1
|
Meng Y, Xu L, Cheng G. Bioelectronics hydrogels for implantable cardiac and brain disease medical treatment application. Int J Biol Macromol 2025; 299:139945. [PMID: 39837454 DOI: 10.1016/j.ijbiomac.2025.139945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Hydrogel-based bioelectronic systems offer significant benefits for point-of-care diagnosis, treatment of cardiac and cerebral disease, surgical procedures, and other medical applications, ushering in a new era of advancements in medical technology. Progress in hydrogel-based bioelectronics has advanced from basic instrument and sensing capabilities to sophisticated multimodal perceptions and feedback systems. Addressing challenges related to immune responses and inflammation regulation after implantation, physiological dynamic mechanism, biological toxicology as well as device size, power consumption, stability, and signal conversion is crucial for the practical implementation of hydrogel-based bioelectronics in medical implants. Therefore, further exploration of hydrogel-based bioelectronics is imperative, and a comprehensive review is necessary to steer the development of these technologies for use in implantable therapies for cardiac and brain/neural conditions. In this review, a concise overview is provided on the fundamental principles underlying ionic electronic and ionic bioelectronic mechanisms. Additionally, a comprehensive examination is conducted on various bioelectronic materials integrated within hydrogels for applications in implantable medical treatments. The analysis encompasses a detailed discussion on the representative structures and physical attributes of hydrogels. This includes an exploration of their intrinsic properties such as mechanical strength, dynamic capabilities, shape-memory features, stability, stretchability, and water retention characteristics. Moreover, the discussion extends to properties related to interactions with tissues or the environment, such as adhesiveness, responsiveness, and degradability. The intricate relationships between the structure and properties of hydrogels are thoroughly examined, along with an elucidation of how these properties influence their applications in implantable medical treatments. The review also delves into the processing techniques and characterization methods employed for hydrogels. Furthermore, recent breakthroughs in the applications of hydrogels are logically explored, covering aspects such as materials, structure, properties, functions, fabrication procedures, and hybridization with other materials. Finally, the review concludes by outlining the future prospects and challenges associated with hydrogels-based bioelectronics systems.
Collapse
Affiliation(s)
- Yanfang Meng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China
| | - Lin Xu
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| | - Guanggui Cheng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| |
Collapse
|
2
|
Vishwanath R, Biswas A, Modi U, Gupta S, Bhatia D, Solanki R. Programmable short peptides for modulating stem cell fate in tissue engineering and regenerative medicine. J Mater Chem B 2025; 13:2573-2591. [PMID: 39871657 DOI: 10.1039/d4tb02102a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Recent advancements in tissue engineering and regenerative medicine have introduced promising strategies to address tissue and organ deficiencies. This review highlights the critical role of short peptides, particularly their ability to self-assemble into matrices that mimic the extracellular matrix (ECM). These low molecular weight peptides exhibit target-specific activities, modulate gene expression, and influence cell differentiation pathways. They are stable, programmable, non-cytotoxic, biocompatible, biodegradable, capable of crossing the cell membrane and easy to synthesize. This review underscores the importance of peptide structure and concentration in directing stem cell differentiation and explores their diverse biomedical applications. Peptides such as Aβ1-40, Aβ1-42, RADA16, A13 and KEDW are discussed for their roles in modulating stem cell differentiation into neuronal, glial, myocardial, osteogenic, hepatocyte and pancreatic lineages. Furthermore, this review delves into the underlying signaling mechanisms, the chemistry and design of short peptides and their potential for engineering biocompatible materials that mimic stem cell microenvironments. Short peptide-based biomaterials and scaffolds represent a promising avenue in stem cell therapy, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Rohan Vishwanath
- School of Life Science, Central University of Gujarat, Gandhinagar-382030, India
| | - Abhijit Biswas
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
3
|
Shi P, Sun P, Lou C, Fang J, Zhang L, Xie B, Zhang C. Adventitial Injection of Hyaluronic Acid/Sodium Alginate Hydrogel Loaded With IL-33 Antibody Decreases Neointimal Hyperplasia. J Surg Res 2025; 305:107-117. [PMID: 39667249 DOI: 10.1016/j.jss.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/30/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Neointimal hyperplasia is one of the persistent complications after vascular interventions, and is the major cause of treatment failure. Interleukin-33 (IL-33) emerges as a crucial factor in many biological processes and plays an important role in vascular diseases. Adventitial injection is catching attention for its effectiveness and fewer side effects. We hypothesize that targeting IL-33 by adventitial injection can be a therapeutic method to inhibit neointimal hyperplasia. METHOD IL-33 expression was examined in human vein graft. The hydrogel was fabricated by the interaction of hyaluronic acid, sodium alginate, and CaCO3; and phosphate buffered saline (PBS) or IL-33 antibody or recombinant IL-33 was mixed within the hydrogel uniformly. A rat aortic wire injury-induced neointimal hyperplasia model was developed; rats were divided into three groups and received an adventitial injection of a hydrogel loaded with PBS or IL-33 antibody or recombinant IL-33 after wire injury. Tissues were harvested at day 21 and analyzed by histology and immunohistochemical staining. Hydrogel loaded with PBS, IL-33 antibody, or IL-33 was also used in a mouse carotid artery ligation neointimal hyperplasia model. RESULT There was a high expression of IL-33 in human vein graft neointima. Hydrogel can be successfully injected into the aortic wall and is encapsulated by the adventitia. The hydrogel could be seen beneath the adventitia after adventitial injection and was partly degraded at day 21. There was a significantly thinner neointimal thickness and less proliferation and inflammation in the IL-33 antibody group compared to the control group. On the contrary, the IL-33 group has a thicker neointima, increased proliferation, and inflammation. The mouse carotid artery ligation model showed similar results. CONCLUSIONS IL-33 plays a role in arterial neointimal hyperplasia in both human and rodent models; adventitial injection of hydrogel loaded with IL-33 antibody can effectively decrease neointimal thickness. Neutralizing IL-33 by IL-33 antibody may be a potential therapeutic method to inhibit intimal hyperplasia after vascular interventions.
Collapse
Affiliation(s)
- Pengfei Shi
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Peng Sun
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Zhengzhou, China; Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Chunyang Lou
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianbang Fang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwei Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boao Xie
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cong Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Maeso L, Eufrásio-da-Silva T, Deveci E, Dolatshahi-Pirouz A, Orive G. Latest progress of self-healing hydrogels in cardiac tissue engineering. Biomed Microdevices 2024; 26:36. [PMID: 39150571 DOI: 10.1007/s10544-024-00716-z] [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] [Accepted: 07/01/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases represent a significant public health challenge and are responsible for more than 4 million deaths annually in Europe alone (45% of all deaths). Among these, coronary-related heart diseases are a leading cause of mortality, accounting for 20% of all deaths. Cardiac tissue engineering has emerged as a promising strategy to address the limitations encountered after myocardial infarction. This approach aims to improve regulation of the inflammatory and cell proliferation phases, thereby reducing scar tissue formation and restoring cardiac function. In cardiac tissue engineering, biomaterials serve as hosts for cells and therapeutics, supporting cardiac restoration by mimicking the native cardiac environment. Various bioengineered systems, such as 3D scaffolds, injectable hydrogels, and patches play crucial roles in cardiac tissue repair. In this context, self-healing hydrogels are particularly suitable substitutes, as they can restore structural integrity when damaged. This structural healing represents a paradigm shift in therapeutic interventions, offering a more native-like environment compared to static, non-healable hydrogels. Herein, we sharply review the most recent advances in self-healing hydrogels in cardiac tissue engineering and their potential to transform cardiovascular healthcare.
Collapse
Affiliation(s)
- Lidia Maeso
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | | | - Enes Deveci
- Faculty of Pharmacy, Lokman Hekim University, Ankara, Turkey
| | | | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain.
- University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, 01007, Spain.
| |
Collapse
|
5
|
Memarian P, Bagher Z, Asghari S, Aleemardani M, Seifalian A. Emergence of graphene as a novel nanomaterial for cardiovascular applications. NANOSCALE 2024; 16:12793-12819. [PMID: 38919053 DOI: 10.1039/d4nr00018h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CDs) are the foremost cause of death worldwide. Several promising therapeutic methods have been developed for this approach, including pharmacological, surgical intervention, cell therapy, or biomaterial implantation since heart tissue is incapable of regenerating and healing on its own. The best treatment for heart failure to date is heart transplantation and invasive surgical intervention, despite their invasiveness, donor limitations, and the possibility of being rejected by the patient's immune system. To address these challenges, research is being conducted on less invasive and efficient methods. Consequently, graphene-based materials (GBMs) have attracted a great deal of interest in the last decade because of their exceptional mechanical, electrical, chemical, antibacterial, and biocompatibility properties. An overview of GBMs' applications in the cardiovascular system has been presented in this article. Following a brief explanation of graphene and its derivatives' properties, the potential of GBMs to improve and restore cardiovascular system function by using them as cardiac tissue engineering, stents, vascular bypass grafts,and heart valve has been discussed.
Collapse
Affiliation(s)
- Paniz Memarian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sheida Asghari
- Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, S3 7HQ, UK.
- Department of Translational Health Science, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK.
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
| |
Collapse
|
6
|
Liang J, Lv R, Li M, Chai J, Wang S, Yan W, Zheng Z, Li P. Hydrogels for the Treatment of Myocardial Infarction: Design and Therapeutic Strategies. Macromol Biosci 2024; 24:e2300302. [PMID: 37815522 DOI: 10.1002/mabi.202300302] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) have become the leading global burden of diseases in recent years and are the primary cause of human mortality and loss of healthy life expectancy. Myocardial infarction (MI) is the top cause of CVDs-related deaths, and its incidence is increasing worldwide every year. Recently, hydrogels have garnered great interest from researchers as a promising therapeutic option for cardiac tissue repair after MI. This is due to their excellent properties, including biocompatibility, mechanical properties, injectable properties, anti-inflammatory properties, antioxidant properties, angiogenic properties, and conductive properties. This review discusses the advantages of hydrogels as a novel treatment for cardiac tissue repair after MI. The design strategies of various hydrogels in MI treatment are then summarized, and the latest research progress in the field is classified. Finally, the future perspectives of this booming field are also discussed at the end of this review.
Collapse
Affiliation(s)
- Jiaheng Liang
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
- Laboratory for Advanced Interfacial Materials and Devices, Department of Applied Biology and Chemical Technology (ABCT), Research Institute for Intelligent Wearable Systems (RI-IWEAR), The Hong Kong Polytechnic University, Hong Kong, SAR, 999077, China
| | - Ronghao Lv
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
| | - Maorui Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
| | - Jin Chai
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
| | - Shuo Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710072, China
| | - Zijian Zheng
- Laboratory for Advanced Interfacial Materials and Devices, Department of Applied Biology and Chemical Technology (ABCT), Research Institute for Intelligent Wearable Systems (RI-IWEAR), The Hong Kong Polytechnic University, Hong Kong, SAR, 999077, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE), Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, China
| |
Collapse
|
7
|
Liu X, Chen B, Chen J, Wang X, Dai X, Li Y, Zhou H, Wu LM, Liu Z, Yang Y. A Cardiac-Targeted Nanozyme Interrupts the Inflammation-Free Radical Cycle in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308477. [PMID: 37985164 DOI: 10.1002/adma.202308477] [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: 08/21/2023] [Revised: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Severe systemic inflammation following myocardial infarction (MI) is a major cause of patient mortality. MI-induced inflammation can trigger the production of free radicals, which in turn ultimately leads to increased inflammation in cardiac lesions (i.e., inflammation-free radicals cycle), resulting in heart failure and patient death. However, currently available anti-inflammatory drugs have limited efficacy due to their weak anti-inflammatory effect and poor accumulation at the cardiac site. Herein, a novel Fe-Cur@TA nanozyme is developed for targeted therapy of MI, which is generated by coordinating Fe3+ and anti-inflammatory drug curcumin (Cur) with further modification of tannic acid (TA). Such Fe-Cur@TA nanozyme exhibits excellent free radicals scavenging and anti-inflammatory properties by reducing immune cell infiltration, promoting macrophage polarization toward the M2-like phenotype, suppressing inflammatory cytokine secretion, and blocking the inflammatory free radicals cycle. Furthermore, due to the high affinity of TA for cardiac tissue, Fe-Cur@TA shows an almost tenfold greater in cardiac retention and uptake than Fe-Cur. In mouse and preclinical beagle dog MI models, Fe-Cur@TA nanozyme preserves cardiac function and reduces scar size, suggesting promising potential for clinical translation in cardiovascular disease.
Collapse
Affiliation(s)
- Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Binghua Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuan Wang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinfeng Dai
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuqing Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huayuan Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lian-Ming Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
8
|
Kafili G, Kabir H, Jalali Kandeloos A, Golafshan E, Ghasemi S, Mashayekhan S, Taebnia N. Recent advances in soluble decellularized extracellular matrix for heart tissue engineering and organ modeling. J Biomater Appl 2023; 38:577-604. [PMID: 38006224 PMCID: PMC10676626 DOI: 10.1177/08853282231207216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Despite the advent of tissue engineering (TE) for the remodeling, restoring, and replacing damaged cardiovascular tissues, the progress is hindered by the optimal mechanical and chemical properties required to induce cardiac tissue-specific cellular behaviors including migration, adhesion, proliferation, and differentiation. Cardiac extracellular matrix (ECM) consists of numerous structural and functional molecules and tissue-specific cells, therefore it plays an important role in stimulating cell proliferation and differentiation, guiding cell migration, and activating regulatory signaling pathways. With the improvement and modification of cell removal methods, decellularized ECM (dECM) preserves biochemical complexity, and bio-inductive properties of the native matrix and improves the process of generating functional tissue. In this review, we first provide an overview of the latest advancements in the utilization of dECM in in vitro model systems for disease and tissue modeling, as well as drug screening. Then, we explore the role of dECM-based biomaterials in cardiovascular regenerative medicine (RM), including both invasive and non-invasive methods. In the next step, we elucidate the engineering and material considerations in the preparation of dECM-based biomaterials, namely various decellularization techniques, dECM sources, modulation, characterizations, and fabrication approaches. Finally, we discuss the limitations and future directions in fabrication of dECM-based biomaterials for cardiovascular modeling, RM, and clinical translation.
Collapse
Affiliation(s)
- Golara Kafili
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Hannaneh Kabir
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, USA
| | | | - Elham Golafshan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Sara Ghasemi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
9
|
Yang Z, Zhang Y, Wang J, Yin J, Wang Z, Pei R. Cardiac organoid: multiple construction approaches and potential applications. J Mater Chem B 2023; 11:7567-7581. [PMID: 37477533 DOI: 10.1039/d3tb00783a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The human cardiac organoid (hCO) is three-dimensional tissue model that is similar to an in vivo organ and has great potential on heart development biology, disease modeling, drug screening and regenerative medicine. However, the construction of hCO presents a unique challenge compared with other organoids such as the lung, small intestine, pancreas, liver. Since heart disease is the dominant cause of death and the treatment of such disease is one of the most unmet medical needs worldwide, developing technologies for the construction and application of hCO is a critical task for the scientific community. In this review, we discuss the current classification and construction methods of hCO. In addition, we describe its applications in drug screening, disease modeling, and regenerative medicine. Finally, we propose the limitations of the cardiac organoid and future research directions. A detailed understanding of hCO will provide ways to improve its construction and expand its applications.
Collapse
Affiliation(s)
- Ziyi Yang
- School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Jine Wang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Jingbo Yin
- School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Zheng Wang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| |
Collapse
|
10
|
Wang J, Zhu X, Wang S, Zhang Y, Hua W, Liu Z, Zheng Y, Lu X. Phosphoproteomic and proteomic profiling in post-infarction chronic heart failure. Front Pharmacol 2023; 14:1181622. [PMID: 37405054 PMCID: PMC10315476 DOI: 10.3389/fphar.2023.1181622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
Background: Post-infarction chronic heart failure is the most common type of heart failure. Patients with chronic heart failure show elevated morbidity and mortality with limited evidence-based therapies. Phosphoproteomic and proteomic analysis can provide insights regarding molecular mechanisms underlying post-infarction chronic heart failure and explore new therapeutic approaches. Methods and results: Global quantitative phosphoproteomic and proteomic analysis of left ventricular tissues from post-infarction chronic heart failure rats were performed. A total of 33 differentially expressed phosphorylated proteins (DPPs) and 129 differentially expressed proteins were identified. Bioinformatic analysis indicated that DPPs were enriched mostly in nucleocytoplasmic transport and mRNA surveillance pathway. Bclaf1 Ser658 was identified after construction of Protein-Protein Interaction Network and intersection with Thanatos Apoptosis Database. Predicted Upstream Kinases of DPPs based on kinase-substrate enrichment analysis (KSEA) app showed 13 kinases enhanced in heart failure. Proteomic analysis showed marked changes in protein expression related to cardiac contractility and metabolism. Conclusion: The present study marked phosphoproteomics and proteomics changes in post-infarction chronic heart failure. Bclaf1 Ser658 might play a critical role in apoptosis in heart failure. PRKAA1, PRKACA, and PAK1 might serve as potential therapeutic targets for post-infarction chronic heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yu Zheng
- *Correspondence: Yu Zheng, ; Xiao Lu,
| | - Xiao Lu
- *Correspondence: Yu Zheng, ; Xiao Lu,
| |
Collapse
|
11
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
12
|
Feng J, Xing M, Qian W, Qiu J, Liu X. An injectable hydrogel combining medicine and matrix with anti-inflammatory and pro-angiogenic properties for potential treatment of myocardial infarction. Regen Biomater 2023; 10:rbad036. [PMID: 37153848 PMCID: PMC10159687 DOI: 10.1093/rb/rbad036] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/20/2023] [Accepted: 04/07/2023] [Indexed: 05/10/2023] Open
Abstract
One of the main illnesses that put people's health in jeopardy is myocardial infarction (MI). After MI, damaged or dead cells set off an initial inflammatory response that thins the ventricle wall and degrades the extracellular matrix. At the same time, the ischemia and hypoxic conditions resulting from MI lead to significant capillary obstruction and rupture, impairing cardiac function and reducing blood flow to the heart. Therefore, attenuating the initial inflammatory response and promoting angiogenesis are very important for the treatment of MI. Here, to reduce inflammation and promote angiogenesis in infarcted area, we report a new kind of injectable hydrogel composed of puerarin and chitosan via in situ self-assembly with simultaneous delivery of mesoporous silica nanoparticles (CHP@Si) for myocardial repair. On the one hand, puerarin degraded from CHP@Si hydrogel modulated the inflammatory response via inhibiting M1-type polarization of macrophages and expression of pro-inflammatory factors. On the other hand, silica ions and puerarin released from CHP@Si hydrogel showed synergistic activity to improve the cell viability, migration and angiogenic gene expression of HUVECs in both conventional and oxygen/glucose-deprived environments. It suggests that this multifunctional injectable CHP@Si hydrogel with good biocompatibility may be an appropriate candidate as a bioactive material for myocardial repair post-MI.
Collapse
Affiliation(s)
- Jiayin Feng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Xing
- Shanghai Xuhui District Dental Center, Shanghai 200032, China
| | - Wenhao Qian
- Shanghai Xuhui District Dental Center, Shanghai 200032, China
| | - Jiajun Qiu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
13
|
Karimi Hajishoreh N, Baheiraei N, Naderi N, Salehnia M, Razavi M. Left Ventricular Geometry and Angiogenesis Improvement in Rat Chronic Ischemic Cardiomyopathy following Injection of Encapsulated Mesenchymal Stem Cells. CELL JOURNAL 2022; 24:741-747. [PMID: 36527346 PMCID: PMC9790069 DOI: 10.22074/cellj.2022.557257.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Injection of hydrogel and cells into myocardial infarction (MI) patients is one of the emerging treatment techniques, however, it has some limitations such as a lack of electromechanical properties and neovascularization. We investigated the therapeutic potential of new electroactive hydrogel [reduced graphene oxide (rGO)/Alginate (ALG)] encapsulated human bone marrow mesenchymal stem cells (BMSCs). MATERIALS AND METHODS The experimental study involved ligating the left anterior descending coronary artery (LAD) in rat models of chronic ischemic cardiomyopathy. Echocardiograms were analyzed at 4 and 8 weeks after MI treatment. In the eighth week after injection in the heart, the rats were sacrificed. Histological and immunohistochemical analyses were performed using Hematoxylin and Eosin (H and E) staining, Masson's trichrome staining and anti-CD31 antibody to analyze tissue structure and detect neovascularization. RESULTS In comparison to the control and other treatment groups, MSCs encapsulated in rGO-ALG showed significant improvements in fractional shortening (FS), ejection fraction (EF), wall thickness and internal diameters (P<0.05). The morphological observation showed several small blood vessels formed around the transplantation site in all treated groups especially in the MSC-ALG-rGO group 8 weeks after the transplantation. Also, Masson's trichrome staining indicated an increased amount of collagen fibers in rGO-ALG-MSC. Microvessel density was significantly higher using MSC-ALG-rGO compared to controls (P<0.01). CONCLUSION This study demonstrates that intramyocardial injection of rGO/ALG, a bio-electroactive hydrogel, is safe for increasing LV function, neovascularization, and adjusting electrical characteristics following MI. The results confirm ALG promising capability as a natural therapeutic for cardiac regeneration.
Collapse
Affiliation(s)
- Negar Karimi Hajishoreh
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat
Modares University, Tehran, Iran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat
Modares University, Tehran, Iran,P.O.Box: 111-14115Tissue Engineering and Applied Cell Sciences DivisionDepartment of Anatomical
SciencesFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
P.O.Box: 1995614331Rajaie CardiovascularMedicaland Research CenterIran University of Medical SciencesTehranIran
Emails:,
| | - Nasim Naderi
- Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran,P.O.Box: 111-14115Tissue Engineering and Applied Cell Sciences DivisionDepartment of Anatomical
SciencesFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
P.O.Box: 1995614331Rajaie CardiovascularMedicaland Research CenterIran University of Medical SciencesTehranIran
Emails:,
| | - Mojdeh Salehnia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants and Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central
Florida, Orlando, Florida, United States
| |
Collapse
|
14
|
Jafari A, Ajji Z, Mousavi A, Naghieh S, Bencherif SA, Savoji H. Latest Advances in 3D Bioprinting of Cardiac Tissues. ADVANCED MATERIALS TECHNOLOGIES 2022; 7:2101636. [PMID: 38044954 PMCID: PMC10691862 DOI: 10.1002/admt.202101636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Indexed: 12/05/2023]
Abstract
Cardiovascular diseases (CVDs) are known as the major cause of death worldwide. In spite of tremendous advancements in medical therapy, the gold standard for CVD treatment is still transplantation. Tissue engineering, on the other hand, has emerged as a pioneering field of study with promising results in tissue regeneration using cells, biological cues, and scaffolds. Three-dimensional (3D) bioprinting is a rapidly growing technique in tissue engineering because of its ability to create complex scaffold structures, encapsulate cells, and perform these tasks with precision. More recently, 3D bioprinting has made its debut in cardiac tissue engineering, and scientists are investigating this technique for development of new strategies for cardiac tissue regeneration. In this review, the fundamentals of cardiac tissue biology, available 3D bioprinting techniques and bioinks, and cells implemented for cardiac regeneration are briefly summarized and presented. Afterwards, the pioneering and state-of-the-art works that have utilized 3D bioprinting for cardiac tissue engineering are thoroughly reviewed. Finally, regulatory pathways and their contemporary limitations and challenges for clinical translation are discussed.
Collapse
Affiliation(s)
- Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Zineb Ajji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Ali Mousavi
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
- Sorbonne University, UTC CNRS UMR 7338, Biomechanics and Bioengineering (BMBI), University of Technology of Compiègne, 60203 Compiègne, France
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02128, United States
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| |
Collapse
|
15
|
Wang Y, Li G, Yang L, Luo R, Guo G. Development of Innovative Biomaterials and Devices for the Treatment of Cardiovascular Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201971. [PMID: 35654586 DOI: 10.1002/adma.202201971] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Cardiovascular diseases have become the leading cause of death worldwide. The increasing burden of cardiovascular diseases has become a major public health problem and how to carry out efficient and reliable treatment of cardiovascular diseases has become an urgent global problem to be solved. Recently, implantable biomaterials and devices, especially minimally invasive interventional ones, such as vascular stents, artificial heart valves, bioprosthetic cardiac occluders, artificial graft cardiac patches, atrial shunts, and injectable hydrogels against heart failure, have become the most effective means in the treatment of cardiovascular diseases. Herein, an overview of the challenges and research frontier of innovative biomaterials and devices for the treatment of cardiovascular diseases is provided, and their future development directions are discussed.
Collapse
Affiliation(s)
- Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China
| | - Gaoyang Guo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China
| |
Collapse
|
16
|
Rocker AJ, Cavasin M, Johnson NR, Shandas R, Park D. Sulfonated Thermoresponsive Injectable Gel for Sequential Release of Therapeutic Proteins to Protect Cardiac Function after Myocardial Infarction. ACS Biomater Sci Eng 2022; 8:3883-3898. [PMID: 35950643 DOI: 10.1021/acsbiomaterials.2c00616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Myocardial infarction causes cardiomyocyte death and persistent inflammatory responses, which generate adverse pathological remodeling. Delivering therapeutic proteins from injectable materials in a controlled-release manner may present an effective biomedical approach for treating this disease. A thermoresponsive injectable gel composed of chitosan, conjugated with poly(N-isopropylacrylamide) and sulfonate groups, was developed for spatiotemporal protein delivery to protect cardiac function after myocardial infarction. The thermoresponsive gel delivered vascular endothelial growth factor (VEGF), interleukin-10 (IL-10), and platelet-derived growth factor (PDGF) in a sequential and sustained manner in vitro. An acute myocardial infarction mouse model was used to evaluate polymer biocompatibility and to determine therapeutic effects from the delivery system on cardiac function. Immunohistochemistry showed biocompatibility of the hydrogel, while the controlled delivery of the proteins reduced macrophage infiltration and increased vascularization. Echocardiography showed an improvement in ejection fraction and fractional shortening after injecting the thermal gel and proteins. A factorial design of experimental study was implemented to optimize the delivery system for the best combination and doses of proteins for further increasing stable vascularization and reducing inflammation using a subcutaneous injection mouse model. The results showed that VEGF, IL-10, and FGF-2 demonstrated significant contributions toward promoting long-term vascularization, while PDGF's effect was minimal.
Collapse
Affiliation(s)
- Adam J Rocker
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Maria Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Noah R Johnson
- Department of Neurology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robin Shandas
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
17
|
Xiao Y, Chen Y, Shao C, Wang Y, Hu S, Lei W. Strategies to improve the therapeutic effect of pluripotent stem cell-derived cardiomyocytes on myocardial infarction. Front Bioeng Biotechnol 2022; 10:973496. [PMID: 35992358 PMCID: PMC9388750 DOI: 10.3389/fbioe.2022.973496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Myocardial infarction (MI) is a common cardiovascular disease caused by permanent loss of cardiomyocytes and the formation of scar tissue due to myocardial ischemia. Mammalian cardiomyocytes lose their ability to proliferate almost completely in adulthood and are unable to repair the damage caused by MI. Therefore, transplantation of exogenous cells into the injured area for treatment becomes a promising strategy. Pluripotent stem cells (PSCs) have the ability to proliferate and differentiate into various cellular populations indefinitely, and pluripotent stem cell-derived cardiomyocytes (PSC-CMs) transplanted into areas of injury can compensate for part of the injuries and are considered to be one of the most promising sources for cell replacement therapy. However, the low transplantation rate and survival rate of currently transplanted PSC-CMs limit their ability to treat MI. This article focuses on the strategies of current research for improving the therapeutic efficacy of PSC-CMs, aiming to provide some inspiration and ideas for subsequent researchers to further enhance the transplantation rate and survival rate of PSC-CMs and ultimately improve cardiac function.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chunlai Shao
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| |
Collapse
|
18
|
Tariq U, Gupta M, Pathak S, Patil R, Dohare A, Misra SK. Role of Biomaterials in Cardiac Repair and Regeneration: Therapeutic Intervention for Myocardial Infarction. ACS Biomater Sci Eng 2022; 8:3271-3298. [PMID: 35867701 DOI: 10.1021/acsbiomaterials.2c00454] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heart failure or myocardial infarction (MI) is one of the world's leading causes of death. Post MI, the heart can develop pathological conditions such as ischemia, inflammation, fibrosis, and left ventricular dysfunction. However, current surgical approaches are sufficient for enhancing myocardial perfusion but are unable to reverse the pathological changes. Tissue engineering and regenerative medicine approaches have shown promising effects in the repair and replacement of injured cardiomyocytes. Additionally, biomaterial scaffolds with or without stem cells are established to provide an effective environment for cardiac regeneration. Excipients loaded with growth factors, cytokines, oligonucleotides, and exosomes are found to help in such cardiac eventualities by promoting angiogenesis, cardiomyocyte proliferation, and reducing fibrosis, inflammation, and apoptosis. Injectable hydrogels, nanocarriers, cardiac patches, and vascular grafts are some excipients that can help the self-renewal in the damaged heart but are not understood well yet, in the context of used biomaterials. This review focuses on the use of various biomaterial-based approaches for the regeneration and repair of cardiac tissue postoccurrence of MI. It also discusses the outlines of cardiac remodeling and current therapeutic approaches after myocardial infarction, which are translationally important with respect to used biomaterials. It provides comprehensive details of the biomaterial-based regenerative approaches, which are currently the focus of the research for cardiac repair and regeneration and can provide a broad outline for further improvements.
Collapse
Affiliation(s)
- Ubaid Tariq
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| | - Mahima Gupta
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| | - Subhajit Pathak
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| | - Ruchira Patil
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| | - Akanksha Dohare
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| | - Santosh K Misra
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India.,Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kalyanpur, Uttar Pradesh 208016, India
| |
Collapse
|
19
|
Hao T, Qian M, Zhang Y, Liu Q, Midgley AC, Liu Y, Che Y, Hou J, Zhao Q. An Injectable Dual-Function Hydrogel Protects Against Myocardial Ischemia/Reperfusion Injury by Modulating ROS/NO Disequilibrium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105408. [PMID: 35319828 PMCID: PMC9130918 DOI: 10.1002/advs.202105408] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/25/2022] [Indexed: 06/14/2023]
Abstract
Acute myocardial infarction (MI) is the leading cause of death worldwide. Exogenous delivery of nitric oxide (NO) to the infarcted myocardium has proven to be an effective strategy for treating MI due to the multiple physiological functions of NO. However, reperfusion of blood flow to the ischemic tissues is accompanied by the overproduction of toxic reactive oxygen species (ROS), which can further exacerbate tissue damage and compromise the therapeutic efficacy. Here, an injectable hydrogel is synthesized from the chitosan modified by boronate-protected diazeniumdiolate (CS-B-NO) that can release NO in response to ROS stimulation and thereby modulate ROS/NO disequilibrium after ischemia/reperfusion (I/R) injury. Furthermore, administration of CS-B-NO efficiently attenuated cardiac damage and adverse cardiac remodeling, promoted repair of the heart, and ameliorated cardiac function, unlike a hydrogel that only released NO, in a mouse model of myocardial I/R injury. Mechanistically, regulation of the ROS/NO balance activated the antioxidant defense system and protected against oxidative stress induced by I/R injury via adaptive regulation of the Nrf2-Keap1 pathway. Inflammation is then reduced by inhibition of the activation of NF-κB signaling. Collectively, these results show that this dual-function hydrogel may be a promising candidate for the protection of tissues and organs after I/R injury.
Collapse
Affiliation(s)
- Tian Hao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yating Zhang
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Qi Liu
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Yongzhe Che
- School of MedicineNankai UniversityTianjin300071China
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| |
Collapse
|
20
|
da Silva IGR, Pantoja BTDS, Almeida GHDR, Carreira ACO, Miglino MA. Bacterial Cellulose and ECM Hydrogels: An Innovative Approach for Cardiovascular Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23073955. [PMID: 35409314 PMCID: PMC8999934 DOI: 10.3390/ijms23073955] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are considered the leading cause of death in the world, accounting for approximately 85% of sudden death cases. In dogs and cats, sudden cardiac death occurs commonly, despite the scarcity of available pathophysiological and prevalence data. Conventional treatments are not able to treat injured myocardium. Despite advances in cardiac therapy in recent decades, transplantation remains the gold standard treatment for most heart diseases in humans. In veterinary medicine, therapy seeks to control clinical signs, delay the evolution of the disease and provide a better quality of life, although transplantation is the ideal treatment. Both human and veterinary medicine face major challenges regarding the transplantation process, although each area presents different realities. In this context, it is necessary to search for alternative methods that overcome the recovery deficiency of injured myocardial tissue. Application of biomaterials is one of the most innovative treatments for heart regeneration, involving the use of hydrogels from decellularized extracellular matrix, and their association with nanomaterials, such as alginate, chitosan, hyaluronic acid and gelatin. A promising material is bacterial cellulose hydrogel, due to its nanostructure and morphology being similar to collagen. Cellulose provides support and immobilization of cells, which can result in better cell adhesion, growth and proliferation, making it a safe and innovative material for cardiovascular repair.
Collapse
Affiliation(s)
- Izabela Gabriela Rodrigues da Silva
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; (I.G.R.d.S.); (B.T.d.S.P.); (G.H.D.R.A.); (A.C.O.C.)
| | - Bruna Tássia dos Santos Pantoja
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; (I.G.R.d.S.); (B.T.d.S.P.); (G.H.D.R.A.); (A.C.O.C.)
| | - Gustavo Henrique Doná Rodrigues Almeida
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; (I.G.R.d.S.); (B.T.d.S.P.); (G.H.D.R.A.); (A.C.O.C.)
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; (I.G.R.d.S.); (B.T.d.S.P.); (G.H.D.R.A.); (A.C.O.C.)
- NUCEL-Cell and Molecular Therapy Center, School of Medicine, Sao Paulo University, Sao Paulo 05508-270, Brazil
| | - Maria Angélica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; (I.G.R.d.S.); (B.T.d.S.P.); (G.H.D.R.A.); (A.C.O.C.)
- Correspondence:
| |
Collapse
|
21
|
Tang J, Cui X, Zhang Z, Xu Y, Guo J, Soliman BG, Lu Y, Qin Z, Wang Q, Zhang H, Lim KS, Woodfield TBF, Zhang J. Injection-Free Delivery of MSC-Derived Extracellular Vesicles for Myocardial Infarction Therapeutics. Adv Healthc Mater 2022; 11:e2100312. [PMID: 34310068 DOI: 10.1002/adhm.202100312] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/09/2021] [Indexed: 12/17/2022]
Abstract
As emerging therapeutic factors, extracellular vesicles (EVs) offer significant potential for myocardial infarction (MI) treatment. Current delivery approaches for EVs involve either intra-myocardial or intravenous injection, where both have inherent limitations for downstream clinical applications such as secondary tissue injury and low delivery efficiency. Herein, an injection-free approach for delivering EVs onto the heart surface to treat MI is proposed. By spraying a mixture of EVs, gelatin methacryloyl (GelMA) precursors, and photoinitiators followed by visible light irradiation for 30 s, EVs are physically entrapped within the GelMA hydrogel network covering the surface of the heart, resulting in an enhanced retention rate. Moreover, EVs are gradually released from the hydrogel network through a combination of diffusion and/or enzymatic degradation of the hydrogel, and they are effectively taken up by the sprayed tissue area. More importantly, the released EVs further migrate deep into myocardium tissue, which exerts an improved therapeutic effect. In an MI-induced mice model, the group treated with EVs-laden GelMA hydrogels shows significant recovery in cardiac function after 4 weeks. The work demonstrates a new strategy for delivering EVs into cardiac tissues for MI treatment in a localized manner with high retention.
Collapse
Affiliation(s)
- Junnan Tang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Zenglei Zhang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Yanyan Xu
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Jiacheng Guo
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Bram G Soliman
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Yongzheng Lu
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Zhen Qin
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials Sichuan University Chengdu Sichuan 61004 China
| | - Hu Zhang
- Henry E. Riggs School of Applied Life Sciences Keck Graduate Institute Claremont CA 91711 USA
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Jinying Zhang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| |
Collapse
|
22
|
Goonoo N. Tunable Biomaterials for Myocardial Tissue Regeneration: Promising New Strategies for Advanced Biointerface Control and Improved Therapeutic Outcomes. Biomater Sci 2022; 10:1626-1646. [DOI: 10.1039/d1bm01641e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Following myocardial infarction (MI) and the natural healing process, the cardiac mechanostructure changes significantly leading to reduced contractile ability and putting additional pressure on the heart muscle thereby increasing the...
Collapse
|
23
|
Li H, Yu B, Yang P, Zhan J, Fan X, Chen P, Liao X, Ou C, Cai Y, Chen M. Injectable AuNP-HA matrix with localized stiffness enhances the formation of gap junction in engrafted human induced pluripotent stem cell-derived cardiomyocytes and promotes cardiac repair. Biomaterials 2021; 279:121231. [PMID: 34739980 DOI: 10.1016/j.biomaterials.2021.121231] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/17/2021] [Accepted: 10/24/2021] [Indexed: 01/14/2023]
Abstract
Cell therapy offers a promising paradigm for heart tissue regeneration. Human induced pluripotent stem cells (hiPS) and their cardiac derivatives are emerging as a novel treatment for post-myocardial infarction repair. However, the immature phenotype and function of hiPS-derived cardiomyocytes (hiPS-CMs), particularly poor electrical coupling, limit their potential as a therapy. Herein, we developed a hybrid gold nanoparticle (AuNP)-hyaluronic acid (HA) hydrogel matrix encapsulating hiPS-CMs to overcome this limitation. Methacrylate-modified-HA was used as the backbone and crosslinked with a matrix metalloproteinase-2 (MMP-2) degradable peptide to obtain a MMP-2-responsive hydrogel; RGD peptide was introduced as an adhesion point to enhance biocompatibility; AuNPs were incorporated to regulate the mechanical and topological properties of the matrix by significantly increasing its stiffness and surface roughness, thereby accelerating gap junction formation in hiPS-CMs and orchestrating calcium handling via the αnβ1integrin-mediated ILK-1/p-AKT/GATA4 pathway. Transplanted AuNP-HA-hydrogel-encapsulated-hiPS-CMs developed more robust gap junctions in the infarcted mice heart and resynchronized electrical conduction of the ventricle post-myocardial infarction. The hiPS-CMs delivered by the hydrogels exerted stronger angiogenic effects, which also contributed to the recovery process. This study provides insight into constructing an injectable biomimetic for structural and functional renovation of the injured heart.
Collapse
Affiliation(s)
- Hekai Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Bin Yu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Pingzhen Yang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Jie Zhan
- Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, Foshan, 528300, China.
| | - Xianglin Fan
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Peier Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Xu Liao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Caiwen Ou
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Minsheng Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
24
|
Recent Advances in Cardiac Tissue Engineering for the Management of Myocardium Infarction. Cells 2021; 10:cells10102538. [PMID: 34685518 PMCID: PMC8533887 DOI: 10.3390/cells10102538] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Myocardium Infarction (MI) is one of the foremost cardiovascular diseases (CVDs) causing death worldwide, and its case numbers are expected to continuously increase in the coming years. Pharmacological interventions have not been at the forefront in ameliorating MI-related morbidity and mortality. Stem cell-based tissue engineering approaches have been extensively explored for their regenerative potential in the infarcted myocardium. Recent studies on microfluidic devices employing stem cells under laboratory set-up have revealed meticulous events pertaining to the pathophysiology of MI occurring at the infarcted site. This discovery also underpins the appropriate conditions in the niche for differentiating stem cells into mature cardiomyocyte-like cells and leads to engineering of the scaffold via mimicking of native cardiac physiological conditions. However, the mode of stem cell-loaded engineered scaffolds delivered to the site of infarction is still a challenging mission, and yet to be translated to the clinical setting. In this review, we have elucidated the various strategies developed using a hydrogel-based system both as encapsulated stem cells and as biocompatible patches loaded with cells and applied at the site of infarction.
Collapse
|
25
|
Mesenchymal Stem Cells for Cardiac Regeneration: from Differentiation to Cell Delivery. Stem Cell Rev Rep 2021; 17:1666-1694. [PMID: 33954876 DOI: 10.1007/s12015-021-10168-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are so far the most widely researched stem cells in clinics and used as an experimental cellular therapy module, particularly in cardiac regeneration and repair. Ever since the discovery of cardiomyogenesis induction in MSCs, a wide variety of differentiation protocols have been extensively used in preclinical models. However, pre differentiated MSC-derived cardiomyocytes have not been used in clinical trials; highlighting discrepancies and limitations in its use as a source of derived cardiomyocytes for transplantation to improve the damaged heart function. Therefore, this review article focuses on the strategies used to derive cardiomyocytes-like cells from MSCs isolated from three widely used tissue sources and their differentiation efficiencies. We have further discussed the role of MSCs in inducing angiogenesis as a cellular precursor to endothelial cells and its secretory aspects including exosomes. We have then discussed the strategies used for delivering cells in the damaged heart and how its retention plays a critical role in the overall outcome of the therapy. We have also conversed about the scope of the local and systemic modes of delivery of MSCs and the application of biomaterials to improve the overall delivery efficacy and function. We have finally discussed the advantages and limitations of cell delivery to the heart and the future scope of MSCs in cardiac regenerative therapy.
Collapse
|
26
|
Bai H, Sun P, Wei S, Xie B, Li M, Xu Y, Wang W, Liu Y, Zhang L, Wu H, Wang Z, Xing Y, Wang Z, Li J. A novel intramural TGF β 1 hydrogel delivery method to decrease murine abdominal aortic aneurysm and rat aortic pseudoaneurysm formation and progression. Biomed Pharmacother 2021; 137:111296. [PMID: 33545663 DOI: 10.1016/j.biopha.2021.111296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Aneurysms are generally the result of dilation of all 3 layers of the vessel wall, and pseudoaneurysms are the result of localized extravasation of blood that is contained by surrounding tissue. Since there is still no recommended protocol to decrease aneurysm formation and progression, we hypothesised that intramural delivery of TGF β1 hydrogel can decrease aneurysm and pseudoaneurysm formation and progression. MATERIALS Male C57BL/6 J mice (12-14 wk), SD rats (200 g) and pig abdominal aortas were used, and hydrogels were fabricated by the interaction of sodium alginate (SA), hyaluronic acid (HA) and CaCO3. METHODS A CaCl2 adventitial incubation model in mice and a decellularized human great saphenous vein patch angioplasty model in rats were used. TGF β1 hydrogel was intramurally delivered after CaCl2 incubation in mice; at day 7, the abdomen in some mice was reopened, and TGF β1 hydrogel was injected intramurally into the aorta. In rats, TGF β1 hydrogel was delivered intramurally after patch angioplasty completion. Tissues were harvested at day 14 and analysed by histology and immunohistochemistry staining. The pig aorta was also intramurally injected with hydrogel. RESULTS In mice, rhodamine hydrogel was still found between the medium and adventitia at day 14. In the mouse aneurysm model, there was a thicker wall and smaller amount of elastin breaks in the TGF β1 hydrogel-delivered groups both at day 0 and day 7 after CaCl2 incubation, and there were larger numbers of p-smad2- and TAK1-positive cells in the TGF β1 hydrogel-injected groups. In the rat decellularized human saphenous vein patch pseudoaneurysm model, there was a higher incidence of pseudoaneurysm formation when the patch was decellularized using 3% SDS, and delivery of TGF β1 hydrogel could effectively decrease the formation of pseudoaneurysm formation and increase p-smad2 and TAK1 expression. In pig aortas, hydrogels can be delivered between the medium and adventitia easily and successfully. CONCLUSIONS Intramural delivery of TGF β1 hydrogel can effectively decease aneurysm and pseudoaneurysm formation and progression in both mice and rats, and pig aortas can also be successfully intramurally injected with hydrogel. This technique may be a promising drug delivery method and therapeutic choice to decrease aneurysm and pseudoaneurysm formation and progression in the clinic.
Collapse
MESH Headings
- Aneurysm, False/metabolism
- Aneurysm, False/pathology
- Aneurysm, False/prevention & control
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Delayed-Action Preparations
- Dilatation, Pathologic
- Disease Models, Animal
- Disease Progression
- Drug Carriers
- Drug Compounding
- Hydrogels
- MAP Kinase Kinase Kinases/metabolism
- Male
- Mice, Inbred C57BL
- Phosphorylation
- Rats, Sprague-Dawley
- Smad2 Protein/metabolism
- Sus scrofa
- Transforming Growth Factor beta1/administration & dosage
- Mice
- Rats
Collapse
Affiliation(s)
- Hualong Bai
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China; Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Henan, China.
| | - Peng Sun
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Shunbo Wei
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Boao Xie
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Yanhua Xu
- Department of Internal Medicine, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Wang Wang
- Department of Physiology, Medical School of Zhengzhou University, Henan, China; Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Henan, China
| | - Yuanfeng Liu
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Liwei Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Haoliang Wu
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhiju Wang
- Department of Physiology, Medical School of Zhengzhou University, Henan, China; Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Henan, China
| | - Ying Xing
- Department of Physiology, Medical School of Zhengzhou University, Henan, China; Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Henan, China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China.
| | - Jing'an Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mould Technology (Ministry of Education), Zhengzhou University, Henan, China.
| |
Collapse
|
27
|
Silveira-Filho LM, Coyan GN, Adamo A, Luketich SK, Menallo G, D'Amore A, Wagner WR. Can a Biohybrid Patch Salvage Ventricular Function at a Late Time Point in the Post-Infarction Remodeling Process? ACTA ACUST UNITED AC 2021; 6:447-463. [PMID: 34095634 PMCID: PMC8165254 DOI: 10.1016/j.jacbts.2021.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/15/2023]
Abstract
A simple, biohybrid patch made of polymer (PECUU) and ECM, without cellular components, was able to induce positive remodeling features when applied over chronic infarcts with severely dilated hearts and high cardiac function impairment in rats. The remodeling benefit was particularly notable in a subgroup of the sickest rats with very low initial ejection fraction in which the echocardiographic endpoints were found to improve after treatment. This technological approach may hold promise for future translation to patients in a chronic scenario.
A biohybrid patch without cellular components was implanted over large infarcted areas in severely dilated hearts. Nonpatched animals were assigned to control or losartan therapy. Patch-implanted animals responded with better morphological and functional echocardiographic endpoints, which were more evident in a subgroup of animals with very low pre-treatment ejection fraction (<35%). Patched animals also had smaller infarcts than both nonpatched groups. This simple approach could hold promise for clinical translation and be applied using minimally invasive procedures over the epicardium in a large set of patients to induce better ventricular remodeling, especially among those who are especially frail.
Collapse
Key Words
- AT1R, angiotensin 1 receptor
- ECM, extracellular matrix
- EDA, end-diastolic area
- EF, ejection fraction
- ESA, end-systolic area
- FS, fractional shortening
- HF, heart failure
- LV, left ventricle
- LVEF, left ventricular ejection fraction
- LVFW, left ventricular free wall
- LVdd, left ventricular end-diastolic diameter
- LVsd, left ventricular end-systolic diameter
- M1, macrophage type 1
- M2, macrophage type 2
- MI, myocardial infarction
- MT, Masson trichrome
- PBS, phosphate-buffered saline
- PECUU, poly(ester carbonate urethane) urea
- PEUU, poly(ester urethane) urea
- SMA, smooth muscle actin
- biomaterial
- cardiac patch
- left ventricular remodeling
- myocardial infarction
Collapse
Affiliation(s)
- Lindemberg M Silveira-Filho
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Campinas, Sao Paulo, Brazil
| | - Garrett N Coyan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Arianna Adamo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samuel K Luketich
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Giorgio Menallo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Antonio D'Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,RiMED Foundation, Palermo, Italy
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
29
|
Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. Stem cell therapy in the heart: Biomaterials as a key route. Tissue Cell 2021; 71:101504. [PMID: 33607524 DOI: 10.1016/j.tice.2021.101504] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are one of the main concerns, nowadays causing a high rate of mortality in the world. The majority of conventional treatment protects the heart from failure progression. As a novel therapeutic way, Regenerative medicine in the heart includes cellular and noncellular approaches. Despite the irrefutable privileges of noncellular aspects such as administration of exosomes, utilizing of miRNAs, and growth factors, they cannot reverse necrotic or ischemic myocardium, hence recruiting of stem cells to help regenerative therapy in the heart seems indispensable. Stem cell lineages are varied and divided into two main groups namely pluripotent and adult stem cells. Not only has each of which own regenerative capacity, benefits, and drawbacks, but their turnover also close correlates with the target organ and/or tissue as well as the stage and level of failure. In addition to inefficient tissue integration due to the defects in delivering methods and poor retention of transplanted cells, the complexity of the heart and its movement also make more rigorous the repair process. Hence, utilizing biomaterials can make a key route to tackle such obstacles. In this review, we evaluate some natural products which can help stem cells in regenerative medicine of the cardiovascular system.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad, Iran; Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Torabi H, Mehdikhani M, Varshosaz J, Shafiee F. An innovative approach to fabricate a thermosensitive melatonin‐loaded conductive pluronic/chitosan hydrogel for myocardial tissue engineering. J Appl Polym Sci 2020. [DOI: 10.1002/app.50327] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hadis Torabi
- Department of Biomedical Engineering, Faculty of Engineering University of Isfahan Isfahan Iran
| | - Mehdi Mehdikhani
- Department of Biomedical Engineering, Faculty of Engineering University of Isfahan Isfahan Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center Isfahan University of Medical Sciences Isfahan Iran
- Department of Pharmaceutics School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences Isfahan Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
31
|
He L, Chen X. Cardiomyocyte Induction and Regeneration for Myocardial Infarction Treatment: Cell Sources and Administration Strategies. Adv Healthc Mater 2020; 9:e2001175. [PMID: 33000909 DOI: 10.1002/adhm.202001175] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Occlusion of coronary artery and subsequent damage or death of myocardium can lead to myocardial infarction (MI) and even heart failure-one of the leading causes of deaths world wide. Notably, myocardium has extremely limited regeneration potential due to the loss or death of cardiomyocytes (i.e., the cells of which the myocardium is comprised) upon MI. A variety of stem cells and stem cell-derived cardiovascular cells, in situ cardiac fibroblasts and endogenous proliferative epicardium, have been exploited to provide renewable cellular sources to treat injured myocardium. Also, different strategies, including direct injection of cell suspensions, bioactive molecules, or cell-incorporated biomaterials, and implantation of artificial cardiac scaffolds (e.g., cell sheets and cardiac patches), have been developed to deliver renewable cells and/or bioactive molecules to the MI site for the myocardium regeneration. This article briefly surveys cell sources and delivery strategies, along with biomaterials and their processing techniques, developed for MI treatment. Key issues and challenges, as well as recommendations for future research, are also discussed.
Collapse
Affiliation(s)
- Lihong He
- Department of Cell Biology Medical College of Soochow University Suzhou 215123 China
| | - Xiongbiao Chen
- Department of Mechanical Engineering Division of Biomedical Engineering University of Saskatchewan Saskatoon S7N5A9 Canada
| |
Collapse
|
32
|
Nguyen-Truong M, Li YV, Wang Z. Mechanical Considerations of Electrospun Scaffolds for Myocardial Tissue and Regenerative Engineering. Bioengineering (Basel) 2020; 7:E122. [PMID: 33022929 PMCID: PMC7711753 DOI: 10.3390/bioengineering7040122] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Biomaterials to facilitate the restoration of cardiac tissue is of emerging importance. While there are many aspects to consider in the design of biomaterials, mechanical properties can be of particular importance in this dynamically remodeling tissue. This review focuses on one specific processing method, electrospinning, that is employed to generate materials with a fibrous microstructure that can be combined with material properties to achieve the desired mechanical behavior. Current methods used to fabricate mechanically relevant micro-/nanofibrous scaffolds, in vivo studies using these scaffolds as therapeutics, and common techniques to characterize the mechanical properties of the scaffolds are covered. We also discuss the discrepancies in the reported elastic modulus for physiological and pathological myocardium in the literature, as well as the emerging area of in vitro mechanobiology studies to investigate the mechanical regulation in cardiac tissue engineering. Lastly, future perspectives and recommendations are offered in order to enhance the understanding of cardiac mechanobiology and foster therapeutic development in myocardial regenerative medicine.
Collapse
Affiliation(s)
- Michael Nguyen-Truong
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
| | - Yan Vivian Li
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
- Department of Design and Merchandising, Colorado State University, Fort Collins, CO 80523, USA
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO 80523, USA
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
33
|
Gaggi G, Di Credico A, Izzicupo P, Sancilio S, Di Mauro M, Iannetti G, Dolci S, Amabile G, Di Baldassarre A, Ghinassi B. Decellularized Extracellular Matrices and Cardiac Differentiation: Study on Human Amniotic Fluid-Stem Cells. Int J Mol Sci 2020; 21:E6317. [PMID: 32878275 PMCID: PMC7504221 DOI: 10.3390/ijms21176317] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Cell therapy with a variety of stem populations is increasingly being investigated as a promising regenerative strategy for cardiovascular (CV) diseases. Their combination with adequate scaffolds represents an improved therapeutic approach. Recently, several biomaterials were investigated as scaffolds for CV tissue repair, with decellularized extracellular matrices (dECMs) arousing increasing interest for cardiac tissue engineering applications. The aim of this study was to analyze whether dECMs support the cardiac differentiation of CardiopoieticAF stem cells. These perinatal stem cells, which can be easily isolated without ethical or safety limitations, display a high cardiac differentiative potential. Differentiation was previously achieved by culturing them on Matrigel, but this 3D scaffold is not transplantable. The identification of a new transplantable scaffold able to support CardiopoieticAF stem cell cardiac differentiation is pivotal prior to encouraging translation of in vitro studies in animal model preclinical investigations. Our data demonstrated that decellularized extracellular matrices already used in cardiac surgery (the porcine CorTMPATCH and the equine MatrixPatchTM) can efficiently support the proliferation and cardiac differentiation of CardiopoieticAF stem cells and represent a useful cellular scaffold to be transplanted with stem cells in animal hosts.
Collapse
Affiliation(s)
- Giulia Gaggi
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Andrea Di Credico
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Pascal Izzicupo
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Silvia Sancilio
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), 6202 Maastricht, The Netherlands;
| | | | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | | | - Angela Di Baldassarre
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Barbara Ghinassi
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| |
Collapse
|
34
|
Karimi Hajishoreh N, Baheiraei N, Naderi N, Salehnia M. Reduced graphene oxide facilitates biocompatibility of alginate for cardiac repair. J BIOACT COMPAT POL 2020. [DOI: 10.1177/0883911520933913] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The benefits of combined cell/material therapy appear promising for myocardial infarction treatment. The safety of alginate, along with its excellent biocompatibility and biodegradability, has been extensively investigated for cardiac tissue engineering. Among graphene-based nanomaterials, reduced graphene oxide has been considered as a promising candidate for cardiac treatment due to its unique physicochemical properties. In this study, the reduced graphene oxide incorporation effect within alginate hydrogels was investigated for cardiac repair application. Reduced graphene oxide reinforced alginate properties, resulting in an increase in gel stiffness. The cytocompatibility of the hydrogels prepared with human bone marrow–derived mesenchymal stem cells was assessed by the 3-(4,5dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide) assay. Following reduced graphene oxide addition, alginate-reduced graphene oxide retained significantly higher cell viability compared to that of alginate and cells cultured on tissue culture plates. Acridine orange/propidium iodide staining was also used to identify both viable and necrotic human bone marrow–derived mesenchymal stem cells within the prepared hydrogels. After a 72-h culture, the percentage of viable cells was twice as much as those cultured on either alginate or tissue culture plate, reaching approximately 80%. Quantitative reverse transcription polymerase chain reaction analysis was performed to assess gene expression of neonatal rat cardiac cells encapsulated on hydrogels for TrpT-2, Conx43, and Actn4 after 7 days. The expression of all genes in alginate-reduced graphene oxide increased significantly compared to that in alginate or tissue culture plate. The results obtained confirmed that the presence of reduced graphene oxide, as an electro-active moiety within alginate, could tune the physicochemical properties of this material, providing a desirable electroactive hydrogel for stem cell therapy in patients with ischemic heart disease.
Collapse
Affiliation(s)
- Negar Karimi Hajishoreh
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nasim Naderi
- Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mojdeh Salehnia
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
35
|
Ramírez WA, Gizzi A, Sack KL, Guccione JM, Hurtado DE. In-silico study of the cardiac arrhythmogenic potential of biomaterial injection therapy. Sci Rep 2020; 10:12990. [PMID: 32737400 PMCID: PMC7395773 DOI: 10.1038/s41598-020-69900-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Biomaterial injection is a novel therapy to treat ischemic heart failure (HF) that has shown to reduce remodeling and restore cardiac function in recent preclinical studies. While the effect of biomaterial injection in reducing mechanical wall stress has been recently demonstrated, the influence of biomaterials on the electrical behavior of treated hearts has not been elucidated. In this work, we developed computational models of swine hearts to study the electrophysiological vulnerability associated with biomaterial injection therapy. The propagation of action potentials on realistic biventricular geometries was simulated by numerically solving the monodomain electrophysiology equations on anatomically-detailed models of normal, HF untreated, and HF treated hearts. Heart geometries were constructed from high-resolution magnetic resonance images (MRI) where the healthy, peri-infarcted, infarcted and gel regions were identified, and the orientation of cardiac fibers was informed from diffusion-tensor MRI. Regional restitution properties in each case were evaluated by constructing a probability density function of the action potential duration (APD) at different cycle lengths. A comparative analysis of the ventricular fibrillation (VF) dynamics for every heart was carried out by measuring the number of filaments formed after wave braking. Our results suggest that biomaterial injection therapy does not affect the regional dispersion of repolarization when comparing untreated and treated failing hearts. Further, we found that the treated failing heart is more prone to sustain VF than the normal heart, and is at least as susceptible to sustained VF as the untreated failing heart. Moreover, we show that the main features of VF dynamics in a treated failing heart are not affected by the level of electrical conductivity of the biogel injectates. This work represents a novel proof-of-concept study demonstrating the feasibility of computer simulations of the heart in understanding the arrhythmic behavior in novel therapies for HF.
Collapse
Affiliation(s)
- William A Ramírez
- Department of Structural and Geotechnical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alessio Gizzi
- Nonlinear Physics and Mathematical Modeling Lab, Department of Engineering, Campus Bio-Medico University of Rome, Rome, Italy
| | - Kevin L Sack
- Department of Surgery, University of California at San Francisco, San Francisco, CA, USA
- Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Julius M Guccione
- Department of Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Daniel E Hurtado
- Department of Structural and Geotechnical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile.
| |
Collapse
|
36
|
Bar A, Cohen S. Inducing Endogenous Cardiac Regeneration: Can Biomaterials Connect the Dots? Front Bioeng Biotechnol 2020; 8:126. [PMID: 32175315 PMCID: PMC7056668 DOI: 10.3389/fbioe.2020.00126] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
Heart failure (HF) after myocardial infarction (MI) due to blockage of coronary arteries is a major public health issue. MI results in massive loss of cardiac muscle due to ischemia. Unfortunately, the adult mammalian myocardium presents a low regenerative potential, leading to two main responses to injury: fibrotic scar formation and hypertrophic remodeling. To date, complete heart transplantation remains the only clinical option to restore heart function. In the last two decades, tissue engineering has emerged as a promising approach to promote cardiac regeneration. Tissue engineering aims to target processes associated with MI, including cardiomyogenesis, modulation of extracellular matrix (ECM) remodeling, and fibrosis. Tissue engineering dogmas suggest the utilization and combination of two key components: bioactive molecules and biomaterials. This chapter will present current therapeutic applications of biomaterials in cardiac regeneration and the challenges still faced ahead. The following biomaterial-based approaches will be discussed: Nano-carriers for cardiac regeneration-inducing biomolecules; corresponding matrices for their controlled release; injectable hydrogels for cell delivery and cardiac patches. The concept of combining cardiac patches with controlled release matrices will be introduced, presenting a promising strategy to promote endogenous cardiac regeneration.
Collapse
Affiliation(s)
- Assaf Bar
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Smadar Cohen
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beersheba, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
37
|
Li W. Biomechanics of infarcted left Ventricle-A review of experiments. J Mech Behav Biomed Mater 2020; 103:103591. [PMID: 32090920 DOI: 10.1016/j.jmbbm.2019.103591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is one of leading diseases to contribute to annual death rate of 5% in the world. In the past decades, significant work has been devoted to this subject. Biomechanics of infarcted left ventricle (LV) is associated with MI diagnosis, understanding of remodelling, MI micro-structure and biomechanical property characterizations as well as MI therapy design and optimization, but the subject has not been reviewed presently. In the article, biomechanics of infarcted LV was reviewed in terms of experiments achieved in the subject so far. The concerned content includes experimental remodelling, kinematics and kinetics of infarcted LVs. A few important issues were discussed and several essential topics that need to be investigated further were summarized. Microstructure of MI tissue should be observed even carefully and compared between different methods for producing MI scar in the same animal model, and eventually correlated to passive biomechanical property by establishing innovative constitutive laws. More uniaxial or biaxial tensile tests are desirable on MI, border and remote tissues, and viscoelastic property identification should be performed in various time scales. Active contraction experiments on LV wall with MI should be conducted to clarify impaired LV pumping function and supply necessary data to the function modelling. Pressure-volume curves of LV with MI during diastole and systole for the human are also desirable to propose and validate constitutive laws for LV walls with MI.
Collapse
Affiliation(s)
- Wenguang Li
- School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
38
|
Yao Y, Ding J, Wang Z, Zhang H, Xie J, Wang Y, Hong L, Mao Z, Gao J, Gao C. ROS-responsive polyurethane fibrous patches loaded with methylprednisolone (MP) for restoring structures and functions of infarcted myocardium in vivo. Biomaterials 2019; 232:119726. [PMID: 31901502 DOI: 10.1016/j.biomaterials.2019.119726] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) play an important role in the pathogenesis of numerous diseases including atherosclerosis, diabetes, inflammation and myocardial infarction (MI). In this study, a ROS-responsive biodegradable elastomeric polyurethane containing thioketal (PUTK) linkages was synthesized from polycaprolactone diol (PCL-diol ), 1,6-hexamethylene diisocyanate (HDI), and ROS-cleavable chain extender. The PUTK was electrospun into fibrous patches with the option to load glucocorticoid methylprednisolone (MP), which were then used to treat MI of rats in vivo. The fibrous patches exhibited suitable mechanical properties and high elasticity. The molecular weight of PUTK was decreased significantly after incubation in 1 mM H2O2 solution for 2 weeks due to the degradation of thioketal bonds on the polymer backbone. Both the PUTK and PUTK/MP fibrous patches showed good antioxidant property in an oxidative environment in vitro. Implantation of the ROS-responsive polyurethane patches in MI of rats in vivo could better protect cardiomyocytes from death in the earlier stage (24 h) than the non ROS-responsive ones. Implantation of the PUTK/MP fibrous patches for 28 days could effectively improve the reconstruction of cardiac functions including increased ejection fraction, decreased infarction size, and enhanced revascularization of the infarct myocardium.
Collapse
Affiliation(s)
- Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhaoyi Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haolan Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Jianqing Gao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
39
|
W. King M, Chen J, Deshpande M, He T, Ramakrishna H, Xie Y, Zhang F, Zhao F. Structural Design, Fabrication and Evaluation of Resorbable Fiber-Based Tissue Engineering Scaffolds. Biotechnol Bioeng 2019. [DOI: 10.5772/intechopen.84643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
40
|
Zhao L. A novel graphene oxide polymer gel platform for cardiac tissue engineering application. 3 Biotech 2019; 9:401. [PMID: 31681522 PMCID: PMC6800416 DOI: 10.1007/s13205-019-1912-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/21/2019] [Indexed: 12/20/2022] Open
Abstract
In this study, we demonstrated a Reverse Thermal Gel (RTG), which is injectable and functionalized with GOs (GO-RTG) that changes at room temperature (24 °C) from a mixture to a three-dimensional (3D) matrix gel soon after approaching its body temperature (37 °C). We also presented investigational evidence, which represents that the system of 3D GO-RTG promotes MCs proliferation as well as alignment, supports in long-standing survival of MCs, and enhances the function of MCs when compared with typical 3D plain RTG system and 2D gelatin control groups. Thus, this system of injectable GO-RTG can be capable of using as a negligibly invasive device for engineering efforts of cardiac tissue.
Collapse
Affiliation(s)
- Li Zhao
- Department of Cardiology, Third Affiliated Hospital of Qiqihar Medical University, NO 27, Taishun Street, Tiefeng District, Qiqihar City, 161000 Heilongjiang Province China
| |
Collapse
|
41
|
Fan C, Shi J, Zhuang Y, Zhang L, Huang L, Yang W, Chen B, Chen Y, Xiao Z, Shen H, Zhao Y, Dai J. Myocardial-Infarction-Responsive Smart Hydrogels Targeting Matrix Metalloproteinase for On-Demand Growth Factor Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902900. [PMID: 31408234 DOI: 10.1002/adma.201902900] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/22/2019] [Indexed: 06/10/2023]
Abstract
Although in situ restoration of blood supply to the infarction region and attenuating pre-existing extracellular matrix degradation remain potential therapeutic approaches for myocardial infarction (MI), local delivery of therapeutics has been limited by low accumulation (inefficacy) and unnecessary diffusion (toxicity). Here, a dual functional MI-responsive hydrogel is fabricated for on-demand drug delivery to promote angiogenesis and inhibit cardiac remodeling by targeting upregulated matrix metalloproteinase-2/9 (MMP-2/9) after MI. A glutathione (GSH)-modified collagen hydrogel (collagen-GSH) is prepared by conjugating collagen amine groups with GSH sulfhydryl groups and the recombinant protein GST-TIMP-bFGF (bFGF: basic fibroblast growth factor) by fusing bFGF with glutathione-S-transferase (GST) and MMP-2/9 cleavable peptide PLGLAG (TIMP). Specific binding between GST and GSH significantly improves the amount of GST-TIMP-bFGF loaded in collagen-GSH hydrogel. The TIMP peptide enclosed between GST and bFGF responds to MMPs for on-demand release during MI. Additionally, the TIMP peptide is a competitive substrate of MMPs that inhibits the excessive degradation of cardiac matrix by MMPs after MI. GST-TIMP-bFGF/collagen-GSH hydrogels promote the recovery of MI rats by enhancing vascularization and ameliorating myocardium remodeling. The results suggest that on-demand growth factor delivery by synchronously controlling binding and responsive release to promote angiogenesis and attenuate cardiac remodeling might be promising for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Caixia Fan
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiajia Shi
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lulu Zhang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lei Huang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Bing Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
42
|
Dolan EB, Hofmann B, de Vaal MH, Bellavia G, Straino S, Kovarova L, Pravda M, Velebny V, Daro D, Braun N, Monahan DS, Levey RE, O'Neill H, Hinderer S, Greensmith R, Monaghan MG, Schenke-Layland K, Dockery P, Murphy BP, Kelly HM, Wildhirt S, Duffy GP. A bioresorbable biomaterial carrier and passive stabilization device to improve heart function post-myocardial infarction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109751. [DOI: 10.1016/j.msec.2019.109751] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
|
43
|
Yang H, Qin X, Wang H, Zhao X, Liu Y, Wo HT, Liu C, Nishiga M, Chen H, Ge J, Sayed N, Abilez OJ, Ding D, Heilshorn SC, Li K. An in Vivo miRNA Delivery System for Restoring Infarcted Myocardium. ACS NANO 2019; 13:9880-9894. [PMID: 31149806 PMCID: PMC7930012 DOI: 10.1021/acsnano.9b03343] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
A major challenge in myocardial infarction (MI)-related heart failure treatment using microRNA is the efficient and sustainable delivery of miRNAs into myocardium to achieve functional improvement through stimulation of intrinsic myocardial restoration. In this study, we established an in vivo delivery system using polymeric nanoparticles to carry miRNA (miNPs) for localized delivery within a shear-thinning injectable hydrogel. The miNPs triggered proliferation of human embryonic stem cell-derived cardiomyocytes and endothelial cells (hESC-CMs and hESC-ECs) and promoted angiogenesis in hypoxic conditions, showing significantly lower cytotoxicity than Lipofectamine. Furthermore, one injected dose of hydrogel/miNP in MI rats demonstrated significantly improved cardiac functions: increased ejection fraction from 45% to 64%, reduced scar size from 20% to 10%, and doubled capillary density in the border zone compared to the control group at 4 weeks. As such, our results indicate that this injectable hydrogel/miNP composite can deliver miRNA to restore injured myocardium efficiently and safely.
Collapse
Affiliation(s)
- Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
- Corresponding Authors.,
| | - Xulei Qin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Huiyuan Wang
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Xin Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Yonggang Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Hung-Ta Wo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Haodong Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jing Ge
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Oscar J. Abilez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Kai Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
- Corresponding Authors.,
| |
Collapse
|
44
|
Fan Y, Ronan W, Teh I, Schneider JE, Varela CE, Whyte W, McHugh P, Leen S, Roche E. A comparison of two quasi-static computational models for assessment of intra-myocardial injection as a therapeutic strategy for heart failure. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2019; 35:e3213. [PMID: 31062508 DOI: 10.1002/cnm.3213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 06/09/2023]
Abstract
Myocardial infarction, or heart attack, is the leading cause of mortality globally. Although the treatment of myocardial infarct has improved significantly, scar tissue that persists can often lead to increased stress and adverse remodeling of surrounding tissue and ultimately to heart failure. Intra-myocardial injection of biomaterials represents a potential treatment to attenuate remodeling, mitigate degeneration, and reverse the disease process in the tissue. In vivo experiments on animal models have shown functional benefits of this therapeutic strategy. However, a poor understanding of the optimal injection pattern, volume, and material properties has acted as a barrier to its widespread clinical adoption. In this study, we developed two quasistatic finite element simulations of the left ventricle to investigate the mechanical effect of intra-myocardial injection. The first model employed an idealized left ventricular geometry with rule-based cardiomyocyte orientation. The second model employed a subject-specific left ventricular geometry with cardiomyocyte orientation from diffusion tensor magnetic resonance imaging. Both models predicted cardiac parameters including ejection fraction, systolic wall thickening, and ventricular twist that matched experimentally reported values. All injection simulations showed cardiomyocyte stress attenuation, offering an explanation for the mechanical reinforcement benefit associated with injection. The study also enabled a comparison of injection location and the corresponding effect on cardiac performance at different stages of the cardiac cycle. While the idealized model has lower fidelity, it predicts cardiac function and differentiates the effects of injection location. Both models represent versatile in silico tools to guide optimal strategy in terms of injection number, volume, site, and material properties.
Collapse
Affiliation(s)
- Yiling Fan
- Mechanical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William Ronan
- Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Irvin Teh
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jurgen E Schneider
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Claudia E Varela
- Institute for Medical Engineering Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, Massachusetts
| | - William Whyte
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, 2, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin, 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI, NUIG & TCD, Dublin, 2, Ireland
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| | - Peter McHugh
- Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Sean Leen
- Mechanical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Ellen Roche
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Institute for Medical Engineering Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
45
|
Chan BT, Ahmad Bakir A, Al Abed A, Dokos S, Leong CN, Ooi EH, Lim R, Lim E. Impact of myocardial infarction on intraventricular vortex and flow energetics assessed using computational simulations. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2019; 35:e3204. [PMID: 30912313 DOI: 10.1002/cnm.3204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 06/09/2023]
Abstract
Flow energetics have been proposed as early indicators of progressive left ventricular (LV) functional impairment in patients with myocardial infarction (MI), but its correlation with individual MI parameters has not been fully explored. Using electro-fluid-structure interaction LV models, this study investigated the correlation between four MI parameters: infarct size, infarct multiplicity, regional enhancement of contractility at the viable myocardium area (RECVM), and LV mechanical dyssynchrony (LVMD) with intraventricular vortex and flow energetics. In LV with small infarcts, our results showed that infarct appearance amplified the energy dissipation index (DI), where substantial viscous energy loss was observed in areas with high flow velocity and near the infarct-vortex interface. The LV with small multiple infarcts and RECVM showed remarkable DI increment during systole and diastole. In correlation analysis, the systolic kinetic energy fluctuation index (E') was positively related to ejection fraction (EF) (R2 = 0.982) but negatively correlated with diastolic E' (R2 = 0.970). Diastolic E' was inversely correlated with vortex kinetic energy (R2 = 0.960) and vortex depth (R2 = 0.876). We showed an excessive systolic DI could differentiate infarcted LV with normal EF from healthy LV. Strong flow acceleration, LVMD, and vortex-infarct interactions were predominant factors that induced excessive DI in infarcted LVs. Instead of causing undesired flow turbulence, high systolic E' suggested the existence of energetic flow acceleration, while high diastolic E' implied an inefficient diastolic filling. Thus, systolic E' is not a suitable early indicator for progressive LV dysfunction in MI patients, while diastolic E' may be a useful index to indicate diastolic impairment in these patients.
Collapse
Affiliation(s)
- Bee Ting Chan
- Department of Mechanical Engineering, Faculty of Engineering, Technology & Built Environment, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Azam Ahmad Bakir
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Amr Al Abed
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Socrates Dokos
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Chin Neng Leong
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ean Hin Ooi
- School of Engineering, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia
| | - Renly Lim
- Quality Use of Medicines and Pharmacy Research Centre, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, 5001, Australia
| | - Einly Lim
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Peña B, Maldonado M, Bonham AJ, Aguado BA, Dominguez-Alfaro A, Laughter M, Rowland TJ, Bardill J, Farnsworth NL, Ramon NA, Taylor MRG, Anseth KS, Prato M, Shandas R, McKinsey TA, Park D, Mestroni L. Gold Nanoparticle-Functionalized Reverse Thermal Gel for Tissue Engineering Applications. ACS APPLIED MATERIALS & INTERFACES 2019; 11:18671-18680. [PMID: 31021594 PMCID: PMC6764451 DOI: 10.1021/acsami.9b00666] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Utilizing polymers in cardiac tissue engineering holds promise for restoring function to the heart following myocardial infarction, which is associated with grave morbidity and mortality. To properly mimic native cardiac tissue, materials must not only support cardiac cell growth but also have inherent conductive properties. Here, we present an injectable reverse thermal gel (RTG)-based cardiac cell scaffold system that is both biocompatible and conductive. Following the synthesis of a highly functionalizable, biomimetic RTG backbone, gold nanoparticles (AuNPs) were chemically conjugated to the backbone to enhance the system's conductivity. The resulting RTG-AuNP hydrogel supported targeted survival of neonatal rat ventricular myocytes (NRVMs) for up to 21 days when cocultured with cardiac fibroblasts, leading to an increase in connexin 43 (Cx43) relative to control cultures (NRVMs cultured on traditional gelatin-coated dishes and RTG hydrogel without AuNPs). This biomimetic and conductive RTG-AuNP hydrogel holds promise for future cardiac tissue engineering applications.
Collapse
Affiliation(s)
- Brisa Peña
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Marcos Maldonado
- Department of Chemistry and Biochemistry, Metropolitan State University of Denver, 1201 5th Street, Denver, Colorado 80206, United States
| | - Andrew J. Bonham
- Department of Chemistry and Biochemistry, Metropolitan State University of Denver, 1201 5th Street, Denver, Colorado 80206, United States
| | - Brian A. Aguado
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
| | - Antonio Dominguez-Alfaro
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
- POLYMAT, University of the Basque Country UPV/EHU, Avenida de Tolosa 72, 20018 Donostia-San Sebastian, Spain
| | - Melissa Laughter
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Teisha J. Rowland
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - James Bardill
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Nikki L. Farnsworth
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Ct., Bldg. M20, Aurora, Colorado 80045, United States
| | - Nuria Alegret Ramon
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- POLYMAT, University of the Basque Country UPV/EHU, Avenida de Tolosa 72, 20018 Donostia-San Sebastian, Spain
| | - Matthew R. G. Taylor
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
| | - Maurizio Prato
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Giorgieri 1, Trieste 34127, Italy
- Basque Fdn Sci, Ikerbasque, Bilbao 48013, Spain
| | - Robin Shandas
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Daewon Park
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Luisa Mestroni
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| |
Collapse
|
47
|
Yang Y, Lei D, Huang S, Yang Q, Song B, Guo Y, Shen A, Yuan Z, Li S, Qing F, Ye X, You Z, Zhao Q. Elastic 3D-Printed Hybrid Polymeric Scaffold Improves Cardiac Remodeling after Myocardial Infarction. Adv Healthc Mater 2019; 8:e1900065. [PMID: 30941925 DOI: 10.1002/adhm.201900065] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/22/2019] [Indexed: 01/12/2023]
Abstract
Myocardial remodeling, including ventricular dilation and wall thinning, is an important pathological process caused by myocardial infarction (MI). To intervene in this pathological process, a new type of cardiac scaffold composed of a thermoset (poly-[glycerol sebacate], PGS) and a thermoplastic (poly-[ε-caprolactone], PCL) is directly printed by employing fused deposition modeling 3D-printing technology. The PGS-PCL scaffold possesses stacked construction with regular crisscrossed filaments and interconnected micropores and exhibits superior mechanical properties. In vitro studies demonstrate favorable biodegradability and biocompatibility of the PGS-PCL scaffold. When implanted onto the infarcted myocardium, this scaffold improves and preserves heart function. Furthermore, the scaffold improves several vital aspects of myocardial remodeling. On the morphological level, the scaffold reduces ventricular wall thinning and attenuated infarct size, and on the cellular level, it enhances vascular density and increases M2 macrophage infiltration, which might further contribute to the mitigated myocardial apoptosis rate. Moreover, the flexible PGS-PCL scaffold can be tailored to any desired shape, showing promise for annular-shaped restraint device application and meeting the demands for minimal invasive operation. Overall, this study demonstrates the therapeutic effects and versatile applications of a novel 3D-printed, biodegradable and biocompatible cardiac scaffold, which represents a promising strategy for improving myocardial remodeling after MI.
Collapse
Affiliation(s)
- Yang Yang
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| | - Dong Lei
- College of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| | - Shixing Huang
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| | - Qi Yang
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| | - Benyan Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of Materials Science and EngineeringDonghua University Shanghai 201620 P. R. China
| | - Yifan Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of Materials Science and EngineeringDonghua University Shanghai 201620 P. R. China
| | - Ao Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of Materials Science and EngineeringDonghua University Shanghai 201620 P. R. China
| | - Zhize Yuan
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| | - Sen Li
- Department of Vascular SurgeryThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang University School of Medicine Zhejiang 310009 P. R. China
| | - Feng‐Ling Qing
- College of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| | - Xiaofeng Ye
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of Materials Science and EngineeringDonghua University Shanghai 201620 P. R. China
| | - Qiang Zhao
- Department of Cardiac SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine Shanghai 200025 P. R. China
| |
Collapse
|
48
|
Cheng Y, Liu DZ, Zhang CX, Cui H, Liu M, Zhang BL, Mei QB, Lu ZF, Zhou SY. Mitochondria-targeted antioxidant delivery for precise treatment of myocardial ischemia–reperfusion injury through a multistage continuous targeted strategy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 16:236-249. [DOI: 10.1016/j.nano.2018.12.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/29/2018] [Accepted: 12/28/2018] [Indexed: 02/06/2023]
|
49
|
Lee DJ, Cavasin MA, Rocker AJ, Soranno DE, Meng X, Shandas R, Park D. An injectable sulfonated reversible thermal gel for therapeutic angiogenesis to protect cardiac function after a myocardial infarction. J Biol Eng 2019; 13:6. [PMID: 30675179 PMCID: PMC6337754 DOI: 10.1186/s13036-019-0142-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/07/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Cardiovascular disease and myocardial infarction are associated with high mortality and morbidity and a more effective treatment remains a major clinical need. The intramyocardial injection of biomaterials has been investigated as a potential treatment for heart failure by providing mechanical support to the myocardium and reducing stress on cardiomyocytes. Another treatment approach that has been explored is therapeutic angiogenesis that requires careful spatiotemporal control of angiogenic drug delivery. An injectable sulfonated reversible thermal gel composed of a polyurea conjugated with poly(N-isopropylacrylamide) and sulfonate groups has been developed for intramyocardial injection with angiogenic factors for the protection of cardiac function after a myocardial infarction. RESULTS The thermal gel allowed for the sustained, localized release of VEGF in vivo with intramyocardial injection after two weeks. A myocardial infarction reperfusion injury model was used to evaluate therapeutic benefits to cardiac function and vascularization. Echocardiography presented improved cardiac function, infarct size and ventricular wall thinning were reduced, and immunohistochemistry showed improved vascularization with thermal gel injections. The thermal gel alone showed cardioprotective and vascularization properties, and slightly improved further with the additional delivery of VEGF. An inflammatory response evaluation demonstrated the infiltration of macrophages due to the myocardial infarction was more significant compared to the foreign body inflammatory response to the thermal gel. Detecting DNA fragments of apoptotic cells also demonstrated potential anti-apoptotic effects of the thermal gel. CONCLUSION The intramyocardial injection of the sulfonated reversible thermal gel has cardioprotective and vascularization properties for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- David J. Lee
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Maria A. Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Adam J. Rocker
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Danielle E. Soranno
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Robin Shandas
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045 USA
| |
Collapse
|
50
|
Curley CJ, Dolan EB, Otten M, Hinderer S, Duffy GP, Murphy BP. An injectable alginate/extra cellular matrix (ECM) hydrogel towards acellular treatment of heart failure. Drug Deliv Transl Res 2018; 9:1-13. [DOI: 10.1007/s13346-018-00601-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|