagen-like functions for cell development in vitro. The interest in using GelMA hydrogels combined with nanomaterials is increasingly growing to promote the responsiveness to external stimuli and improve certain properties of these hydrogels by exploring the incorporation of nanomaterials into these hydrogels to serve as electrical signaling conductive elements. This review highlights the applications of electrically conductive nanomaterials associated with GelMA hydrogels for the development of structures for cardiac tissue engineering, by focusing on studies that report the combination of GelMA with nanomaterials, such as gold and carbon derivatives (carbon nanotubes and graphene), in addition to the possibility of applying these materials in 3D tissue engineering, developing new possibilities for cardiac studies.
Collapse
Affiliation(s)
- Erika S Lisboa
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Carine Serafim
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Wanessa Santana
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Victoria L S Dos Santos
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Ricardo L C de Albuquerque-Junior
- Post-Graduate Program in Dentistry, Department of Dentistry, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil; Department of Pathology, Health Sciences Center, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil
| | - Marco V Chaud
- Laboratory of Biomaterials and Nanotechnology of UNISO (LaBNUS), University of Sorocaba, Sorocaba, São Paulo, Brazil
| | - Juliana C Cardoso
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Sona Jain
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Patrícia Severino
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil.
| | - Eliana B Souto
- Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, MEDTECH, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
26
|
Zhou Y, Yue T, Ding Y, Tan H, Weng J, Luo S, Zheng X. Nanotechnology translation in vascular diseases: From design to the bench. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1919. [PMID: 37548140 DOI: 10.1002/wnan.1919] [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: 02/21/2022] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Atherosclerosis is a systemic pathophysiological condition contributing to the development of majority of polyvascular diseases. Nanomedicine is a novel and rapidly developing science. Due to their small size, nanoparticles are freely transported in vasculature, and have been widely employed as tools in analytical imaging techniques. Furthermore, the application of nanoparticles also allows target intervention, such as drug delivery and tissue engineering regenerative methods, in the management of major vascular diseases. Therefore, by summarizing the physical and chemical characteristics of common nanoparticles used in diagnosis and treatment of vascular diseases, we discuss the details of these applications from cellular, molecular, and in vivo perspectives in this review. Furthermore, we also summarize the status and challenges of the application of nanoparticles in clinical translation. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Yongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tong Yue
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Ding
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huiling Tan
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sihui Luo
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xueying Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
27
|
Yoon J, Han H, Jang J. Nanomaterials-incorporated hydrogels for 3D bioprinting technology. NANO CONVERGENCE 2023; 10:52. [PMID: 37968379 PMCID: PMC10651626 DOI: 10.1186/s40580-023-00402-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
In the field of tissue engineering and regenerative medicine, various hydrogels derived from the extracellular matrix have been utilized for creating engineered tissues and implantable scaffolds. While these hydrogels hold immense promise in the healthcare landscape, conventional bioinks based on ECM hydrogels face several challenges, particularly in terms of lacking the necessary mechanical properties required for 3D bioprinting process. To address these limitations, researchers are actively exploring novel nanomaterial-reinforced ECM hydrogels for both mechanical and functional aspects. In this review, we focused on discussing recent advancements in the fabrication of engineered tissues and monitoring systems using nanobioinks and nanomaterials via 3D bioprinting technology. We highlighted the synergistic benefits of combining numerous nanomaterials into ECM hydrogels and imposing geometrical effects by 3D bioprinting technology. Furthermore, we also elaborated on critical issues remaining at the moment, such as the inhomogeneous dispersion of nanomaterials and consequent technical and practical issues, in the fabrication of complex 3D structures with nanobioinks and nanomaterials. Finally, we elaborated on plausible outlooks for facilitating the use of nanomaterials in biofabrication and advancing the function of engineered tissues.
Collapse
Affiliation(s)
- Jungbin Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hohyeon Han
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Institute of Convergence Science, Yonsei University, Seoul, South Korea.
| |
Collapse
|
28
|
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
|
29
|
Gu Y, Zou Y, Huang Y, Liang R, Wu Y, Hu Y, Hong Y, Zhang X, Toh YC, Ouyang H, Zhang S. 3D-printed biomimetic scaffolds with precisely controlled and tunable structures guide cell migration and promote regeneration of osteochondral defect. Biofabrication 2023; 16:015003. [PMID: 37797606 DOI: 10.1088/1758-5090/ad0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 10/07/2023]
Abstract
Untreated osteochondral defects will develop into osteoarthritis, affecting patients' quality of life. Since articular cartilage and subchondral bone exhibit distinct biological characteristics, repairing osteochondral defects remains a major challenge. Previous studies have tried to fabricate multilayer scaffolds with traditional methods or 3D printing technology. However, the efficacy is unsatisfactory because of poor control over internal structures or a lack of integrity between adjacent layers, severely compromising repair outcomes. Therefore, there is a need for a biomimetic scaffold that can simultaneously boost osteochondral defect regeneration in both structure and function. Herein, an integrated bilayer scaffold with precisely controlled structures is successfully 3D-printed in one step via digital light processing (DLP) technology. The upper layer has both 'lotus- and radial-' distribution pores, and the bottom layer has 'lotus-' pores to guide and facilitate the migration of chondrocytes and bone marrow mesenchymal stem cells, respectively, to the defect area. Tuning pore sizes could modulate the mechanical properties of scaffolds easily. Results show that 3D-printed porous structures allow significantly more cells to infiltrate into the area of 'lotus- and radial-' distribution pores during cell migration assay, subcutaneous implantation, andin situtransplantation, which are essential for osteochondral repair. Transplantation of this 3D-printed bilayer scaffold exhibits a promising osteochondral repair effect in rabbits. Incorporation of Kartogenin into the upper layer of scaffolds further induces better cartilage formation. Combining small molecules/drugs and precisely size-controlled and layer-specific porous structure via DLP technology, this 3D-printed bilayer scaffold is expected to be a potential strategy for osteochondral regeneration.
Collapse
Affiliation(s)
- Yuqing Gu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yiwei Zou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yuxuan Huang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yicong Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yifan Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yi Hong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane 4059, Australia
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, People's Republic of China
- Department of Sports Medicine, School of Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, People's Republic of China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|
30
|
Gaytan SL, Beaven E, Gadad SS, Nurunnabi M. Progress and prospect of nanotechnology for cardiac fibrosis treatment. INTERDISCIPLINARY MEDICINE 2023; 1:e20230018. [PMID: 38089921 PMCID: PMC10712437 DOI: 10.1002/inmd.20230018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 02/01/2024]
Abstract
Cardiac fibrosis is the excessive accumulation of extracellular matrix components in the heart, leading to reduced cardiac functionality and heart failure. This review provides an overview of the therapeutic applications of nanotechnology for the treatment of cardiac fibrosis. We first delve into the fundamental pathophysiology of cardiac fibrosis, highlighting the key molecular players, including Matrix Metalloproteinases, Transforming Growth Factor-beta, and several growth factors, cytokines, and signaling molecules. Each target presents a unique opportunity to develop targeted nano-therapies. We then focus on recent advancements in nanotechnology and how nanoparticles can be engineered to deliver drugs or therapeutic genes. These advanced delivery approaches have shown significant potential to inhibit fibrosis-promoting factors, thereby mitigating the fibrotic response and potentially reversing disease progression. In addition, we discuss the challenges associated with developing and translating nanotechnology-based drug delivery systems, including ensuring biocompatibility, safety, and regulatory compliance. This review highlights how nanotechnology can bridge the gap between lab research and clinical practice for treating cardiac fibrosis.
Collapse
Affiliation(s)
- Samantha L. Gaytan
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Interdisciplinary Health SciencesCollege of Health SciencesThe University of Texas El PasoEl PasoTexasUSA
| | - Elfa Beaven
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Biomedical EngineeringCollege of EngineeringThe University of Texas El PasoEl PasoTexasUSA
| | - Shrikanth S. Gadad
- Center of Emphasis in CancerDepartment of Molecular and Translational MedicinePaul L. Foster School of MedicineTexas Tech University Health Sciences Center El PasoEl PasoTexasUSA
| | - Md Nurunnabi
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Interdisciplinary Health SciencesCollege of Health SciencesThe University of Texas El PasoEl PasoTexasUSA
- Department of Biomedical EngineeringCollege of EngineeringThe University of Texas El PasoEl PasoTexasUSA
- Border Biomedical Research CenterThe University of Texas El PasoEl PasoTexasUSA
| |
Collapse
|
31
|
Cao S, Wei Y, Bo R, Yun X, Xu S, Guan Y, Zhao J, Lan Y, Zhang B, Xiong Y, Jin T, Lai Y, Chang J, Zhao Q, Wei M, Shao Y, Quan Q, Zhang Y. Inversely engineered biomimetic flexible network scaffolds for soft tissue regeneration. SCIENCE ADVANCES 2023; 9:eadi8606. [PMID: 37756408 PMCID: PMC10530085 DOI: 10.1126/sciadv.adi8606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
Graft-host mechanical mismatch has been a longstanding issue in clinical applications of synthetic scaffolds for soft tissue regeneration. Although numerous efforts have been devoted to resolve this grand challenge, the regenerative performance of existing synthetic scaffolds remains limited by slow tissue growth (comparing to autograft) and mechanical failures. We demonstrate a class of rationally designed flexible network scaffolds that can precisely replicate nonlinear mechanical responses of soft tissues and enhance tissue regeneration via reduced graft-host mechanical mismatch. Such flexible network scaffold includes a tubular network frame containing inversely engineered curved microstructures to produce desired mechanical properties, with an electrospun ultrathin film wrapped around the network to offer a proper microenvironment for cell growth. Using rat models with sciatic nerve defects or Achilles tendon injuries, our network scaffolds show regenerative performances evidently superior to that of clinically approved electrospun conduit scaffolds and achieve similar outcomes to autologous nerve transplantation in prevention of target organ atrophy and recovery of static sciatic index.
Collapse
Affiliation(s)
- Shunze Cao
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Yu Wei
- Department of Orthopedic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100142, P.R. China
| | - Renheng Bo
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Xing Yun
- Department of Orthopedic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100142, P.R. China
| | - Shiwei Xu
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Yanjun Guan
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100142, P.R. China
- Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100142, Beijing, P.R. China
| | - Jianzhong Zhao
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Yu Lan
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Bin Zhang
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Yingjie Xiong
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100142, P.R. China
- Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100142, Beijing, P.R. China
| | - Tianqi Jin
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Yuchen Lai
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| | - Jiahui Chang
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Qing Zhao
- Department of Orthopedic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100142, P.R. China
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100142, P.R. China
- Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100142, Beijing, P.R. China
| | - Min Wei
- Department of Orthopedic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100142, P.R. China
| | - Yue Shao
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Qi Quan
- Department of Orthopedic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100142, P.R. China
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100142, P.R. China
- Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100142, Beijing, P.R. China
| | - Yihui Zhang
- AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
32
|
Kim SD, Kim K, Shin M. Recent advances in 3D printable conductive hydrogel inks for neural engineering. NANO CONVERGENCE 2023; 10:41. [PMID: 37679589 PMCID: PMC10484881 DOI: 10.1186/s40580-023-00389-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
Recently, the 3D printing of conductive hydrogels has undergone remarkable advances in the fabrication of complex and functional structures. In the field of neural engineering, an increasing number of reports have been published on tissue engineering and bioelectronic approaches over the last few years. The convergence of 3D printing methods and electrically conducting hydrogels may create new clinical and therapeutic possibilities for precision regenerative medicine and implants. In this review, we summarize (i) advancements in preparation strategies for conductive materials, (ii) various printing techniques enabling the fabrication of electroconductive hydrogels, (iii) the required physicochemical properties of the printed constructs, (iv) their applications in bioelectronics and tissue regeneration for neural engineering, and (v) unconventional approaches and outlooks for the 3D printing of conductive hydrogels. This review provides technical insights into 3D printable conductive hydrogels and encompasses recent developments, specifically over the last few years of research in the neural engineering field.
Collapse
Affiliation(s)
- Sung Dong Kim
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
| | - Kyoungryong Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mikyung Shin
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
33
|
He J, Sun Y, Gao Q, He C, Yao K, Wang T, Xie M, Yu K, Nie J, Chen Y, He Y. Gelatin Methacryloyl Hydrogel, from Standardization, Performance, to Biomedical Application. Adv Healthc Mater 2023; 12:e2300395. [PMID: 37115708 DOI: 10.1002/adhm.202300395] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/23/2023] [Indexed: 04/29/2023]
Abstract
Gelatin methacryloyl (GelMA), a photocurable hydrogel, is widely used in 3D culture, particularly in 3D bioprinting, due to its high biocompatibility, tunable physicochemical properties, and excellent formability. However, as the properties and performances of GelMA vary under different synthetic conditions, there is a lack of standardization, leading to conflicting results. In this study, a uniform standard is established to understand and enhance GelMA applications. First, the basic concept of GelMA and the density of the molecular network (DMN) are defined. Second, two properties, degrees of substitution and ratio of solid content, as the main measurable parameters determining the DMN are used. Third, the mechanisms and relationships between DMN and its performance in various applications in terms of porosity, viscosity, formability, mechanical strength, swelling, biodegradation, and cytocompatibility are theoretically explained. The main questions that are answered: what does performance mean, why is it important, how to optimize the basic parameters to improve the performance, and how to characterize it reasonably and accurately? Finally, it is hoped that this knowledge will eliminate the need for researchers to conduct tedious and repetitive pre-experiments, enable easy communication for achievements between groups under the same standard, and fully explore the potential of the GelMA hydrogel.
Collapse
Affiliation(s)
- Jing He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
| | - Chanfan He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ke Yao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tongyao Wang
- State Key Laboratory of Catalysis, National Laboratory for Clean Energy, 2011-Collaborative Innovation Center of Chemistry for Energy Materials, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Kang Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
34
|
Edrisi F, Baheiraei N, Razavi M, Roshanbinfar K, Imani R, Jalilinejad N. Potential of graphene-based nanomaterials for cardiac tissue engineering. J Mater Chem B 2023; 11:7280-7299. [PMID: 37427687 DOI: 10.1039/d3tb00654a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Cardiovascular diseases are the primary cause of death worldwide. Despite significant advances in pharmacological treatments and surgical interventions to restore heart function after myocardial infarction, it can progress to heart failure due to the restricted inherent potential of adult cardiomyocytes to self-regenerate. Hence, the evolution of new therapeutic methods is critical. Nowadays, novel approaches in tissue engineering have assisted in restoring biological and physical specifications of the injured myocardium and, hence, cardiac function. The incorporation of a supporting matrix that could mechanically and electronically support the heart tissue and stimulate the cells to proliferate and regenerate will be advantageous. Electroconductive nanomaterials can facilitate intracellular communication and aid synchronous contraction via electroactive substrate creation, preventing the issue of arrhythmia in the heart. Among a wide range of electroconductive materials, graphene-based nanomaterials (GBNs) are promising for cardiac tissue engineering (CTE) due to their outstanding features including high mechanical strength, angiogenesis, antibacterial and antioxidant properties, low cost, and scalable fabrication. In the present review, we discuss the effect of applying GBNs on angiogenesis, proliferation, and differentiation of implanted stem cells, their antibacterial and antioxidant properties, and their role in improving the electrical and mechanical properties of the scaffolds for CTE. Also, we summarize the recent research that has applied GBNs in CTE. Finally, we present a concise discussion on the challenges and prospects.
Collapse
Affiliation(s)
- Fatemeh Edrisi
- Modern Technologies in Engineering Group, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran.
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, Florida 32827, USA
- Department of Material Sciences and Engineering, University of Central Florida, Orlando, Florida 32816, USA
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054 Erlangen, Germany
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran 1591634311, Iran
| | - Negin Jalilinejad
- Biomaterial Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
35
|
Lee M, Park J, Choe G, Lee S, Kang BG, Jun JH, Shin Y, Kim MC, Kim YS, Ahn Y, Lee JY. A Conductive and Adhesive Hydrogel Composed of MXene Nanoflakes as a Paintable Cardiac Patch for Infarcted Heart Repair. ACS NANO 2023. [PMID: 37339066 DOI: 10.1021/acsnano.3c00933] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Myocardial infarction (MI) is a major cause of death worldwide. After the occurrence of MI, the heart frequently undergoes serious pathological remodeling, leading to excessive dilation, electrical disconnection between cardiac cells, and fatal functional damage. Hence, extensive efforts have been made to suppress pathological remodeling and promote the repair of the infarcted heart. In this study, we developed a hydrogel cardiac patch that can provide mechanical support, electrical conduction, and tissue adhesiveness to aid in the recovery of an infarcted heart function. Specifically, we developed a conductive and adhesive hydrogel (CAH) by combining the two-dimensional titanium carbide (Ti3C2Tx) MXene with natural biocompatible polymers [i.e., gelatin and dextran aldehyde (dex-ald)]. The CAH was formed within 250 s of mixing the precursor solution and could be painted. The hydrogel containing 3.0 mg/mL MXene, 10% gelatin, and 5% dex-ald exhibited appropriate material characteristics for cardiac patch applications, including a uniform distribution of MXene, a high electrical conductivity (18.3 mS/cm), cardiac tissue-like elasticity (30.4 kPa), strong tissue adhesion (6.8 kPa), and resistance to various mechanical deformations. The CAH was cytocompatible and induced cardiomyocyte (CM) maturation in vitro, as indicated by the upregulation of connexin 43 expression and a faster beating rate. Furthermore, CAH could be painted onto the heart tissue and remained stably adhered to the beating epicardium. In vivo animal studies revealed that CAH cardiac patch treatment significantly improved cardiac function and alleviated the pathological remodeling of an infarcted heart. Thus, we believe that our MXene-based CAH can potentially serve as a promising platform for the effective repair of various electroactive tissues including the heart, muscle, and nerve tissues.
Collapse
Affiliation(s)
- Mingyu Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sanghun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University, Gwangju 61005, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University, Gwangju 61005, Republic of Korea
| | - Yoonmin Shin
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61005, Republic of Korea
| | - Min Chul Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61005, Republic of Korea
- Chonnam National University Medical School, Gwangju 61005, Republic of Korea
| | - Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University, Gwangju 61005, Republic of Korea
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61005, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University, Gwangju 61005, Republic of Korea
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61005, Republic of Korea
- Chonnam National University Medical School, Gwangju 61005, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
36
|
Adhikari B, Stager MA, Krebs MD. Cell-instructive biomaterials in tissue engineering and regenerative medicine. J Biomed Mater Res A 2023; 111:660-681. [PMID: 36779265 DOI: 10.1002/jbm.a.37510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/14/2023]
Abstract
The field of biomaterials aims to improve regenerative outcomes or scientific understanding for a wide range of tissue types and ailments. Biomaterials can be fabricated from natural or synthetic sources and display a plethora of mechanical, electrical, and geometrical properties dependent on their desired application. To date, most biomaterial systems designed for eventual translation to the clinic rely on soluble signaling moieties, such as growth factors, to elicit a specific cellular response. However, these soluble factors are often limited by high cost, convoluted synthesis, low stability, and difficulty in regulation, making the translation of these biomaterials systems to clinical or commercial applications a long and arduous process. In response to this, significant effort has been dedicated to researching cell-directive biomaterials which can signal for specific cell behavior in the absence of soluble factors. Cells of all tissue types have been shown to be innately in tune with their microenvironment, which is a biological phenomenon that can be exploited by researchers to design materials that direct cell behavior based on their intrinsic characteristics. This review will focus on recent developments in biomaterials that direct cell behavior using biomaterial properties such as charge, peptide presentation, and micro- or nano-geometry. These next generation biomaterials could offer significant strides in the development of clinically relevant medical devices which improve our understanding of the cellular microenvironment and enhance patient care in a variety of ailments.
Collapse
Affiliation(s)
- Bikram Adhikari
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Michael A Stager
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Melissa D Krebs
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, USA
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| |
Collapse
|
37
|
Tan Y, Lu T, Chen Y, Witman N, Yan B, Yang L, Liu M, Gong Y, Ai X, Luo R, Wang H, Wang W, Fu W. Engineering a conduction-consistent cardiac patch with graphene oxide modified butterfly wings and human pluripotent stem cell-derived cardiomyocytes. Bioeng Transl Med 2023; 8:e10522. [PMID: 37206241 PMCID: PMC10189447 DOI: 10.1002/btm2.10522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 03/12/2023] [Accepted: 03/29/2023] [Indexed: 05/21/2023] Open
Abstract
Engineering a conduction-consistent cardiac patch has direct implications to biomedical research. However, there is difficulty in obtaining and maintaining a system that allows researchers to study physiologically relevant cardiac development, maturation, and drug screening due to the issues around inconsistent contractions of cardiomyocytes. Butterfly wings have special nanostructures arranged in parallel, which could help generate the alignment of cardiomyocytes to better mimic the natural heart tissue structure. Here, we construct a conduction-consistent human cardiac muscle patch by assembling human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on graphene oxide (GO) modified butterfly wings. We also show this system functions as a versatile model to study human cardiomyogenesis by assembling human induced pluripotent stem cell-derived cardiac progenitor cells (hiPSC-CPCs) on the GO modified butterfly wings. The GO modified butterfly wing platform facilitated the parallel orientation of hiPSC-CMs, enhanced relative maturation as well as improved conduction consistency of the cardiomyocytes. In addition, GO modified butterfly wings enhanced the proliferation and maturation characteristics of the hiPSC-CPCs. In accordance with data obtained from RNA-sequencing and gene signatures, assembling hiPSC-CPCs on GO modified butterfly wings stimulated the differentiation of the progenitors into relatively mature hiPSC-CMs. These characteristics and capabilities of GO modified butterfly wings make them an ideal platform for heart research and drug screening.
Collapse
Affiliation(s)
- Yao Tan
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Tingting Lu
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Ying Chen
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Nevin Witman
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Bingqian Yan
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Li Yang
- Department of AnesthesiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Minglu Liu
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Runjiao Luo
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Huijing Wang
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Wang
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Fu
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Tissue EngineeringShanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
38
|
Sathuvan M, Thangam R, Cheong KL, Kang H, Liu Y. κ-Carrageenan-essential oil loaded composite biomaterial film facilitates mechanosensing and tissue regenerative wound healing. Int J Biol Macromol 2023; 241:124490. [PMID: 37076080 DOI: 10.1016/j.ijbiomac.2023.124490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Polysaccharides κ-carrageenan (κ-Car) have become a predominant source in developing bioactive materials. We aimed to develop biopolymer composite materials of κ-Car with coriander essential oil (CEO) (κ-Car-CEO) films for fibroblast-associated wound healing. Initially, we loaded the CEO in to κ-Car and CEO through homogenization and ultrasonication to fabricate composite film bioactive materials. After performing morphological and chemical characterizations, we validated the developed material functionalities in both in vitro and in vivo models. The chemical and morphological analysis with physical structure, swelling ratio, encapsulation efficiency, CEO release, and water barrier properties of films examined and showed the structural interaction of κ-Car and CEO-loaded into the polymer network. Furthermore, the bioactive applications of CEO release showed initial burst release followed by controlled release from the κ-Car composite film with fibroblast (L929) cell adhesive capabilities and mechanosensing. Our results proved that the CEO-loaded into the κ-Car film impacts cell adhesion, F-actin organization, and collagen synthesis, followed by in vitro mechanosensing activation, further promoting wound healing in vivo. Our innovative perspectives of active polysaccharide (κ-Car)-based CEO functional film materials could potentially accomplish regenerative medicine.
Collapse
Affiliation(s)
- Malairaj Sathuvan
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea; Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Republic of Korea
| | - Kit-Leong Cheong
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yang Liu
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China.
| |
Collapse
|
39
|
Shi T, Wang P, Ren Y, Zhang W, Ma J, Li S, Tan X, Chi B. Conductive Hydrogel Patches with High Elasticity and Fatigue Resistance for Cardiac Microenvironment Remodeling. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 36880699 DOI: 10.1021/acsami.2c22673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Remodeling the conductive zone to assist normal myocardial contraction and relaxation during myocardial fibrosis has become the primary concern of myocardial infarction (MI) regeneration. Herein, we report an unbreakable and self-recoverable hyaluronic acid conductive cardiac patch for MI treatment, which can maintain structural integrity under mechanical load and integrate mechanical and electrical conduction and biological cues to restore cardiac electrical conduction and diastolic contraction function. Using the free carboxyl groups and aldehyde groups in the hydrogel system, excellent adhesion properties are achieved in the interface between the myocardial patch and the tissue, which can be closely integrated with the rabbit myocardial tissue, reducing the need for suture. Interestingly, the hydrogel patch exhibits sensitive conductivity (ΔR/R0 ≈ 2.5) for 100 cycles and mechanical stability for 500 continuous loading cycles without collapse, which allows the patch to withstand mechanical damage caused by sustained contraction and relaxation of the myocardial tissue. Moreover, considering the oxidative stress state caused by excessive ROS in the MI area, we incorporated Rg1 into the hydrogel to improve the abnormal myocardial microenvironment, which achieved more than 80% free radicalscavenging efficiency in the local infarcted region and promoted myocardial reconstruction. Overall, these Rg1-loaded conductive hydrogels with highly elastic fatigue resistance have great potential in restoring the abnormal electrical conduction pathway and promoting the myocardial microenvironment, thereby repairing the heart and improving the cardiac function.
Collapse
Affiliation(s)
- Tianqi Shi
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Penghui Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yanhan Ren
- University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, United States
| | - Wenjie Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Juping Ma
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Shuang Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Xiaoyan Tan
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials Nanjing Tech University, Nanjing 211816, China
| | - Bo Chi
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
40
|
Nanocomposite Hydrogels as Functional Extracellular Matrices. Gels 2023; 9:gels9020153. [PMID: 36826323 PMCID: PMC9957407 DOI: 10.3390/gels9020153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Over recent years, nano-engineered materials have become an important component of artificial extracellular matrices. On one hand, these materials enable static enhancement of the bulk properties of cell scaffolds, for instance, they can alter mechanical properties or electrical conductivity, in order to better mimic the in vivo cell environment. Yet, many nanomaterials also exhibit dynamic, remotely tunable optical, electrical, magnetic, or acoustic properties, and therefore, can be used to non-invasively deliver localized, dynamic stimuli to cells cultured in artificial ECMs in three dimensions. Vice versa, the same, functional nanomaterials, can also report changing environmental conditions-whether or not, as a result of a dynamically applied stimulus-and as such provide means for wireless, long-term monitoring of the cell status inside the culture. In this review article, we present an overview of the technological advances regarding the incorporation of functional nanomaterials in artificial extracellular matrices, highlighting both passive and dynamically tunable nano-engineered components.
Collapse
|
41
|
Tan Y, Chen Y, Lu T, Witman N, Yan B, Gong Y, Ai X, Yang L, Liu M, Luo R, Wang H, Ministrini S, Dong W, Wang W, Fu W. Engineering a conduction-consistent cardiac patch with rGO/PLCL electrospun nanofibrous membranes and human iPSC-derived cardiomyocytes. Front Bioeng Biotechnol 2023; 11:1094397. [PMID: 36845196 PMCID: PMC9944832 DOI: 10.3389/fbioe.2023.1094397] [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: 11/10/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
The healthy human heart has special directional arrangement of cardiomyocytes and a unique electrical conduction system, which is critical for the maintenance of effective contractions. The precise arrangement of cardiomyocytes (CMs) along with conduction consistency between CMs is essential for enhancing the physiological accuracy of in vitro cardiac model systems. Here, we prepared aligned electrospun rGO/PLCL membranes using electrospinning technology to mimic the natural heart structure. The physical, chemical and biocompatible properties of the membranes were rigorously tested. We next assembled human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on electrospun rGO/PLCL membranes in order to construct a myocardial muscle patch. The conduction consistency of cardiomyocytes on the patches were carefully recorded. We found that cells cultivated on the electrospun rGO/PLCL fibers presented with an ordered and arranged structure, excellent mechanical properties, oxidation resistance and effective guidance. The addition of rGO was found to be beneficial for the maturation and synchronous electrical conductivity of hiPSC-CMs within the cardiac patch. This study verified the possibility of using conduction-consistent cardiac patches to enhance drug screening and disease modeling applications. Implementation of such a system could one day lead to in vivo cardiac repair applications.
Collapse
Affiliation(s)
- Yao Tan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Chen
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Lu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nevin Witman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Bingqian Yan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minglu Liu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Runjiao Luo
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland,Department of Medicine and Surgery, Internal Medicine, Angiology and Atherosclerosis, University of Perugia, Perugia, Italy
| | - Wei Dong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| |
Collapse
|
42
|
Kumar R, Parashar A. Atomistic simulations of pristine and nanoparticle reinforced hydrogels: A review. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2023. [DOI: 10.1002/wcms.1655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Raju Kumar
- Department of Mechanical and Industrial Engineering Indian Institute of Technology Roorkee Uttarakhand India
| | - Avinash Parashar
- Department of Mechanical and Industrial Engineering Indian Institute of Technology Roorkee Uttarakhand India
| |
Collapse
|
43
|
Wang Y, Lv Q, Chen Y, Xu L, Feng M, Xiong Z, Li J, Ren J, Liu J, Liu B. Bilayer hydrogel dressing with lysozyme-enhanced photothermal therapy for biofilm eradication and accelerated chronic wound repair. Acta Pharm Sin B 2023; 13:284-297. [PMID: 36811095 PMCID: PMC9939289 DOI: 10.1016/j.apsb.2022.03.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022] Open
Abstract
Biofilms are closely associated with the tough healing and dysfunctional inflammation of chronic wounds. Photothermal therapy (PTT) emerged as a suitable alternative which could destroy the structure of biofilms with local physical heat. However, the efficacy of PTT is limited because the excessive hyperthermia could damage surrounding tissues. Besides, the difficult reserve and delivery of photothermal agents makes PTT hard to eradicate biofilms as expectation. Herein, we present a GelMA-EGF/Gelatin-MPDA-LZM bilayer hydrogel dressing to perform lysozyme-enhanced PTT for biofilms eradication and a further acceleration to the repair of chronic wounds. Gelatin was used as inner layer hydrogel to reserve lysozyme (LZM) loaded mesoporous polydopamine (MPDA) (MPDA-LZM) nanoparticles, which could rapidly liquefy while temperature rising so as to achieve a bulk release of nanoparticles. MPDA-LZM nanoparticles serve as photothermal agents with antibacterial capability, could deeply penetrate and destroy biofilms. In addition, the outer layer hydrogel consisted of gelatin methacryloyl (GelMA) and epidermal growth factor (EGF) promoted wound healing and tissue regeneration. It displayed remarkable efficacy on alleviating infection and accelerating wound healing in vivo. Overall, the innovative therapeutic strategy we came up with has significant effect on biofilms eradication and shows promising application in promoting the repair of clinical chronic wounds.
Collapse
Affiliation(s)
- Yizhen Wang
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Qijun Lv
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - You Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Langtao Xu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Miao Feng
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiyong Xiong
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jiajun Li
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jie Ren
- Department of Ultrasound, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jie Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Bo Liu
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
44
|
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: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
45
|
Han N, Zhang W, Fang XX, Li QC, Pi W. Reduced graphene oxide-embedded nerve conduits loaded with bone marrow mesenchymal stem cell-derived extracellular vesicles promote peripheral nerve regeneration. Neural Regen Res 2023. [PMID: 35799543 PMCID: PMC9241414 DOI: 10.4103/1673-5374.343889] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We previously combined reduced graphene oxide (rGO) with gelatin-methacryloyl (GelMA) and polycaprolactone (PCL) to create an rGO-GelMA-PCL nerve conduit and found that the conductivity and biocompatibility were improved. However, the rGO-GelMA-PCL nerve conduits differed greatly from autologous nerve transplants in their ability to promote the regeneration of injured peripheral nerves and axonal sprouting. Extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSCs) can be loaded into rGO-GelMA-PCL nerve conduits for repair of rat sciatic nerve injury because they can promote angiogenesis at the injured site. In this study, 12 weeks after surgery, sciatic nerve function was measured by electrophysiology and sciatic nerve function index, and myelin sheath and axon regeneration were observed by electron microscopy, immunohistochemistry, and immunofluorescence. The regeneration of microvessel was observed by immunofluorescence. Our results showed that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles were superior to rGO-GelMA-PCL conduits alone in their ability to increase the number of newly formed vessels and axonal sprouts at the injury site as well as the recovery of neurological function. These findings indicate that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles can promote peripheral nerve regeneration and neurological function recovery, and provide a new direction for the curation of peripheral nerve defect in the clinic.
Collapse
|
46
|
Lee M, Kim MC, Lee JY. Nanomaterial-Based Electrically Conductive Hydrogels for Cardiac Tissue Repair. Int J Nanomedicine 2022; 17:6181-6200. [PMID: 36531116 PMCID: PMC9748845 DOI: 10.2147/ijn.s386763] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/23/2022] [Indexed: 08/28/2023] Open
Abstract
Cardiovascular disease is one of major causes of deaths, and its incidence has gradually increased worldwide. For cardiovascular diseases, several therapeutic approaches, such as drugs, cell-based therapy, and heart transplantation, are currently employed; however, their therapeutic efficacy and/or practical availability are still limited. Recently, biomaterial-based tissue engineering approaches have been recognized as promising for regenerating cardiac function in patients with cardiovascular diseases, including myocardial infarction (MI). In particular, materials mimicking the characteristics of native cardiac tissues can potentially prevent pathological progression and promote cardiac repair of the heart tissues post-MI. The mechanical (softness) and electrical (conductivity) properties of biomaterials as non-biochemical cues can improve the cardiac functions of infarcted hearts by mitigating myocardial cell death and subsequent fibrosis, which often leads to cardiac tissue stiffening and high electrical resistance. Consequently, electrically conductive hydrogels that can provide mechanical strength and augment the electrical activity of the infarcted heart tissue are considered new functional materials capable of mitigating the pathological progression to heart failure and stimulating cardiac regeneration. In this review, we highlight nanomaterial-incorporated hydrogels that can induce cardiac repair after MI. Nanomaterials, including carbon-based nanomaterials and recently discovered two-dimensional nanomaterials, offer great opportunities for developing functional conductive hydrogels owing to their excellent electrical conductivity, large surface area, and ease of modification. We describe recent results using nanomaterial-incorporated conductive hydrogels as cardiac patches and injectable hydrogels for cardiac repair. While further evaluations are required to confirm the therapeutic efficacy and toxicity of these materials, they could potentially be used for the regeneration of other electrically active tissues, such as nerves and muscles.
Collapse
Affiliation(s)
- Mingyu Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Min Chul Kim
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
47
|
Li Y, Liu Y, Peng B, Li X, Fang T, Liu S, Liu J, Li B, Li F. Stretchable, conductive, breathable and moisture-sensitive e-skin based on CNTs/graphene/GelMA mat for wound monitoring. BIOMATERIALS ADVANCES 2022; 143:213172. [PMID: 36343392 DOI: 10.1016/j.bioadv.2022.213172] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/07/2022] [Accepted: 10/22/2022] [Indexed: 06/16/2023]
Abstract
Deep skin wound needs a long wound healing process, in which external force on skin around wound can result in a sharp pain, wound re-damage and interstitial fluid flowing out, increasing the risk of deterioration and even amputation. While the conventional wound dressings cannot provide timely feedback of abnormal wound status and lose best time for wound treatment, real-time monitoring wound status is thus urgently needed for wound management. In this work, a breathable and stretchable electronic skin (i.e., e-skin) named CNTs/graphene/GelMA mat has been developed through electrospinning, ice-templating and in-situ loading method for evaluating wound status. The obtained porosity, swelling ratio and vapor transmission rate of the CNTs/graphene/GelMA mat are 55 %, 180 % and 3378.2 h-1 day-1, respectively. And owing to the good porous, nanofibrous architecture and excellent breathability of the mat, L929 cells grow and well spread on the CNTs/graphene/GelMA mat. In addition, the gauge factors of the prepared conductive CNTs/graphene/GelMA mat as a strain sensor are 15.4 and 72.9 in the strain ranges of 0-70 % and 70-85 %, respectively, matching the mechanical performance of human skin. The sensitivity coefficient of the mat for moisture sensing is 12.05, indicating its high efficiency for monitoring and warning interstitial fluid outflow from wound. Furthermore, the integration of CNTs/graphene/GelMA mat with a portable device is feasible to monitor strain and moisture on a rat model with abdominal wound. The healing process of the wounds treated with CNTs/graphene/GelMA mat is similar to that of GelMA mat, indicating that the dosage of CNTs and graphene in the CNTs/graphene/GelMA mat has negligible effect on the mat histocompatibility. The CNTs/graphene/GelMA mat demonstrates the application potential in wound management, home medical diagnosis and human-machine interactions.
Collapse
Affiliation(s)
- Yingchun Li
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China; Advanced Interdisciplinary Research Center for Flexible Electronics, School of Microelectronics, Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an 710071, P. R. China
| | - Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, P. R. China
| | - Bo Peng
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, P. R. China
| | - Xinyue Li
- Advanced Interdisciplinary Research Center for Flexible Electronics, School of Microelectronics, Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an 710071, P. R. China
| | - Tianshu Fang
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Shuai Liu
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Jiachen Liu
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Bo Li
- State key Laboratory for Manufacturing Engineering System, Shaanxi Province Key Laboratory for Intelligent Robots, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| |
Collapse
|
48
|
Cao L, Huang Y, Parakhonskiy B, Skirtach AG. Nanoarchitectonics beyond perfect order - not quite perfect but quite useful. NANOSCALE 2022; 14:15964-16002. [PMID: 36278502 DOI: 10.1039/d2nr02537j] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nanoarchitectonics, like architectonics, allows the design and building of structures, but at the nanoscale. Unlike those in architectonics, and even macro-, micro-, and atomic-scale architectonics, the assembled structures at the nanoscale do not always follow the projected design. In fact, they do follow the projected design but only for self-assembly processes producing structures with perfect order. Here, we look at nanoarchitectonics allowing the building of nanostructures without a perfect arrangement of building blocks. Here, fabrication of structures from molecules, polymers, nanoparticles, and nanosheets to polymer brushes, layer-by-layer assembly structures, and hydrogels through self-assembly processes is discussed, where perfect order is not necessarily the aim to be achieved. Both planar substrate and spherical template-based assemblies are discussed, showing the challenging nature of research in this field and the usefulness of such structures for numerous applications, which are also discussed here.
Collapse
Affiliation(s)
- Lin Cao
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| | - Yanqi Huang
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| | - Bogdan Parakhonskiy
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| | - Andre G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
49
|
Hui Y, Yan Z, Yang H, Xu X, Yuan WE, Qian Y. Graphene Family Nanomaterials for Stem Cell Neurogenic Differentiation and Peripheral Nerve Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:4741-4759. [PMID: 36102324 DOI: 10.1021/acsabm.2c00663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Stem cells play a critical role in peripheral nerve regeneration. Nerve scaffolds fabricated by specific materials can help induce the neurogenic differentiation of stem cells. Therefore, it is a potential strategy to enhance therapeutic efficiency. Graphene family nanomaterials are widely applied in repairing peripheral nerves. However, the mechanism underlying the pro-regeneration effects remains elusive. In this review, we first discuss the properties of graphene family nanomaterials, including monolayer and multilayer graphene, few-layer graphene, graphene oxide, reduced graphene oxide, and graphene quantum dots. We also introduce their applications in regulating stem cell differentiation. Then, we review the potential mechanisms of the neurogenic differentiation of stem cells facilitated by the materials. Finally, we discuss the existing challenges in this field to advance the development of nerve biomaterials.
Collapse
Affiliation(s)
- Yuxuan Hui
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| | - Zhiwen Yan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| | - Hao Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| | - Xingxing Xu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| |
Collapse
|
50
|
Su G, Li G, Wang W, Xu L. Application Prospect and Preliminary Exploration of GelMA in Corneal Stroma Regeneration. Polymers (Basel) 2022; 14:4227. [PMID: 36236174 PMCID: PMC9571618 DOI: 10.3390/polym14194227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Corneal regeneration has become a prominent study area in recent decades. Because the corneal stroma contributes about 90% of the corneal thickness in the corneal structure, corneal stromal regeneration is critical for the treatment of cornea disease. Numerous materials, including deacetylated chitosan, hydrophilic gel, collagen, gelatin methacrylate (GelMA), serine protein, glycerol sebacate, and decellularized extracellular matrix, have been explored for keratocytes regeneration. GelMA is one of the most prominent materials, which is becoming more and more popular because of its outstanding three-dimensional scaffold structure, strong mechanics, good optical transmittance, and biocompatibility. This review discussed recent research on corneal stroma regeneration materials and related GelMA.
Collapse
Affiliation(s)
| | | | | | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|