1
|
Chen S, Tan L, Serpooshan V, Chen H. A 3D bioprinted adhesive tissue engineering scaffold to repair ischemic heart injury. Biomater Sci 2024. [PMID: 39639799 DOI: 10.1039/d4bm00988f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Adhesive tissue engineering scaffold (ATES) devices can be secured on tissues by relying on their intrinsic adhesive properties, hence, avoiding the complications such as host tissue/scaffold damage that are associated with conventional scaffold fixation methods like suturing or bioglue. This study introduces a new generation of three-dimensional (3D) bioprinted ATES systems for use as cardiac patches to regenerate the adult human heart. Tyramine-modified methacrylated hyaluronic acid (HAMA-tyr), gelatin methacrylate (GelMA), and gelatin were used to create the hybrid bioink formulation with self-adhesive properties. ATESs were bioprinted and further modified to improve the adhesion properties. In-depth characterization of printing fidelity, pore size, mechanical properties, swelling behavior, as well as biocompatibility was used to create ATESs with optimal biological function. Following in vitro testing, the ATESs were tested in a mouse model of myocardial infarction to study the scaffold adhesive strength in biological milieu. The method developed in this study can be used to manufacture off-the-shelf ATESs with complex cellular and extracellular architecture, with robust potential for clinical translation into a variety of personalized tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Lindan Tan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| |
Collapse
|
2
|
Jafari A, Al‐Ostaz A, Nouranian S. Recent Advances in Multifunctional Naturally Derived Bioadhesives for Tissue Engineering and Wound Management. POLYM ADVAN TECHNOL 2024; 35. [DOI: 10.1002/pat.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/17/2024] [Indexed: 01/03/2025]
Abstract
ABSTRACTRecent advancements in naturally derived bioadhesives have transformed their application across diverse medical fields, including tissue engineering, wound management, and surgery. This review focuses on the innovative development and multifunctional nature of these bioadhesives, particularly emphasizing their role in enhancing adhesion performance in wet environments and optimizing mechanical properties for use in dynamic tissues. Key areas covered include the chemical and physical mechanisms of adhesion, the incorporation of multi‐adhesion strategies that combine covalent and non‐covalent bonding, and bioinspired designs mimicking natural adhesives such as those of barnacles and mussels. Additionally, the review discusses emerging applications of bioadhesives in the regeneration of musculoskeletal, cardiac, neural, and ocular tissues, highlighting the potential for bioadhesive‐based therapies in complex biological settings. Despite substantial progress, challenges such as scaling lab‐based innovations for clinical use and overcoming environmental and mechanical constraints remain critical. Ongoing research in bioadhesive technologies aims to bridge these gaps, promising significant improvements in medical adhesives tailored for diverse therapeutic needs.
Collapse
Affiliation(s)
- Aliakbar Jafari
- Department of Chemical Engineering University of Mississippi University Mississippi USA
- Center for Graphene Research and Innovation University of Mississippi University Mississippi USA
| | - Ahmed Al‐Ostaz
- Center for Graphene Research and Innovation University of Mississippi University Mississippi USA
- Department of Civil Engineering University of Mississippi University Mississippi USA
| | - Sasan Nouranian
- Department of Chemical Engineering University of Mississippi University Mississippi USA
- Center for Graphene Research and Innovation University of Mississippi University Mississippi USA
| |
Collapse
|
3
|
Liu J, Shen Y, Duan K, He X, Wang R, Chen Y, Li R, Sun J, Qiu X, Chen T, Wang J, Wang H. Novel biomimetic sandwich-structured electrospun cardiac patches with moderate adhesiveness and excellent electrical conductivity. J Mech Behav Biomed Mater 2024; 163:106828. [PMID: 39647339 DOI: 10.1016/j.jmbbm.2024.106828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
Clinical cardiac patches exhibit unsatisfied biocompatibility, low adhesion, and inadequate compliance and suboptimal mechanical properties for cardiac disorders repair. To address these challenges, herein we have innovatively proposed a biomimetic nanofiber electrospun membrane with a sandwich structure strategy. The composite patch comprises a stretchable polyurethane (PU) as basic material, then infiltrated with biocompatible silk fibroin methacryloyl (Silk-MA) as the middle layer via electrospinning and finally covered with Bio-ILs (chemically modified biocompatible ionic liquids) to impart electrical conductivity. Results indicated that the incorporation of Bio-ILs significantly enhances the conductivity reaching 2877 mS/m; particularly due to the positive charges of Bio-ILs, the composite film exhibits mild adhesive properties, inducing minimal damage to the substrate tissue. Furthermore, the basic PU of bilayer nanofiber membrane increased the film's stretching strain to approximately 250%, the Silk-MA hydrogel coating changed the film from hydrophobic to hydrophilic, creating a favorable and biocompatible microenvironment. Finally, in vitro experiments on cardiomyocytes confirmed that the material exhibits low cytotoxicity and excellent biocompatibility. Overall, the biomimetic sandwich electrospun membrane could restore electrical conduction and synchronized contraction function, providing a promising strategy for the treatment of cardiac tissue engineering.
Collapse
Affiliation(s)
- Jing Liu
- The Second Rehabilitation Hospital of Shanghai, China; Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yinyang Shen
- Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kaikai Duan
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiangming He
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruoyu Wang
- Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yeping Chen
- The Second Rehabilitation Hospital of Shanghai, China
| | - Ruoyu Li
- Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jialu Sun
- The Second Rehabilitation Hospital of Shanghai, China
| | - Xiaoyi Qiu
- The Second Rehabilitation Hospital of Shanghai, China
| | - Tao Chen
- Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jie Wang
- The Second Rehabilitation Hospital of Shanghai, China.
| | - Hui Wang
- The Second Rehabilitation Hospital of Shanghai, China; Engineering Research Center of Intelligent Rehabilitation for Traditional Chinese Medicine, Ministry of Education, School of Rehabilitation Science, Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
4
|
Xu Y, Yu Y, Guo Z. Hydrogels in cardiac tissue engineering: application and challenges. Mol Cell Biochem 2024:10.1007/s11010-024-05145-3. [PMID: 39495368 DOI: 10.1007/s11010-024-05145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Cardiovascular disease remains the leading cause of global mortality. Current stem cell therapy and heart transplant therapy have limited long-term stability in cardiac function. Cardiac tissue engineering may be one of the key methods for regenerating damaged myocardial tissue. As an ideal scaffold material, hydrogel has become a viable tissue engineering therapy for the heart. Hydrogel can not only provide mechanical support for infarcted myocardium but also serve as a carrier for various drugs, bioactive factors, and cells to increase myocardial contractility and improve the cell microenvironment in the infarcted area, thereby improving cardiac function. This paper reviews the applications of hydrogels and biomedical mechanisms in cardiac tissue engineering and discusses the challenge of clinical transformation of hydrogel in cardiac tissue engineering, providing new strategies for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yaping Xu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Yuexin Yu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China.
| |
Collapse
|
5
|
Casella A, Lowen J, Griffin KH, Shimamoto N, Ramos-Rodriguez DH, Panitch A, Leach JK. Conductive Microgel Annealed Scaffolds Enhance Myogenic Potential of Myoblastic Cells. Adv Healthc Mater 2024; 13:e2302500. [PMID: 38069833 DOI: 10.1002/adhm.202302500] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/15/2023] [Indexed: 12/19/2023]
Abstract
Conductive biomaterials may capture native or exogenous bioelectric signaling, but incorporation of conductive moieties is limited by cytotoxicity, poor injectability, or insufficient stimulation. Microgel annealed scaffolds are promising as hydrogel-based materials due to their inherent void space that facilitates cell migration and proliferation better than nanoporous bulk hydrogels. Conductive microgels are generated from poly(ethylene) glycol (PEG and poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT: PSS) to explore the interplay of void volume and conductivity on myogenic differentiation. PEDOT: PSS increases microgel conductivity two-fold while maintaining stiffness, annealing strength, and viability of associated myoblastic cells. C2C12 myoblasts exhibit increases in the late-stage differentiation marker myosin heavy chain as a function of both porosity and conductivity. Myogenin, an earlier marker, is influenced only by porosity. Human skeletal muscle-derived cells exhibit increased Myod1, insulin like growth factor-1, and insulin-like growth factor binding protein 2 at earlier time points on conductive microgel scaffolds compared to non-conductive scaffolds. They also secrete more vascular endothelial growth factor at early time points and express factors that led to macrophage polarization patterns observe during muscle repair. These data indicate that conductivity aids myogenic differentiation of myogenic cell lines and primary cells, motivating the need for future translational studies to promote muscle repair.
Collapse
Affiliation(s)
- Alena Casella
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
| | - Jeremy Lowen
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
| | - Katherine H Griffin
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
- School of Veterinary Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Nathan Shimamoto
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
| | - David H Ramos-Rodriguez
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Biomedical Engineering, Emory University, Atlanta, GA, 30322, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, 95817, USA
| |
Collapse
|
6
|
Roy A, Zenker S, Jain S, Afshari R, Oz Y, Zheng Y, Annabi N. A Highly Stretchable, Conductive, and Transparent Bioadhesive Hydrogel as a Flexible Sensor for Enhanced Real-Time Human Health Monitoring. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404225. [PMID: 38970527 PMCID: PMC11407428 DOI: 10.1002/adma.202404225] [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: 03/22/2024] [Revised: 06/05/2024] [Indexed: 07/08/2024]
Abstract
Real-time continuous monitoring of non-cognitive markers is crucial for the early detection and management of chronic conditions. Current diagnostic methods are often invasive and not suitable for at-home monitoring. An elastic, adhesive, and biodegradable hydrogel-based wearable sensor with superior accuracy and durability for monitoring real-time human health is developed. Employing a supramolecular engineering strategy, a pseudo-slide-ring hydrogel is synthesized by combining polyacrylamide (pAAm), β-cyclodextrin (β-CD), and poly 2-(acryloyloxy)ethyltrimethylammonium chloride (AETAc) bio ionic liquid (Bio-IL). This novel approach decouples conflicting mechano-chemical effects arising from different molecular building blocks and provides a balance of mechanical toughness (1.1 × 106 Jm-3), flexibility, conductivity (≈0.29 S m-1), and tissue adhesion (≈27 kPa), along with rapid self-healing and remarkable stretchability (≈3000%). Unlike traditional hydrogels, the one-pot synthesis avoids chemical crosslinkers and metallic nanofillers, reducing cytotoxicity. While the pAAm provides mechanical strength, the formation of the pseudo-slide-ring structure ensures high stretchability and flexibility. Combining pAAm with β-CD and pAETAc enhances biocompatibility and biodegradability, as confirmed by in vitro and in vivo studies. The hydrogel also offers transparency, passive-cooling, ultraviolet (UV)-shielding, and 3D printability, enhancing its practicality for everyday use. The engineered sensor demonstratesimproved efficiency, stability, and sensitivity in motion/haptic sensing, advancing real-time human healthcare monitoring.
Collapse
Affiliation(s)
- Arpita Roy
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Shea Zenker
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Saumya Jain
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ronak Afshari
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yavuz Oz
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuting Zheng
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
7
|
Carvalho T, Bártolo R, Correia A, Vilela C, Wang S, Santos HA, Freire CSR. Implantable Patch of Oxidized Nanofibrillated Cellulose and Lysozyme Amyloid Nanofibrils for the Regeneration of Infarcted Myocardium Tissue and Local Delivery of RNA-Loaded Nanoparticles. Macromol Rapid Commun 2024; 45:e2400129. [PMID: 38778746 DOI: 10.1002/marc.202400129] [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: 03/05/2024] [Revised: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Biopolymeric implantable patches are popular scaffolds for myocardial regeneration applications. Besides being biocompatible, they can be tailored to have required properties and functionalities for this application. Recently, fibrillar biobased nanostructures prove to be valuable in the development of functional biomaterials for tissue regeneration applications. Here, periodate-oxidized nanofibrillated cellulose (OxNFC) is blended with lysozyme amyloid nanofibrils (LNFs) to prepare a self-crosslinkable patch for myocardial implantation. The OxNFC:LNFs patch shows superior wet mechanical properties (60 MPa for Young's modulus and 1.5 MPa for tensile stress at tensile strength), antioxidant activity (70% scavenging activity under 24 h), and bioresorbability ratio (80% under 91 days), when compared to the patches composed solely of NFC or OxNFC. These improvements are achieved while preserving the morphology, required thermal stability for sterilization, and biocompatibility toward rat cardiomyoblast cells. Additionally, both OxNFC and OxNFC:LNFs patches reveal the ability to act as efficient vehicles to deliver spermine modified acetalated dextran nanoparticles, loaded with small interfering RNA, with 80% of delivery after 5 days. This study highlights the value of simply blending OxNFC and LNFs, synergistically combining their key properties and functionalities, resulting in a biopolymeric patch that comprises valuable characteristics for myocardial regeneration applications.
Collapse
Affiliation(s)
- Tiago Carvalho
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Raquel Bártolo
- Department of Biomaterials and Biomedical Technology, PRECISION - Personalized medicine Research Institute, University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Carla Vilela
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomaterials and Biomedical Technology, PRECISION - Personalized medicine Research Institute, University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Carmen S R Freire
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
8
|
Yu C, Qiu Y, Yao F, Wang C, Li J. Chemically Programmed Hydrogels for Spatiotemporal Modulation of the Cardiac Pathological Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404264. [PMID: 38830198 DOI: 10.1002/adma.202404264] [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: 03/24/2024] [Revised: 05/20/2024] [Indexed: 06/05/2024]
Abstract
After myocardial infarction (MI), sustained ischemic events induce pathological microenvironments characterized by ischemia-hypoxia, oxidative stress, inflammatory responses, matrix remodeling, and fibrous scarring. Conventional clinical therapies lack spatially targeted and temporally responsive modulation of the infarct microenvironment, leading to limited myocardial repair. Engineered hydrogels have a chemically programmed toolbox for minimally invasive localization of the pathological microenvironment and personalized responsive modulation over different pathological periods. Chemically programmed strategies for crosslinking interactions, interfacial binding, and topological microstructures in hydrogels enable minimally invasive implantation and in situ integration tailored to the myocardium. This enhances substance exchange and signal interactions within the infarcted microenvironment. Programmed responsive polymer networks, intelligent micro/nanoplatforms, and biological therapeutic cues contribute to the formation of microenvironment-modulated hydrogels with precise targeting, spatiotemporal control, and on-demand feedback. Therefore, this review summarizes the features of the MI microenvironment and chemically programmed schemes for hydrogels to conform, integrate, and modulate the cardiac pathological microenvironment. Chemically programmed strategies for oxygen-generating, antioxidant, anti-inflammatory, provascular, and electrointegrated hydrogels to stimulate iterative and translational cardiac tissue engineering are discussed.
Collapse
Affiliation(s)
- Chaojie Yu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Yuwei Qiu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Fanglian Yao
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Changyong Wang
- Tissue Engineering Research Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Junjie Li
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| |
Collapse
|
9
|
Liu T, Hao Y, Zhang Z, Zhou H, Peng S, Zhang D, Li K, Chen Y, Chen M. Advanced Cardiac Patches for the Treatment of Myocardial Infarction. Circulation 2024; 149:2002-2020. [PMID: 38885303 PMCID: PMC11191561 DOI: 10.1161/circulationaha.123.067097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Myocardial infarction is a cardiovascular disease characterized by a high incidence rate and mortality. It leads to various cardiac pathophysiological changes, including ischemia/reperfusion injury, inflammation, fibrosis, and ventricular remodeling, which ultimately result in heart failure and pose a significant threat to global health. Although clinical reperfusion therapies and conventional pharmacological interventions improve emergency survival rates and short-term prognoses, they are still limited in providing long-lasting improvements in cardiac function or reversing pathological progression. Recently, cardiac patches have gained considerable attention as a promising therapy for myocardial infarction. These patches consist of scaffolds or loaded therapeutic agents that provide mechanical reinforcement, synchronous electrical conduction, and localized delivery within the infarct zone to promote cardiac restoration. This review elucidates the pathophysiological progression from myocardial infarction to heart failure, highlighting therapeutic targets and various cardiac patches. The review considers the primary scaffold materials, including synthetic, natural, and conductive materials, and the prevalent fabrication techniques and optimal properties of the patch, as well as advanced delivery strategies. Last, the current limitations and prospects of cardiac patch research are considered, with the goal of shedding light on innovative products poised for clinical application.
Collapse
Affiliation(s)
- Tailuo Liu
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Ying Hao
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Zixuan Zhang
- West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, PR China (Z.Z.)
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Shiqin Peng
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Dingyi Zhang
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Mao Chen
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
10
|
Chen C, Tang Q, Wu L, Gu G, Huang X, Chen K, Li Z, Wang J, Qu G, Jiang Y, Liu Y, Li S, Huang J, Jia X, Zhu T, Zhao Y, Zhang Q, Ren J, Wu X. Hybrid Double-Sided Tape with Asymmetrical Adhesion and Burst Pressure Tolerance for Abdominal Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30430-30442. [PMID: 38814614 DOI: 10.1021/acsami.4c05400] [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: 05/31/2024]
Abstract
Patients with open abdominal (OA) wounds have a mortality risk of up to 30%, and the resulting disabilities would have profound effects on patients. Here, we present a novel double-sided adhesive tape developed for the management of OA wounds. The tape features an asymmetrical structure and employs an acellular dermal matrix (ADM) with asymmetric wettability as a scaffold. It is constructed by integrating a tissue-adhesive hydrogel composed of polydopamine (pDA), quaternary ammonium chitosan (QCS), and acrylic acid cross-linking onto the bottom side of the ADM. Following surface modification with pDA, the ADM would exhibit characteristics resistant to bacterial adhesion. Furthermore, the presence of a developed hydrogel ensures that the tape not only possesses tissue adhesiveness and noninvasive peelability but also effectively mitigates damage caused by oxidative stress. Besides, the ADM inherits the strength of the skin, imparting high burst pressure tolerance to the tape. Based on these remarkable attributes, we demonstrate that this double-sided (D-S) tape facilitates the repair of OA wounds, mitigates damage to exposed intestinal tubes, and reduces the risk of intestinal fistulae and complications. Additionally, the D-S tape is equally applicable to treating other abdominal injuries, such as gastric perforations. It effectively seals the perforation, promotes injury repair, and prevents the formation of postoperative adhesions. These notable features indicate that the presented double-sided tape holds significant potential value in the biomedical field.
Collapse
Affiliation(s)
- Canwen Chen
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Qinqing Tang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Lei Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing 210002, China
| | - Guosheng Gu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
- Department of General Surgery, Anhui No.2 Provincial Peoples' Hospital, Anhui 230041, P. R. China
| | - Xinxin Huang
- Key Laboratory of High Performance Polymer Material and Technology of MOE, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Kang Chen
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Ze Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Jiajie Wang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Guiwen Qu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Yungang Jiang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Ye Liu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Sicheng Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Jinjian Huang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Xudong Jia
- Key Laboratory of High Performance Polymer Material and Technology of MOE, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Tangsong Zhu
- Key Laboratory of High Performance Polymer Material and Technology of MOE, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yun Zhao
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, P. R. China
| | - Qiuhong Zhang
- Key Laboratory of High Performance Polymer Material and Technology of MOE, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, P. R. China
| |
Collapse
|
11
|
Ma P, Liang W, Huang R, Zheng B, Feng K, He W, Huang Z, Shen H, Wang H, Wu D. Super-Structured Wet-Adhesive Hydrogel with Ultralow Swelling, Ultrahigh Burst Pressure Tolerance, and Anti-Postoperative Adhesion Properties for Tissue Adhesion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305400. [PMID: 38010313 DOI: 10.1002/adma.202305400] [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: 06/06/2023] [Revised: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Wet-adhesive hydrogels have been developed as an attractive strategy for tissue repair. However, achieving simultaneously low swelling and high burst pressure tolerance of wet-adhesive hydrogels is crucial for in vivo application which remains challenges. Herein, a novel super-structured porous hydrogel (denoted as PVA/PAAc-N+ ) is designed via facile moisture-induced phase separation-solvent exchange process for obtaining porous polyvinyl alcohol (PVA) hydrogel as dissipative layer and in situ photocuring technology for entangling quaternary ammonium-functionalized poly(acrylic acid)-based wet-adhesive layer (PAAc-N+ ) with the porous surface of PVA layer. Benefitting from the ionic crosslinking between quaternary ammonium ions and carboxylate ions in PAAc-N+ wet-adhesive layer as well as the high crystallinity induced by abundant hydrogen bonds of PVA layer, the hydrogel has unique ultralow swelling property (0.29) without sacrificing adhesion strength (63.1 kPa). The porous structure of PVA facilitates the mechanical interlock at the interface between PAAc-N+ wet-adhesive layer and tough PVA dissipative layer, leading to the ultrahigh burst pressure tolerance up to 493 mm Hg and effective repair for porcine heart rupture; the PVA layer surface of PVA/PAAc-N+ hydrogel can prevent postoperative adhesion. By integrating ultralow swelling, ultrahigh burst pressure tolerance, and anti-postoperative adhesion properties, PVA/PAAc-N+ hydrogel shows an appealing application prospect for tissue repair.
Collapse
Affiliation(s)
- Pengwei Ma
- PCFM Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, P. R. China
| | - Weiwen Liang
- Department of General Surgery (Colorectal Surgery), Guangdong Institute of Gastroenterology, Biomedical Innovation Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Rongkang Huang
- Department of General Surgery (Colorectal Surgery), Guangdong Institute of Gastroenterology, Biomedical Innovation Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Bingna Zheng
- Center of Accurate Diagnosis, Treatment and Transformation of Bone and Joint Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, P. R. China
| | - Kangni Feng
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Wenyi He
- PCFM Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, P. R. China
| | - Zeping Huang
- Department of General Surgery (Colorectal Surgery), Guangdong Institute of Gastroenterology, Biomedical Innovation Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Huiyong Shen
- Center of Accurate Diagnosis, Treatment and Transformation of Bone and Joint Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, P. R. China
| | - Hui Wang
- Department of General Surgery (Colorectal Surgery), Guangdong Institute of Gastroenterology, Biomedical Innovation Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Dingcai Wu
- PCFM Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, P. R. China
- Center of Accurate Diagnosis, Treatment and Transformation of Bone and Joint Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, P. R. China
| |
Collapse
|
12
|
Wu J, Yun Z, Song W, Yu T, Xue W, Liu Q, Sun X. Highly oriented hydrogels for tissue regeneration: design strategies, cellular mechanisms, and biomedical applications. Theranostics 2024; 14:1982-2035. [PMID: 38505623 PMCID: PMC10945336 DOI: 10.7150/thno.89493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/19/2024] [Indexed: 03/21/2024] Open
Abstract
Many human tissues exhibit a highly oriented architecture that confers them with distinct mechanical properties, enabling adaptation to diverse and challenging environments. Hydrogels, with their water-rich "soft and wet" structure, have emerged as promising biomimetic materials in tissue engineering for repairing and replacing damaged tissues and organs. Highly oriented hydrogels can especially emulate the structural orientation found in human tissue, exhibiting unique physiological functions and properties absent in traditional homogeneous isotropic hydrogels. The design and preparation of highly oriented hydrogels involve strategies like including hydrogels with highly oriented nanofillers, polymer-chain networks, void channels, and microfabricated structures. Understanding the specific mechanism of action of how these highly oriented hydrogels affect cell behavior and their biological applications for repairing highly oriented tissues such as the cornea, skin, skeletal muscle, tendon, ligament, cartilage, bone, blood vessels, heart, etc., requires further exploration and generalization. Therefore, this review aims to fill that gap by focusing on the design strategy of highly oriented hydrogels and their application in the field of tissue engineering. Furthermore, we provide a detailed discussion on the application of highly oriented hydrogels in various tissues and organs and the mechanisms through which highly oriented structures influence cell behavior.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130023, China
| | - Tao Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
13
|
Lee M, Kim YS, Park J, Choe G, Lee S, Kang BG, Jun JH, Shin Y, Kim M, Ahn Y, Lee JY. A paintable and adhesive hydrogel cardiac patch with sustained release of ANGPTL4 for infarcted heart repair. Bioact Mater 2024; 31:395-407. [PMID: 37680586 PMCID: PMC10481188 DOI: 10.1016/j.bioactmat.2023.08.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/05/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The infarcted heart undergoes irreversible pathological remodeling after reperfusion involving left ventricle dilation and excessive inflammatory reactions in the infarcted heart, frequently leading to fatal functional damage. Extensive attempts have been made to attenuate pathological remodeling in infarcted hearts using cardiac patches and anti-inflammatory drug delivery. In this study, we developed a paintable and adhesive hydrogel patch using dextran-aldehyde (dex-ald) and gelatin, incorporating the anti-inflammatory protein, ANGPTL4, into the hydrogel for sustained release directly to the infarcted heart to alleviate inflammation. We optimized the material composition, including polymer concentration and molecular weight, to achieve a paintable, adhesive hydrogel using 10% gelatin and 5% dex-ald, which displayed in-situ gel formation within 135 s, cardiac tissue-like modulus (40.5 kPa), suitable tissue adhesiveness (4.3 kPa), and excellent mechanical stability. ANGPTL4 was continuously released from the gelatin/dex-ald hydrogel without substantial burst release. The gelatin/dex-ald hydrogel could be conveniently painted onto the beating heart and degraded in vivo. Moreover, in vivo studies using animal models of acute myocardial infarction revealed that our hydrogel cardiac patch containing ANGPTL4 significantly improved heart tissue repair, evaluated by echocardiography and histological evaluation. The heart tissues treated with ANGPTL4-loaded hydrogel patches exhibited increased vascularization, reduced inflammatory macrophages, and structural maturation of cardiac cells. Our novel hydrogel system, which allows for facile paintability, appropriate tissue adhesiveness, and sustained release of anti-inflammatory drugs, will serve as an effective platform for the repair of various tissues, including heart, muscle, and cartilage.
Collapse
Affiliation(s)
- Mingyu Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Sanghun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
| | - Yoonmin Shin
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Minchul Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- 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, 61005, Republic of Korea
| |
Collapse
|
14
|
Zhang M, An H, Gu Z, Zhang YC, Wan T, Jiang HR, Zhang FS, Jiang BG, Han N, Wen YQ, Zhang PX. Multifunctional wet-adhesive chitosan/acrylic conduit for sutureless repair of peripheral nerve injuries. Int J Biol Macromol 2023; 253:126793. [PMID: 37709238 DOI: 10.1016/j.ijbiomac.2023.126793] [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: 05/25/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The incidence of peripheral nerve injury (PNI) is high worldwide, and a poor prognosis is common. Surgical closure and repair of the affected area are crucial to ensure the effective treatment of peripheral nerve injuries. Despite being the standard treatment approach, reliance on sutures to seal the severed nerve ends introduces several limitations and restrictions. This technique is intricate and time-consuming, and the application of threading and punctate sutures may lead to tissue damage and heightened tension concentrations, thus increasing the risk of fixation failure and local inflammation. This study aimed to develop easily implantable chitosan-based peripheral nerve repair conduits that combine acrylic acid and cleavable N-hydroxysuccinimide to reduce nerve damage during repair. In ex vivo tissue adhesion tests, the conduit achieved maximal interfacial toughness of 705 J m-2 ± 30 J m-2, allowing continuous bridging of the severed nerve ends. Adhesive repair significantly reduces local inflammation caused by conventional sutures, and the positive charge of chitosan disrupts the bacterial cell wall and reduces implant-related infections. This promises to open new avenues for sutureless nerve repair and reliable medical implants.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Heng An
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Zhen Gu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Yi-Chong Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Teng Wan
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Hao-Ran Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Feng-Shi Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Bao-Guo Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Na Han
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| | - Yong-Qiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Pei-Xun Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration, Peking University, National Center for Trauma Medicine, Beijing 100044, China.
| |
Collapse
|
15
|
Fang Z, Lv B, Zhan J, Xing X, Ding C, Liu J, Wang L, Zou X, Qiu X. Flexible Conductive Decellularized Fish Skin Matrix as a Functional Scaffold for Myocardial Infarction Repair. Macromol Biosci 2023; 23:e2300207. [PMID: 37534715 DOI: 10.1002/mabi.202300207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Engineering cardiac patches are proven to be effective in myocardial infarction (MI) repair, but it is still a tricky problem in tissue engineering to construct a scaffold with good biocompatibility, suitable mechanical properties, and solid structure. Herein, decellularized fish skin matrix is utilized with good biocompatibility to prepare a flexible conductive cardiac patch through polymerization of polydopamine (PDA) and polypyrrole (PPy). Compared with single modification, the double modification strategy facilitated the efficiency of pyrrole polymerization, so that the patch conductivity is improved. According to the results of experiments in vivo and in vitro, the scaffold can promote the maturation and functionalization of cardiomyocytes (CMs). It can also reduce the inflammatory response, increase local microcirculation, and reconstruct the conductive microenvironment in infarcted myocardia, thus improving the cardiac function of MI rats. In addition, the excellent flexibility of the scaffold, which enables it to be implanted in vivo through "folding-delivering-re-stretehing" pathway, provides the possibility of microoperation under endoscope, which avoids the secondary damage to myocardium by traditional thoracotomy for implantation surgery.
Collapse
Affiliation(s)
- Zhanhong Fang
- The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, 528244, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Bingyang Lv
- The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, 528244, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiamian Zhan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xianglong Xing
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chengbin Ding
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianing Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoming Zou
- The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, 528244, China
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
16
|
Xia M, Wu M, Li Y, Liu Y, Jia G, Lou Y, Ma J, Gao Q, Xie M, Chen Y, He Y, Li H, Li W. Varying mechanical forces drive sensory epithelium formation. SCIENCE ADVANCES 2023; 9:eadf2664. [PMID: 37922362 PMCID: PMC10624343 DOI: 10.1126/sciadv.adf2664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/05/2023] [Indexed: 11/05/2023]
Abstract
The mechanical cues of the external microenvironment have been recognized as essential clues driving cell behavior. Although intracellular signals modulating cell fate during sensory epithelium development is well understood, the driving force of sensory epithelium formation remains elusive. Here, we manufactured a hybrid hydrogel with tunable mechanical properties for the cochlear organoids culture and revealed that the extracellular matrix (ECM) drives sensory epithelium formation through shifting stiffness in a stage-dependent pattern. As the driving force, moderate ECM stiffness activated the expansion of cochlear progenitor cell (CPC)-derived epithelial organoids by modulating the integrin α3 (ITGA3)/F-actin cytoskeleton/YAP signaling. Higher stiffness induced the transition of CPCs into sensory hair cells (HCs) through increasing the intracellular Ca2+ signaling mediated by PIEZO2 and then activating KLF2 to accomplish the cell specification . Our results identify the molecular mechanism of sensory epithelium formation guided by ECM mechanical force and contribute to developing therapeutic approaches for HC regeneration.
Collapse
Affiliation(s)
- Mingyu Xia
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Mingxuan Wu
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanrong Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yaoqian Liu
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Gaogan Jia
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yiyun Lou
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jiaoyao Ma
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, 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
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, 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
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huawei Li
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Centre of Cochlear Implant, Shanghai 200031, China
| | - Wenyan Li
- ENT institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Liu S, Yu Q, Guo R, Chen K, Xia J, Guo Z, He L, Wu Q, Liu L, Li Y, Zhang B, Lu L, Sheng X, Zhu J, Zhao L, Qi H, Liu K, Yin L. A Biodegradable, Adhesive, and Stretchable Hydrogel and Potential Applications for Allergic Rhinitis and Epistaxis. Adv Healthc Mater 2023; 12:e2302059. [PMID: 37610041 DOI: 10.1002/adhm.202302059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Indexed: 08/24/2023]
Abstract
Bioadhesive hydrogels have attracted considerable attention as innovative materials in medical interventions and human-machine interface engineering. Despite significant advances in their application, it remains critical to develop adhesive hydrogels that meet the requirements for biocompatibility, biodegradability, long-term strong adhesion, and efficient drug delivery vehicles in moist conditions. A biocompatible, biodegradable, soft, and stretchable hydrogel made from a combination of a biopolymer (unmodified natural gelatin) and stretchable biodegradable poly(ethylene glycol) diacrylate is proposed to achieve durable and tough adhesion and explore its use for convenient and effective intranasal hemostasis and drug administration. Desirable hemostasis efficacy and enhanced therapeutic outcomes for allergic rhinitis are accomplished. Biodegradation enables the spontaneous removal of materials without causing secondary damage and minimizes medical waste. Preliminary trials on human subjects provide an essential foundation for practical applications. This work elucidates material strategies for biodegradable adhesive hydrogels, which are critical to achieving robust material interfaces and advanced drug delivery platforms for novel clinical treatments.
Collapse
Affiliation(s)
- Shengnan Liu
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Qianru Yu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Rui Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Kuntao Chen
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Jiao Xia
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhenhu Guo
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Lu He
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Qian Wu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lan Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yunxuan Li
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Bozhen Zhang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Lin Lu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Jiahua Zhu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lingyun Zhao
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Hui Qi
- Laboratory of Musculoskeletal Regenerative Medicine, Beijing Institute of Traumatology and Orthopaedics, Beijing, 100035, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Beijing Clinical Research Institute, Beijing, 100050, China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
18
|
Corral-Nájera K, Chauhan G, Serna-Saldívar SO, Martínez-Chapa SO, Aeinehvand MM. Polymeric and biological membranes for organ-on-a-chip devices. MICROSYSTEMS & NANOENGINEERING 2023; 9:107. [PMID: 37649779 PMCID: PMC10462672 DOI: 10.1038/s41378-023-00579-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/18/2023] [Accepted: 06/20/2023] [Indexed: 09/01/2023]
Abstract
Membranes are fundamental elements within organ-on-a-chip (OOC) platforms, as they provide adherent cells with support, allow nutrients (and other relevant molecules) to permeate/exchange through membrane pores, and enable the delivery of mechanical or chemical stimuli. Through OOC platforms, physiological processes can be studied in vitro, whereas OOC membranes broaden knowledge of how mechanical and chemical cues affect cells and organs. OOCs with membranes are in vitro microfluidic models that are used to replace animal testing for various applications, such as drug discovery and disease modeling. In this review, the relevance of OOCs with membranes is discussed as well as their scaffold and actuation roles, properties (physical and material), and fabrication methods in different organ models. The purpose was to aid readers with membrane selection for the development of OOCs with specific applications in the fields of mechanistic, pathological, and drug testing studies. Mechanical stimulation from liquid flow and cyclic strain, as well as their effects on the cell's increased physiological relevance (IPR), are described in the first section. The review also contains methods to fabricate synthetic and ECM (extracellular matrix) protein membranes, their characteristics (e.g., thickness and porosity, which can be adjusted depending on the application, as shown in the graphical abstract), and the biological materials used for their coatings. The discussion section joins and describes the roles of membranes for different research purposes and their advantages and challenges.
Collapse
Affiliation(s)
- Kendra Corral-Nájera
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Gaurav Chauhan
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Sergio O. Serna-Saldívar
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Sergio O. Martínez-Chapa
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Mohammad Mahdi Aeinehvand
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| |
Collapse
|
19
|
Casella A, Lowen J, Shimamoto N, Griffin KH, Filler AC, Panitch A, Leach JK. Conductive microgel annealed scaffolds enhance myogenic potential of myoblastic cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551533. [PMID: 37577583 PMCID: PMC10418230 DOI: 10.1101/2023.08.01.551533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Bioelectricity is an understudied phenomenon to guide tissue homeostasis and regeneration. Conductive biomaterials may capture native or exogenous bioelectric signaling, but incorporation of conductive moieties is limited by cytotoxicity, poor injectability, or insufficient stimulation. Microgel annealed scaffolds are promising as hydrogel-based materials due to their inherent void space that facilitates cell migration and proliferation better than nanoporous bulk hydrogels. We generated conductive microgels from poly(ethylene) glycol and poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS) to explore the interplay of void volume and conductivity on myogenic differentiation. PEDOT:PSS increased microgel conductivity over 2-fold while maintaining stiffness, annealing strength, and viability of associated myoblastic cells. C2C12 myoblasts exhibited increases in the late-stage differentiation marker myosin heavy chain as a function of both porosity and conductivity. Myogenin, an earlier marker, was influenced only by porosity. Human skeletal muscle derived cells exhibited increased Myod1 , IGF-1, and IGFBP-2 at earlier timepoints on conductive microgel scaffolds compared to non-conductive scaffolds. They also secreted higher levels of VEGF at early timepoints and expressed factors that led to macrophage polarization patterns observed during muscle repair. These data indicate that conductivity aids myogenic differentiation of myogenic cell lines and primary cells, motivating the need for future translational studies to promote muscle repair.
Collapse
|
20
|
Zhang M, An H, Gu Z, Huang Z, Zhang F, Jiang BG, Wen Y, Zhang P. Mimosa-Inspired Stimuli-Responsive Curling Bioadhesive Tape Promotes Peripheral Nerve Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2212015. [PMID: 37205796 DOI: 10.1002/adma.202212015] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/14/2023] [Indexed: 05/21/2023]
Abstract
Trauma often results in peripheral nerve injuries (PNIs). These injuries are particularly challenging therapeutically because of variable nerve diameters, slow axonal regeneration, infection of severed ends, fragility of the nerve tissue, and the intricacy of surgical intervention. Surgical suturing is likely to cause additional damage to peripheral nerves. Therefore, an ideal nerve scaffold should possess good biocompatibility, diameter adaptability, and a stable biological interface for seamless biointegration with tissues. Inspired by the curl of Mimosa pudica, this study aimed to design and develop a diameter-adaptable, suture-free, stimulated curling bioadhesive tape (SCT) hydrogel for repairing PNI. The hydrogel is fabricated from chitosan and acrylic acid-N-hydroxysuccinimide lipid via gradient crosslinking using glutaraldehyde. It closely matches the nerves of different individuals and regions, thereby providing a bionic scaffold for axonal regeneration. In addition, this hydrogel rapidly absorbs tissue fluid from the nerve surface achieving durable wet-interface adhesion. Furthermore, the chitosan-based SCT hydrogel loaded with insulin-like growth factor-I effectively promotes peripheral nerve regeneration with excellent bioactivity. This procedure for peripheral nerve injury repair using the SCT hydrogel is simple and reduces the difficulty and duration of surgery, thereby advancing adaptive biointerfaces and reliable materials for nerve repair.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Heng An
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhen Gu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhe Huang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Fengshi Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Bao-Guo Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Peixun Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| |
Collapse
|
21
|
Basara G, Bahcecioglu G, Ren X, Zorlutuna P. An Experimental and Numerical Investigation of Cardiac Tissue-Patch Interrelation. J Biomech Eng 2023; 145:081004. [PMID: 37337466 PMCID: PMC10321148 DOI: 10.1115/1.4062736] [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: 11/21/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Tissue engineered cardiac patches have great potential as a regenerative therapy for myocardial infarction. Yet, the mutual interaction of cardiac patches with healthy tissue has not been completely understood. Here, we investigated the impact of acellular and cellular patches on a beating two-dimensional (2D) cardiac cell layer, and the effect of the beating of this layer on the cells encapsulated in the patch. We cultured human-induced pluripotent stem cell-derived cardiomyocytes (iCMs) on a coverslip and placed gelatin methacryloyl hydrogel alone or with encapsulated iCMs to create acellular and cellular patches, respectively. When the acellular patch was placed on the cardiac cell layer, the beating characteristics and Ca+2 handling properties reduced, whereas placing the cellular patch restored these characteristics. To better understand the effects of the cyclic contraction and relaxation induced by the beating cardiac cell layer on the patch placed on top of it, a simulation model was developed, and the calculated strain values were in agreement with the values measured experimentally. Moreover, this dynamic culture induced by the beating 2D iCM layer on the iCMs encapsulated in the cellular patch improved their beating velocity and frequency. Additionally, the encapsulated iCMs were observed to be coupled with the underlying beating 2D iCM layer. Overall, this study provides a detailed investigation on the mutual relationship of acellular/cellular patches with the beating 2D iCM layer, understanding of which would be valuable for developing more advanced cardiac patches.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, 225 Multidisciplinary Research Building, Notre Dame, IN 46556
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, 108B Multidisciplinary Research Building, Notre Dame, IN 46556
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556; Department of Chemical and Biomolecular Engineering, University of Notre Dame, 143 Multidisciplinary Research Building, Notre Dame, IN 46556
| |
Collapse
|
22
|
Saghebasl S, Nobakht A, Saghebasl H, Hayati S, Naturi O, Rahbarghazi R. Sandwich-like electro-conductive polyurethane-based gelatin/soybean oil nanofibrous scaffolds with a targeted release of simvastatin for cardiac tissue engineering. J Biol Eng 2023; 17:42. [PMID: 37415188 DOI: 10.1186/s13036-023-00364-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Cardiac tissue engineering (CTE) is a promising way for the restoration of injured cardiac tissue in the healthcare system. The development of biodegradable scaffolds with appropriate chemical, electrical, mechanical, and biological properties is an unmet need for the success of CTE. Electrospinning is a versatile technique that has shown potential applications in CTE. Herein, four different types of multifunctional scaffolds, including synthetic-based poly (glycerol sebacate)-polyurethane (PGU), PGU-Soy scaffold, and a series of trilayer scaffolds containing two outer layers of PGU-Soy and a middle (inner) layer of gelatin (G) as a natural and biodegradable macromolecule without simvastatin (S) and with simvastatin (GS), an anti-inflammatory agent, were fabricated in the sandwich-like structure using electrospinning technique. This approach offers a combination of the advantages of both synthetic and natural polymers to enhance the bioactivity and the cell-to-cell and cell-to-matrix intercommunication. An in vitro drug release analysis was performed after the incorporation of soybean oil (Soy) and G. Soy is used as a semiconducting material was introduced to improve the electrical conductivity of nanofibrous scaffolds. The physicochemical properties, contact angle, and biodegradability of the electrospun scaffolds were also assessed. Moreover, the blood compatibility of nanofibrous scaffolds was studied through activated partial thromboplastin time (APTT), prothrombin time (PT), and hemolytic assay. The results showed that all scaffolds exhibited defect-free morphologies with mean fiber diameters in the range of 361 ± 109 to 417 ± 167 nm. A delay in blood clotting was observed, demonstrating the anticoagulant nature of nanofibrous scaffolds. Furthermore, rat cardiomyoblast cell lines (H9C2) were cultured on scaffolds for 7 days, and the morphology and cell arrangement were monitored. Data indicated an appropriate cytocompatibility. Of note, in the PGU-Soy/GS nanofibrous scaffold, a high survival rate was indicated compared to other groups. Our findings exhibited that the simvastatin-loaded polymeric system had positive effects on cardiomyoblasts attachment and growth and could be utilized as a drug release carrier in the field of CTE.
Collapse
Affiliation(s)
- Solmaz Saghebasl
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Nobakht
- Research Center of Biosciences & Biotechnology (RCBB), University of Tabriz, Tabriz, Iran
| | - Hesam Saghebasl
- Faculty of Medicine, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Sanya Hayati
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Zhao W, Hu C, Xu T. In vivo bioprinting: Broadening the therapeutic horizon for tissue injuries. Bioact Mater 2023; 25:201-222. [PMID: 36817820 PMCID: PMC9932583 DOI: 10.1016/j.bioactmat.2023.01.018] [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: 09/08/2022] [Revised: 01/06/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Tissue injury is a collective term for various disorders associated with organs and tissues induced by extrinsic or intrinsic factors, which significantly concerns human health. In vivo bioprinting, an emerging tissue engineering approach, allows for the direct deposition of bioink into the defect sites inside the patient's body, effectively addressing the challenges associated with the fabrication and implantation of irregularly shaped scaffolds and enabling the rapid on-site management of tissue injuries. This strategy complements operative therapy as well as pharmacotherapy, and broadens the therapeutic horizon for tissue injuries. The implementation of in vivo bioprinting requires targeted investigations in printing modalities, bioinks, and devices to accommodate the unique intracorporal microenvironment, as well as effective integrations with intraoperative procedures to facilitate its clinical application. In this review, we summarize the developments of in vivo bioprinting from three perspectives: modalities and bioinks, devices, and clinical integrations, and further discuss the current challenges and potential improvements in the future.
Collapse
Affiliation(s)
- Wenxiang Zhao
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory of Precision/Ultra-Precision Manufacturing Equipments and Control, Tsinghua University, Beijing, 100084, China
| | - Chuxiong Hu
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory of Precision/Ultra-Precision Manufacturing Equipments and Control, Tsinghua University, Beijing, 100084, China
| | - Tao Xu
- Center for Bio-intelligent Manufacturing and Living Matter Bioprinting, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, 518057, China
| |
Collapse
|
24
|
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
|
25
|
Song X, Zhang J, Shen S, Liu D, Zhang J, Yin W, Ye G, Wang L, Cai L, Hou H, Qiu X. Cardiac-Adaptive Conductive Hydrogel Patch Enabling Construction of Mechanical-Electrical Anisotropic Microenvironment for Heart Repair. RESEARCH (WASHINGTON, D.C.) 2023; 6:0161. [PMID: 37303598 PMCID: PMC10250027 DOI: 10.34133/research.0161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/14/2023] [Indexed: 06/13/2023]
Abstract
The biomimetic construction of a microstructural-mechanical-electrical anisotropic microenvironment adaptive to the native cardiac tissue is essential to repair myocardial infarction (MI). Inspired by the 3D anisotropic characteristic of the natural fish swim bladder (FSB), a novel flexible, anisotropic, and conductive hydrogel was developed for tissue-specific adaptation to the anisotropic structural, conductive, and mechanical features of the native cardiac extracellular matrix. The results revealed that the originally stiff, homogeneous FSB film was tailored to a highly flexible anisotropic hydrogel, enabling its potential as a functional engineered cardiac patch (ECP). In vitro and in vivo experiments demonstrated the enhanced electrophysiological activity, maturation, elongation, and orientation of cardiomyocytes (CMs), and marked MI repair performance with reduced CM apoptosis and myocardial fibrosis, thereby promoting cell retention, myogenesis, and vascularization, as well as improving electrical integration. Our findings offer a potential strategy for functional ECP and provides a novel strategy to bionically simulate the complex cardiac repair environment.
Collapse
Affiliation(s)
- Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital,
Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou 510630, China
| | - Si Shen
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Dan Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jie Zhang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wenming Yin
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Genlan Ye
- Central Laboratory, The Fifth Affiliated Hospital,
Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Liu Cai
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaozhong Qiu
- Central Laboratory, The Fifth Affiliated Hospital,
Southern Medical University, Guangzhou, Guangdong 510910, China
| |
Collapse
|
26
|
Deng C, Jin Q, Xu J, Fu W, He M, Xu L, Song Y, Wang W, Yi L, Chen Y, Gao T, Wang J, Lv Q, Yang Y, Zhang L, Xie M. Electrospun polymer fibers modified with FK506 for the long-term treatment of acute cardiac allograft rejection in a heart transplantation model. Biomater Sci 2023; 11:4032-4042. [PMID: 37129635 DOI: 10.1039/d3bm00374d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
FK506, a first-line immunosuppressant, is routinely administered orally and intravenously following heart transplantation. However, frequent administration can result in a substantial psychological burden to patients, resulting in non-adherence to medication. The purpose of our study is to overcome the disadvantages of systemic drug administration by developing a polymer-based delivery system that is tunable and biodegradable and that can release highly hydrophobic FK506 over extended periods to treat or prevent acute cardiac allograft rejection. Using an electrospinning method, long-acting microfibers were prepared, and FK506 appeared to be continuously released for up to 14 days based on the in vitro release profiles. After implanting the microfiber subcutaneously into the abdominals of transplanted rats, it was found that the infiltration of T cells and macrophages and the secretion of interleukin-2 (IL-2) and IL-1β were significantly reduced compared with those of the free FK506 groups. More importantly, the mean survival time (MST) of the PCL-FK506 group was significantly extended in comparison with that of untreated control recipients and free FK506 (MST of untreated control recipients, free FK506, and PCL-FK506 was 8, 26.1, and 37, respectively). In conclusion, we propose that this drug delivery approach would be suitable for developing long-lasting immunomodulatory agents that prolong cardiac graft survival safely and effectively.
Collapse
Affiliation(s)
- Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenpei Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengrong He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
27
|
Casella A, Panitch A, Leach JK. Electroconductive agarose hydrogels modulate mesenchymal stromal cell adhesion and spreading through protein adsorption. J Biomed Mater Res A 2023; 111:596-608. [PMID: 36680496 PMCID: PMC10023318 DOI: 10.1002/jbm.a.37503] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
Electrically conductive biomaterials direct cell behavior by capitalizing on the effect of bioelectricity in tissue homeostasis and healing. Many studies have leveraged conductive biomaterials to influence cells and improve tissue healing, even in the absence of external stimulation. However, most studies using electroactive materials neglect characterizing how the inclusion of conductive additives affects the material's mechanical properties, and the interplay between substrate electrical and mechanical properties on cell behavior is poorly understood. Furthermore, mechanisms dictating how electrically conductive materials affect cell behavior in the absence of external stimulation are not explicit. In this study, we developed a mechanically and electrically tunable conductive hydrogel using agarose and the conductive polymer PEDOT:PSS. Under certain conditions, we observed that the hydrogel physical and electrical properties were decoupled. We then seeded human mesenchymal stromal cells (MSCs) onto the hydrogels and observed enhanced adhesion and spreading of MSCs on conductive substrates, regardless of the hydrogel mechanical properties, and despite the gels having no cell-binding sites. To explain this observation, we measured protein interaction with the gels and found that charged proteins adsorbed significantly more to conductive hydrogels. These data demonstrate that conductivity promotes cell adhesion, likely by facilitating increased adsorption of proteins associated with cell binding, providing a better understanding of the mechanism of action of electrically conductive materials.
Collapse
Affiliation(s)
- Alena Casella
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817
| | - Alyssa Panitch
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817
| |
Collapse
|
28
|
Xu W, Zhang M, Du W, Ling G, Yuan Y, Zhang P. Engineering a naturally-derived wound dressing based on bio-ionic liquid conjugation. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.112055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
29
|
Gil-Cabrerizo P, Scaccheti I, Garbayo E, Blanco-Prieto MJ. Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023; 185:106439. [PMID: 37003408 DOI: 10.1016/j.ejps.2023.106439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ilaria Scaccheti
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| |
Collapse
|
30
|
Liu W, Zhao N, Yin Q, Zhao X, Guo K, Xian Y, Li S, Wang C, Zhu M, Du Y, Xu FJ, Wang C, Zhou J. Injectable Hydrogels Encapsulating Dual-Functional Au@Pt Core-Shell Nanoparticles Regulate Infarcted Microenvironments and Enhance the Therapeutic Efficacy of Stem Cells through Antioxidant and Electrical Integration. ACS NANO 2023; 17:2053-2066. [PMID: 36695873 PMCID: PMC9933615 DOI: 10.1021/acsnano.2c07436] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/20/2023] [Indexed: 06/17/2023]
Abstract
Injectable functional biomaterials have made significant progress in cardiac regenerative. In addition, how to adjust the abominable infarction microenvironment and introduce therapeutic stem cells to improve the healing effect has become a hotspot. Herein, injectable stem cell vector is prepared by combining natural alginate hydrogel and Au@Pt nanoparticles (Au@Pt/Alg hydrogel) to encapsulate brown adipose stem cells (BASCs). Au@Pt nanoparticles with both antioxidative and conductive properties could effectively eliminate reactive oxygen species, enhance the frequency of action potential release of cardiomyocytes, and further reduce the inflammatory factors of macrophage in vitro. The Au@Pt/Alg hydrogel enhances the antioxidant, differentiation, and paracrine capability of BASCs. The effect of BASCs loaded Au@Pt/Alg hydrogel is evaluated in a rat myocardial infarction (MI) model. The antioxidant, anti-inflammatory, and heart electrical integration are showed in the MI model. More interestingly, Au@Pt/Alg hydrogel can effectively maintain the paracrine efficiency and pro-angiogenesis effects of BASCs in the infarcted area. This study led us to recognize the great value of Au@Pt/Alg hydrogels for their ability to actively regulate the microenvironment and carry stem cells for MI treatment.
Collapse
Affiliation(s)
- Wei Liu
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Nana Zhao
- Key
Lab of Biomedical Materials of Natural Macromolecules (Beijing University
of Chemical Technology, Ministry of Education), Beijing, 100029, China
- Beijing Laboratory
of Biomedical Materials, Beijing University
of Chemical Technology, Beijing, 100029, China
- Beijing
Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- College
of Materials Science and Engineering, Beijing
University of Chemical Technology, Beijing, 100029, China
| | - Qi Yin
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Xiaoyi Zhao
- Key
Lab of Biomedical Materials of Natural Macromolecules (Beijing University
of Chemical Technology, Ministry of Education), Beijing, 100029, China
- Beijing Laboratory
of Biomedical Materials, Beijing University
of Chemical Technology, Beijing, 100029, China
- Beijing
Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- College
of Materials Science and Engineering, Beijing
University of Chemical Technology, Beijing, 100029, China
| | - Kangli Guo
- Key
Lab of Biomedical Materials of Natural Macromolecules (Beijing University
of Chemical Technology, Ministry of Education), Beijing, 100029, China
- Beijing Laboratory
of Biomedical Materials, Beijing University
of Chemical Technology, Beijing, 100029, China
- Beijing
Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- College
of Materials Science and Engineering, Beijing
University of Chemical Technology, Beijing, 100029, China
| | - Yifan Xian
- Key
Lab of Biomedical Materials of Natural Macromolecules (Beijing University
of Chemical Technology, Ministry of Education), Beijing, 100029, China
- Beijing
Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- College
of Materials Science and Engineering, Beijing
University of Chemical Technology, Beijing, 100029, China
| | - Siwei Li
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Chunlan Wang
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Miaomiao Zhu
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Yurong Du
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Fu-Jian Xu
- Key
Lab of Biomedical Materials of Natural Macromolecules (Beijing University
of Chemical Technology, Ministry of Education), Beijing, 100029, China
- Beijing Laboratory
of Biomedical Materials, Beijing University
of Chemical Technology, Beijing, 100029, China
- Beijing
Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- College
of Materials Science and Engineering, Beijing
University of Chemical Technology, Beijing, 100029, China
| | - Changyong Wang
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Jin Zhou
- Beijing
Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
31
|
Feng Q, Li Q, Zhou H, Sun L, Lin C, Jin Y, Wang D, Guo G. The role of major immune cells in myocardial infarction. Front Immunol 2023; 13:1084460. [PMID: 36741418 PMCID: PMC9892933 DOI: 10.3389/fimmu.2022.1084460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Myocardial infarction (MI) is a cardiovascular disease (CVD) with high morbidity and mortality worldwide, often leading to adverse cardiac remodeling and heart failure, which is a serious threat to human life and health. The immune system makes an important contribution to the maintenance of normal cardiac function. In the disease process of MI, necrotic cardiomyocytes release signals that activate nonspecific immunity and trigger the action of specific immunity. Complex immune cells play an important role in all stages of MI progression by removing necrotic cardiomyocytes and tissue and promoting the healing of damaged tissue cells. With the development of biomaterials, cardiac patches have become an emerging method of repairing MI, and the development of engineered cardiac patches through the construction of multiple animal models of MI can help treat MI. This review introduces immune cells involved in the development of MI, summarizes the commonly used animal models of MI and the newly developed cardiac patch, so as to provide scientific reference for the accurate diagnosis and effective treatment of MI.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China,Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Liqun Sun
- Department of Pathogenobiology, Jilin University Mycology Research Center, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China,*Correspondence: Gongliang Guo,
| | - Gongliang Guo
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China,*Correspondence: Gongliang Guo,
| |
Collapse
|
32
|
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: 10] [Impact Index Per Article: 3.3] [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
|
33
|
Meng J, Xiao B, Wu F, Sun L, Li B, Guo W, Hu X, Xu X, Wen T, Liu J, Xu H. Co-axial fibrous scaffolds integrating with carbon fiber promote cardiac tissue regeneration post myocardial infarction. Mater Today Bio 2022; 16:100415. [PMID: 36105673 PMCID: PMC9465342 DOI: 10.1016/j.mtbio.2022.100415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Myocardium is an excitable tissue with electrical conductivity and mechanical strength. In this work, carbon fibers (CFs) and co-axial fibrous mesh were integrated which combined the high modulus and excellent electrical conductivity of CFs and the fibrous and porous structures of the electrospun fibers. The scaffold was fabricated by simply integrating coaxial electrospun fibers and carbon fibers through a freeze-drying procedure. It was shown that the integration of carbon fibers have the conductivity and Young's modulus of the fibrous mesh increased significantly, meanwhile, upregulated the expression of CX43, α-actinin, RhoA of the neonatal rat primary cardiomyocytes and primary human umbilical vein endothelial cells (HUVECs), and promoted the secretion of VEGF of HUVECs. Moreover, the cardiomyocytes grown on the scaffolds increased the ability of HUVECs migration. When implanted to the injury area post myocardial infraction, the scaffolds were able to effectively enhance the tissue regeneration and new vessel formation, which rescued the heart dysfunction induced by the myocardial infraction, evidenced by the results of echocardiography and histochemical analysis. In conclusion, the composite scaffolds could promote the myocardium regeneration and function's recovery by enhancing cardiomyocytes maturation and angiogenesis and establishing the crosstalk between the cardiomyocytes and the vascular endothelial cells.
Collapse
Affiliation(s)
- Jie Meng
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Bo Xiao
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Fengxin Wu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Lihong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Bo Li
- Peking Union Medical College, Beijing, 100730, China
| | - Wen Guo
- Peking Union Medical College, Beijing, 100730, China
| | - Xuechun Hu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Xuegai Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Tao Wen
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jian Liu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
34
|
Montazerian H, Davoodi E, Baidya A, Badv M, Haghniaz R, Dalili A, Milani AS, Hoorfar M, Annabi N, Khademhosseini A, Weiss PS. Bio-macromolecular design roadmap towards tough bioadhesives. Chem Soc Rev 2022; 51:9127-9173. [PMID: 36269075 PMCID: PMC9810209 DOI: 10.1039/d2cs00618a] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Emerging sutureless wound-closure techniques have led to paradigm shifts in wound management. State-of-the-art biomaterials offer biocompatible and biodegradable platforms enabling high cohesion (toughness) and adhesion for rapid bleeding control as well as robust attachment of implantable devices. Tough bioadhesion stems from the synergistic contributions of cohesive and adhesive interactions. This Review provides a biomacromolecular design roadmap for the development of tough adhesive surgical sealants. We discuss a library of materials and methods to introduce toughness and adhesion to biomaterials. Intrinsically tough and elastic polymers are leveraged primarily by introducing strong but dynamic inter- and intramolecular interactions either through polymer chain design or using crosslink regulating additives. In addition, many efforts have been made to promote underwater adhesion via covalent/noncovalent bonds, or through micro/macro-interlock mechanisms at the tissue interfaces. The materials settings and functional additives for this purpose and the related characterization methods are reviewed. Measurements and reporting needs for fair comparisons of different materials and their properties are discussed. Finally, future directions and further research opportunities for developing tough bioadhesive surgical sealants are highlighted.
Collapse
Affiliation(s)
- Hossein Montazerian
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, Los Angeles, California 90024, USA.
| | - Elham Davoodi
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, Los Angeles, California 90024, USA.
- Multi-Scale Additive Manufacturing Lab, Mechanical and Mechatronics Engineering Department, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Avijit Baidya
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
| | - Maryam Badv
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, Los Angeles, California 90024, USA.
| | - Arash Dalili
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Abbas S Milani
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Mina Hoorfar
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
- School of Engineering and Computer Science, University of Victoria, Victoria, British Columbia V8P 3E6, Canada
| | - Nasim Annabi
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, Los Angeles, California 90024, USA.
| | - Paul S Weiss
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, USA
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
35
|
Xu K, Wu X, Zhang X, Xing M. Bridging wounds: tissue adhesives' essential mechanisms, synthesis and characterization, bioinspired adhesives and future perspectives. BURNS & TRAUMA 2022; 10:tkac033. [PMID: 36225327 PMCID: PMC9548443 DOI: 10.1093/burnst/tkac033] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/29/2022] [Indexed: 11/05/2022]
Abstract
Bioadhesives act as a bridge in wound closure by forming an effective interface to protect against liquid and gas leakage and aid the stoppage of bleeding. To their credit, tissue adhesives have made an indelible impact on almost all wound-related surgeries. Their unique properties include minimal damage to tissues, low chance of infection, ease of use and short wound-closure time. In contrast, classic closures, like suturing and stapling, exhibit potential additional complications with long operation times and undesirable inflammatory responses. Although tremendous progress has been made in the development of tissue adhesives, they are not yet ideal. Therefore, highlighting and summarizing existing adhesive designs and synthesis, and comparing the different products will contribute to future development. This review first provides a summary of current commercial traditional tissue adhesives. Then, based on adhesion interaction mechanisms, the tissue adhesives are categorized into three main types: adhesive patches that bind molecularly with tissue, tissue-stitching adhesives based on pre-polymer or precursor solutions, and bioinspired or biomimetic tissue adhesives. Their specific adhesion mechanisms, properties and related applications are discussed. The adhesion mechanisms of commercial traditional adhesives as well as their limitations and shortcomings are also reviewed. Finally, we also discuss the future perspectives of tissue adhesives.
Collapse
Affiliation(s)
- Kaige Xu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Xiaozhuo Wu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Xingying Zhang
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | | |
Collapse
|
36
|
Xia D, Chen J, Zhang Z, Dong M. Emerging polymeric biomaterials and manufacturing techniques in regenerative medicine. AGGREGATE 2022; 3. [DOI: 10.1002/agt2.176] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
AbstractThe current demand for patients’ organ and tissue repair and regeneration is continually increasing, where autologous or allograft is the golden standard treatment in the clinic. However, due to the shortage of donors, mismatched size and modality, functional loss of the donor region, possible immune rejection, and so forth, the application of auto‐/allo‐grafts is frequently hindered in many cases. In order to solve these problems, artificial constructs structurally and functionally imitating the extracellular matrix have been developed as substitutes to promoting cell attachment, proliferation, and differentiation, and ultimately forming functional tissues or organs for better tissue regeneration. Particularly, polymeric materials have been widely utilized in regenerative medicine because of their ease of manufacturing, flexibility, biocompatibility, as well as good mechanical, chemical, and thermal properties. This review presents a comprehensive overview of a variety of polymeric materials, their fabrication methods as well applications in regenerative medicine. Finally, we discussed the future challenges and perspectives in the development and clinical transformation of polymeric biomaterials.
Collapse
Affiliation(s)
- Dan Xia
- Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, School of Materials Science and Engineering Hebei University of Technology Tianjin China
| | - Jiatian Chen
- Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, School of Materials Science and Engineering Hebei University of Technology Tianjin China
| | - Zhongyang Zhang
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Aarhus Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Aarhus Denmark
| |
Collapse
|
37
|
Wang S, Zhao Q, Li J, Du X. Morphing-to-Adhesion Polysaccharide Hydrogel for Adaptive Biointerfaces. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42420-42429. [PMID: 36083279 DOI: 10.1021/acsami.2c10117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Reliable functions of medical implants highly depend on biocompatible, conformal, and stable biointerfaces for seamless biointegration with biological tissues. Though flexible biointerfaces based on synthetic hydrogels have shown promise in optimizing implant biointegration via surgical suturing, physical attachment, or manual preshaping, they still suffer from poor adaptability, such as tissue damage by surgical suturing, low bioactivity, and difficulties in conformal contact and stable fixation, especially for specific tissues of large surface curvatures. Here, we report a bilayer hydrogel-based adaptive biointerface (HAB) made of two polysaccharide derivates, N-hydroxysuccinimide (NHS) ester-activated alginate and chitosan, harnessing dual advantages of their different swelling and active groups. Leveraging on the differential swelling between the two hydrogel layers and covalent linkages with active groups at hydrogel interfaces, HABs can be programmed into sealed tubes with tunable diameters via water-induced compliable shape morphing and instant interfacial adhesion. We further demonstrate that the polysaccharide-based morphing-to-adhesion HAB possesses outstanding bioactivity in directing cellular focal adhesion and intercellular junction, versatile geometrical adaptability to diverse tubular tissues with a wide range of surface curvatures (2.8 × 102-1.3 × 103 m-1), and excellent mechanical stability in high load-/shear-bearing physiological environments (blood flow volume: 85 mm·s-1). HABs overcome the limitations of existing biointerfaces in terms of poor bioactivity and difficult biointegration with biological tissues of large surface curvatures, holding promise to open new avenues for adaptive biointerfaces and reliable medical implants.
Collapse
Affiliation(s)
- Shanshan Wang
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences, Beijing 100083, China
| | - Qilong Zhao
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
| | - Jinhong Li
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences, Beijing 100083, China
| | - Xuemin Du
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
| |
Collapse
|
38
|
Csöbönyeiová M, Beerová N, Klein M, Debreová-Čeháková M, Danišovič Ľ. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci 2022; 23:10314. [PMID: 36142245 PMCID: PMC9499607 DOI: 10.3390/ijms231810314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Because of cardiomyocyte death or dysfunction frequently caused by myocardial infarction (MI), heart failure is a leading cause of morbidity and mortality in modern society. Paradoxically, only limited and non-curative therapies for heart failure or MI are currently available. As a result, over the past two decades research has focused on developing cell-based approaches promoting the regeneration of infarcted tissue. Cell-based therapies for myocardial regeneration include powerful candidates, such as multipotent stem cells (mesenchymal stem cells (MSCs), bone-marrow-derived stem cells, endothelial progenitor cells, and hematopoietic stem cells) and induced pluripotent stem cells (iPSCs). These possess unique properties, such as potency to differentiate into desired cell types, proliferation capacity, and patient specificity. Preclinical and clinical studies have demonstrated modest improvement in the myocardial regeneration and reduced infarcted areas upon transplantation of pluripotent or multipotent stem cells. Another cell population that need to be considered as a potential source for cardiac regeneration are telocytes found in different organs, including the heart. Their therapeutic effect has been studied in various heart pathologies, such as MI, arrhythmias, or atrial amyloidosis. The most recent cell-free therapeutic tool relies on the cardioprotective effect of complex cargo carried by small membrane-bound vesicles-exosomes-released from stem cells via exocytosis. The MSC/iPSC-derived exosomes could be considered a novel exosome-based therapy for cardiovascular diseases thanks to their unique content. There are also other cell-free approaches, e.g., gene therapy, or acellular cardiac patches. Therefore, our review provides the most recent insights into the novel strategies for myocardial repair based on the regenerative potential of different cell types and cell-free approaches.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Nikoleta Beerová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Martin Klein
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Debreová-Čeháková
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
39
|
Mishra K, Devi N, Siwal SS, Zhang Q, Alsanie WF, Scarpa F, Thakur VK. Ionic Liquid-Based Polymer Nanocomposites for Sensors, Energy, Biomedicine, and Environmental Applications: Roadmap to the Future. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202187. [PMID: 35853696 PMCID: PMC9475560 DOI: 10.1002/advs.202202187] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/30/2022] [Indexed: 05/19/2023]
Abstract
Current interest toward ionic liquids (ILs) stems from some of their novel characteristics, like low vapor pressure, thermal stability, and nonflammability, integrated through high ionic conductivity and broad range of electrochemical strength. Nowadays, ionic liquids represent a new category of chemical-based compounds for developing superior and multifunctional substances with potential in several fields. ILs can be used in solvents such as salt electrolyte and additional materials. By adding functional physiochemical characteristics, a variety of IL-based electrolytes can also be used for energy storage purposes. It is hoped that the present review will supply guidance for future research focused on IL-based polymer nanocomposites electrolytes for sensors, high performance, biomedicine, and environmental applications. Additionally, a comprehensive overview about the polymer-based composites' ILs components, including a classification of the types of polymer matrix available is provided in this review. More focus is placed upon ILs-based polymeric nanocomposites used in multiple applications such as electrochemical biosensors, energy-related materials, biomedicine, actuators, environmental, and the aviation and aerospace industries. At last, existing challenges and prospects in this field are discussed and concluding remarks are provided.
Collapse
Affiliation(s)
- Kirti Mishra
- Department of ChemistryM.M. Engineering CollegeMaharishi Markandeshwar (Deemed to be University)Mullana‐AmbalaHaryana133207India
| | - Nishu Devi
- Mechanics and Energy LaboratoryDepartment of Civil and Environmental EngineeringNorthwestern University2145 Sheridan RoadEvanstonIL60208USA
| | - Samarjeet Singh Siwal
- Department of ChemistryM.M. Engineering CollegeMaharishi Markandeshwar (Deemed to be University)Mullana‐AmbalaHaryana133207India
| | - Qibo Zhang
- Key Laboratory of Ionic Liquids MetallurgyFaculty of Metallurgical and Energy EngineeringKunming University of Science and TechnologyKunming650093P. R. China
- State Key Laboratory of Complex Nonferrous Metal Resources Cleaning Utilization in Yunnan ProvinceKunming650093P. R. China
| | - Walaa F. Alsanie
- Department of Clinical Laboratories SciencesThe Faculty of Applied Medical SciencesTaif UniversityP.O. Box 11099Taif21944Saudi Arabia
| | - Fabrizio Scarpa
- Bristol Composites InstituteUniversity of BristolBristolBS8 1TRUK
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research CenterScotland's Rural College (SRUC)Kings Buildings, West Mains RoadEdinburghEH9 3JGUK
- School of EngineeringUniversity of Petroleum and Energy Studies (UPES)DehradunUttarakhand248007India
| |
Collapse
|
40
|
Mondal P, Chakraborty I, Chatterjee K. Injectable Adhesive Hydrogels for Soft tissue Reconstruction: A Materials Chemistry Perspective. CHEM REC 2022; 22:e202200155. [PMID: 35997710 DOI: 10.1002/tcr.202200155] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/30/2022] [Indexed: 11/09/2022]
Abstract
Injectable bioadhesives offer several advantages over conventional staples and sutures in surgery to seal and close incisions or wounds. Despite the growing research in recent years few injectable bioadhesives are available for clinical use. This review summarizes the key chemical features that enable the development and improvements in the use of polymeric injectable hydrogels as bioadhesives or sealants, their design requirements, the gelation mechanism, synthesis routes, and the role of adhesion mechanisms and strategies in different biomedical applications. It is envisaged that developing a deep understanding of the underlying materials chemistry principles will enable researchers to effectively translate bioadhesive technologies into clinically-relevant products.
Collapse
Affiliation(s)
- Pritiranjan Mondal
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Indranil Chakraborty
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| |
Collapse
|
41
|
Cao W, Peng S, Yao Y, Xie J, Li S, Tu C, Gao C. A nanofibrous membrane loaded with doxycycline and printed with conductive hydrogel strips promotes diabetic wound healing in vivo. Acta Biomater 2022; 152:60-73. [DOI: 10.1016/j.actbio.2022.08.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/01/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
|
42
|
Zhang Y, Chen H, Li J. Recent advances on gelatin methacrylate hydrogels with controlled microstructures for tissue engineering. Int J Biol Macromol 2022; 221:91-107. [DOI: 10.1016/j.ijbiomac.2022.08.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 12/12/2022]
|
43
|
Li Y, Qiu X. Bioelectricity-coupling patches for repairing impaired myocardium. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1787. [PMID: 35233963 DOI: 10.1002/wnan.1787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/27/2021] [Accepted: 01/31/2022] [Indexed: 11/10/2022]
Abstract
Cardiac abnormalities, which account for extensive burdens on public health and economy, drive necessary attempts to revolutionize the traditional therapeutic system. Advances in cardiac tissue engineering have expanded a highly efficacious platform to address cardiovascular events, especially cardiac infarction. Current efforts to overcome biocompatible limitations highlight the constructs of a conductive cardiac patch to accelerate the industrial and clinical landscape that is amenable for patient-accurate therapy, regenerative medicine, disease modeling, and drug delivery. With the notion that cardiac tissue synchronically contracts triggered by electrical pulses, the cardiac patches based on conductive materials are developed and treated on the dysfunctional heart. In this review, we systematically summarize distinct conductive materials serving as the most promising alternatives (conductive nanomaterials, conductive polymers, piezoelectric polymers, and ionic electrolytes) to achieve electric signal transmission and engineered cardiac tissues. Existing applications are discussed considering how these patches containing conductive candidates are fabricated into diverse forms with major strategies. Ultimately, we try to define a new concept as a bioelectricity-coupling patch that provides a favorable cardiac micro-environment for cardiac functional activities. Underlying challenges and prospects are presented regarding industrial processing and cardiovascular treatment of conductive patch progress. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease.
Collapse
Affiliation(s)
- Yuedan Li
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaozhong Qiu
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Zhu S, Yu C, Liu N, Zhao M, Chen Z, Liu J, Li G, Huang H, Guo H, Sun T, Chen J, Zhuang J, Zhu P. Injectable conductive gelatin methacrylate / oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact Mater 2022; 13:119-134. [PMID: 35224296 PMCID: PMC8844712 DOI: 10.1016/j.bioactmat.2021.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 02/03/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) transplantation has been proposed as a promising treatment modality for myocardial infarction (MI), but the low retention rate remains a considerable challenge. Injectable natural polymer hydrogels with conductivity ability are highly desirable as cell delivery vehicles to repair infarct myocardium and restore the cardiac function. In this work, we developed a hydrogel system based on gelatin methacrylate (GelMA) and oxidized dextran (ODEX) as cell delivery vehicles for MI. And dopamine could be used as a reductant of graphene oxide (GO) to form reductive GO (rGO). By adjusting the amount of rGO, the conductivity of hydrogels with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. In vitro cell experiments showed that the prepared hydrogels had excellent biocompatibility and cell delivery ability of UCMSCs. More importantly, GelMA-O5/rGO hydrogel could promote UCMSCs growth and proliferation, improve the myocardial differentiation ability of UCMSCs, and up-regulate the expression of cTnI and Cx43. Further in vivo experiments demonstrated that GelMA-O5/rGO/UCMSCs Hydrogel could significantly improve the ejection fraction (EF) of rats and significantly reduce myocardial infarct area compared to PBS group, promote the survival of UCMSCs, enhance the expression level of cTnI and Cx43, and decrease the expression level of caspase-3. The findings of this study suggested that the injectable conductive GelMA-O5/rGO hydrogel encapsulating UCMSCs could improve damaged myocardial tissue and reconstruct myocardial function, which will be a promising therapeutic strategy for cardiac repair. Conducting interpenetrating polymer network (IPN) hydrogels were synthesized for myocardial infarction treatment. The conductivity of hydrogel with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. The hydrogel could promote the growth and proliferation of UCMSCs, and improve the myocardial differentiation ability of UCMSCs. The hydrogel could reduce infarct size and cardiac fibrosis in the infarct zone, increase ventricular ejection fraction. The hydrogel could promote the survival of UCMSCs, up-regulate the expression level of cTnI and Cx43, down-regulate the expression level of caspase-3.
Collapse
|
45
|
Ghovvati M, Kharaziha M, Ardehali R, Annabi N. Recent Advances in Designing Electroconductive Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200055. [PMID: 35368150 PMCID: PMC9262872 DOI: 10.1002/adhm.202200055] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/12/2022] [Indexed: 12/19/2022]
Abstract
Implantable cardiac patches and injectable hydrogels are among the most promising therapies for cardiac tissue regeneration following myocardial infarction. Incorporating electrical conductivity into these patches and hydrogels is found to be an efficient method to improve cardiac tissue function. Conductive nanomaterials such as carbon nanotube, graphene oxide, gold nanorod, as well as conductive polymers such as polyaniline, polypyrrole, and poly(3,4-ethylenedioxythiophene):polystyrene sulfonate are appealing because they possess the electroconductive properties of semiconductors with ease of processing and have potential to restore electrical signaling propagation through the infarct area. Numerous studies have utilized these materials for regeneration of biological tissues that possess electrical activities, such as cardiac tissue. In this review, recent studies on the use of electroconductive materials for cardiac tissue engineering and their fabrication methods are summarized. Moreover, recent advances in developing electroconductive materials for delivering therapeutic agents as one of emerging approaches for treating heart diseases and regenerating damaged cardiac tissues are highlighted.
Collapse
Affiliation(s)
- Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahshid Kharaziha
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
46
|
Zhao G, Zhou H, Jin G, Jin B, Geng S, Luo Z, Ge Z, Xu F. Rational Design of Electrically Conductive Biomaterials toward Excitable Tissues Regeneration. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Demirci G, Niedźwiedź MJ, Kantor-Malujdy N, El Fray M. Elastomer-Hydrogel Systems: From Bio-Inspired Interfaces to Medical Applications. Polymers (Basel) 2022; 14:1822. [PMID: 35566990 PMCID: PMC9104885 DOI: 10.3390/polym14091822] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Novel advanced biomaterials have recently gained great attention, especially in minimally invasive surgical techniques. By applying sophisticated design and engineering methods, various elastomer-hydrogel systems (EHS) with outstanding performance have been developed in the last decades. These systems composed of elastomers and hydrogels are very attractive due to their high biocompatibility, injectability, controlled porosity and often antimicrobial properties. Moreover, their elastomeric properties and bioadhesiveness are making them suitable for soft tissue engineering. Herein, we present the advances in the current state-of-the-art design principles and strategies for strong interface formation inspired by nature (bio-inspiration), the diverse properties and applications of elastomer-hydrogel systems in different medical fields, in particular, in tissue engineering. The functionalities of these systems, including adhesive properties, injectability, antimicrobial properties and degradability, applicable to tissue engineering will be discussed in a context of future efforts towards the development of advanced biomaterials.
Collapse
Affiliation(s)
| | | | | | - Miroslawa El Fray
- Department of Polymer and Biomaterials Science, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Al. Piastów 45, 70-311 Szczecin, Poland; (G.D.); (M.J.N.); (N.K.-M.)
| |
Collapse
|
48
|
Saghebasl S, Akbarzadeh A, Gorabi AM, Nikzamir N, SeyedSadjadi M, Mostafavi E. Biodegradable functional macromolecules as promising scaffolds for cardiac tissue engineering. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Solmaz Saghebasl
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Abolfazl Akbarzadeh
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Universal Scientific Education and Research Network (USERN) Tabriz Iran
| | - Armita Mahdavi Gorabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Nasrin Nikzamir
- Department of Chemistry, Science and Research Branch Islamic Azad University Tehran Iran
| | | | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford California USA
- Department of Medicine Stanford University School of Medicine Stanford California USA
| |
Collapse
|
49
|
Nair RS, Sobhan PK, Shenoy SJ, Prabhu MA, Rema AM, Ramachandran S, C Geetha S, V Pratheesh K, Mony MP, Raj R, Anilkumar TV. A porcine cholecystic extracellular matrix conductive scaffold for cardiac tissue repair. J Biomed Mater Res B Appl Biomater 2022; 110:2039-2049. [PMID: 35305082 DOI: 10.1002/jbm.b.35058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/22/2021] [Indexed: 11/08/2022]
Abstract
Cardiac tissue engineering using cells, scaffolds or signaling molecules is a promising approach for replacement or repair of damaged myocardium. This study addressed the contemporary need for a conductive biomimetic nanocomposite scaffold for cardiac tissue engineering by examining the use of a gold nanoparticle-incorporated porcine cholecystic extracellular matrix for the same. The scaffold had an electrical conductivity (0.74 ± 0.03 S/m) within the range of native myocardium. It was a suitable substrate for the growth and differentiation of cardiomyoblast (H9c2) as well as rat mesenchymal stem cells to cardiomyocyte-like cells. Moreover, as an epicardial patch, the scaffold promoted neovascularisation and cell proliferation in infarcted myocardium of rats. It was concluded that the gold nanoparticle coated cholecystic extracellular matrix is a prospective biomaterial for cardiac tissue engineering.
Collapse
Affiliation(s)
- Reshma S Nair
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Praveen K Sobhan
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Sachin J Shenoy
- Division of In Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Mukund A Prabhu
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Aswathy M Rema
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Surendran C Geetha
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Kanakarajan V Pratheesh
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Manjula P Mony
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Reshmi Raj
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Thapasimuthu V Anilkumar
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India.,School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| |
Collapse
|
50
|
Li M, Wu H, Yuan Y, Hu B, Gu N. Recent fabrications and applications of cardiac patch in myocardial infarction treatment. VIEW 2022. [DOI: 10.1002/viw.20200153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mei Li
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- The Laboratory Center for Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Hao Wu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Yuehui Yuan
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Benhui Hu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Ning Gu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Sciences and Medical Engineering Southeast University Nanjing China
| |
Collapse
|