1
|
Mu L, Dong R, Li C, Chen J, Huang Y, Li T, Guo B. ROS responsive conductive microspheres loaded with salvianolic acid B as adipose derived stem cell carriers for acute myocardial infarction treatment. Biomaterials 2025; 314:122849. [PMID: 39357150 DOI: 10.1016/j.biomaterials.2024.122849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Stem cell therapy is currently the most promising strategy for the treatment of myocardial infarction. However, the development of injectable cell carriers that can scavenge reactive oxygen species (ROS) in the infarct zone to improve transplanted cell survival remains a challenge. Here, we developed a ROS responsive conductive microsphere based on chitosan (CS) and dextran (DEX) with 4-formylphenylboronic acid (4-FPBA) as a cross-linking agent and the addition of graphite oxide (GO) and the anti-inflammatory agent salvianolic acid B (SalB), as a cell delivery carrier for myocardial infarction. These microspheres were crosslinked by dual dynamic networks of Schiff base and phenylborate bonds. The relationship between CS concentration and microsphere particle size, as well as the biocompatibility, ROS responsiveness, anti-inflammatory properties, and effects on myogenic differentiation of H9C2 cells were fully investigated. The microspheres exhibit good biocompatibility, proliferation promoting, differentiation promoting, antioxidant, and anti-inflammatory properties. When applied to mice myocardial infarction models, the ROS responsive conductive microspheres loaded with SalB and adipose derived stem cells (ADSC) exhibited excellent in vivo repair ability. In addition, they reduced myocardial fibrosis and promoted ventricular wall regeneration by promoting the expression of Connexin 43 (Cx43) and CD31, ultimately reshaping the infarcted myocardium, suggesting their great potential as cell delivery carriers for myocardial infarction treatment.
Collapse
Affiliation(s)
- Lei Mu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ruonan Dong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, China
| | - Jiangwei Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Medical Rehabilitation, School of Stomatology, the Fourth Military Medical University, Xi'an, 710032, China
| | - Ying Huang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Tongyang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Baolin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
2
|
Nam M, Lee JW, Cha GD. Biomedical Application of Enzymatically Crosslinked Injectable Hydrogels. Gels 2024; 10:640. [PMID: 39451293 PMCID: PMC11507637 DOI: 10.3390/gels10100640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant interest in the biomedical field owing to their tissue-like properties and capability to incorporate various fillers. Among these, injectable hydrogels have been highlighted for their unique advantages, especially their minimally invasive administration mode for implantable use. These injectable hydrogels can be utilized in their pristine forms or as composites by integrating them with therapeutic filler materials. Given their primary application in implantable platforms, enzymatically crosslinked injectable hydrogels have been actively explored due to their excellent biocompatibility and easily controllable mechanical properties for the desired use. This review introduces the crosslinking mechanisms of such hydrogels, focusing on those mediated by horseradish peroxidase (HRP), transglutaminase (TG), and tyrosinase. Furthermore, several parameters and their relationships with the intrinsic properties of hydrogels are investigated. Subsequently, the representative biomedical applications of enzymatically crosslinked-injectable hydrogels are presented, including those for wound healing, preventing post-operative adhesion (POA), and hemostasis. Furthermore, hydrogel composites containing filler materials, such as therapeutic cells, proteins, and drugs, are analyzed. In conclusion, we examine the scientific challenges and directions for future developments in the field of enzymatically crosslinked-injectable hydrogels, focusing on material selection, intrinsic properties, and filler integration.
Collapse
Affiliation(s)
| | | | - Gi Doo Cha
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea; (M.N.); (J.W.L.)
| |
Collapse
|
3
|
Zhang Z, Tang R, Liu X, Liang G, Sun X. Recent Advances in Self-Assembling Peptide-Based Nanomaterials for Enhanced Photodynamic Therapy. Macromol Biosci 2024:e2400409. [PMID: 39360584 DOI: 10.1002/mabi.202400409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Self-assembling peptide-based materials with ordered nanostructures possess advantages such as good biocompatibility and biodegradability, superior controllability, and ease of chemical modification. Through covalent conjugation or non-covalent encapsulation, photosensitizers (PSs) can be carried by self-assembling peptide-based nanomaterials for targeted delivery towards tumor tissues. This improves the stability, solubility, and tumor accumulation of PSs, as well as reduces their dark toxicity. More importantly, these nanomaterials can be tailored with responsiveness to tumor microenvironment, which enables smart release of PSs for precise and enhanced photodynamic therapy (PDT). In this review, the self-assembly of peptide from the perspective of driving forces is first described, and various self-assembling peptide materials with zero to 3D nanostructures are subsequently highlighted for PDT of cancers in recent years. Finally, an outlook in this field is provided to motivate fabrication of advanced PDT nanomaterials.
Collapse
Affiliation(s)
- Ziyi Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Runqun Tang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
- Handan Norman Technology Co., Ltd, Guantao, 057750, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| |
Collapse
|
4
|
Luo Q, Li Z, Liu B, Ding J. Hydrogel formulations for orthotopic treatment of myocardial infarction. Expert Opin Drug Deliv 2024; 21:1463-1478. [PMID: 39323051 DOI: 10.1080/17425247.2024.2409906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/25/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Myocardial infarction (MI) causes extensive structural and functional damage to the cardiac tissue due to the significant loss of cardiomyocytes. Early reperfusion is the standard treatment strategy for acute MI, but it is associated with adverse effects. Additionally, current therapies to alleviate pathological changes post-MI are not effective. Subsequent pathological remodeling of the damaged myocardium often results in heart failure. Oral drugs aimed at reducing myocardial damage and remodeling require repeated administration of high doses to maintain therapeutic levels. This compromises efficacy and patient adherence and may cause adverse effects, such as hypotension and liver and/or kidney dysfunction. Hydrogels have emerged as an effective delivery platform for orthotopic treatment of MI due to their high water content and excellent tissue compatibility. AREA COVERED Hydrogels create an optimal microenvironment for delivering drugs, proteins, and cells, preserving their efficacy and increasing their bioavailability. Current research focuses on discovering functional hydrogels for mitigating myocardial damage and regulating repair processes in MI treatment. EXPERT OPINION Hydrogels offer a promising approach in enhancing cardiac repair and improving patient outcomes post-MI. Advancements in hydrogel technology are poised to transform MI therapy, paving the way for personalized treatment strategies and enhanced recovery.
Collapse
Affiliation(s)
- Qiang Luo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P. R. China
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, P. R. China
| |
Collapse
|
5
|
Al Musaimi O, Ng KW, Gavva V, Mercado-Valenzo OM, Haroon HB, Williams DR. Elastin-Derived Peptide-Based Hydrogels as a Potential Drug Delivery System. Gels 2024; 10:531. [PMID: 39195060 DOI: 10.3390/gels10080531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
A peptide-based hydrogel sequence was computationally predicted from the Ala-rich cross-linked domains of elastin. Three candidate peptides were subsequently synthesised and characterised as potential drug delivery vehicles. The elastin-derived peptides are Fmoc-FFAAAAKAA-NH2, Fmoc-FFAAAKAA-NH2, and Fmoc-FFAAAKAAA-NH2. All three peptide sequences were able to self-assemble into nanofibers. However, only the first two could form hydrogels, which are preferred as delivery systems compared to solutions. Both of these peptides also exhibited favourable nanofiber lengths of at least 1.86 and 4.57 µm, respectively, which are beneficial for the successful delivery and stability of drugs. The shorter fibre lengths of the third peptide (maximum 0.649 µm) could have inhibited their self-assembly into the three-dimensional networks crucial to hydrogel formation.
Collapse
Affiliation(s)
- Othman Al Musaimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Keng Wooi Ng
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Varshitha Gavva
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| | | | - Hajira Banu Haroon
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Daryl R Williams
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
6
|
Lv Q, Lin J, Huang H, Ma B, Li W, Chen J, Wang M, Wang X, Fu G, Xiao Y. Nanosponge for Iron Chelation and Efflux: A Ferroptosis-Inhibiting Approach for Myocardial Infarction Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305895. [PMID: 38671590 PMCID: PMC11220697 DOI: 10.1002/advs.202305895] [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: 08/22/2023] [Revised: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Myocardial infarction (MI), a consequence of coronary artery occlusion, triggers the degradation of ferritin, resulting in elevated levels of free iron in the heart and thereby inducing ferroptosis. Targeting myocardial ferroptosis through the chelation of excess iron has therapeutic potential for MI treatment. However, iron chelation in post ischemic injury areas using conventional iron-specific chelators is hindered by ineffective myocardial intracellular chelation, rapid clearance, and high systemic toxicity. A chitosan-desferrioxamine nanosponge (CDNS) is designed by co-crosslinking chitosan and deferoxamine through noncovalent gelation to address these challenges. This architecture facilitates direct iron chelation regardless of deferoxamine (DFO) release due to its sponge-like porous hydrogel structure. Upon cellular internalization, CDNS can effectively chelate cellular iron and facilitate the efflux of captured iron, thereby inhibiting ferroptosis and associated oxidative stress and lipid peroxidation. In MI mouse models, myocardial injection of CDNS promotes sustainable retention and the suppression of ferroptosis in the infarcted heart. This intervention improves cardiac function and alleviates adverse cardiac remodeling post-MI, leading to decreased oxidative stress and the promotion of angiogenesis due to ferroptosis inhibition by CDNS in the infarcted heart. This study reveals a nanosponge-based nanomedicine targeting myocardial ferroptosis with efficient iron chelation and efflux, offering a promising MI treatment.
Collapse
Affiliation(s)
- Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
- Department of Cardiovascular SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Boxuan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Wujiao Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced StudiesZhejiang UniversityHangzhou310058China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| |
Collapse
|
7
|
Song L, Jia K, Yang F, Wang J. Advanced Nanomedicine Approaches for Myocardial Infarction Treatment. Int J Nanomedicine 2024; 19:6399-6425. [PMID: 38952676 PMCID: PMC11215519 DOI: 10.2147/ijn.s467219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Abstract
Myocardial infarction, usually caused by the rupture of atherosclerotic plaque, leads to irreversible ischemic cardiomyocyte death within hours followed by impaired cardiac performance or even heart failure. Current interventional reperfusion strategies for myocardial infarction still face high mortality with the development of heart failure. Nanomaterial-based therapy has made great progress in reducing infarct size and promoting cardiac repair after MI, although most studies are preclinical trials. This review focuses primarily on recent progress (2016-now) in the development of various nanomedicines in the treatment of myocardial infarction. We summarize these applications with the strategy of mechanism including anti-cardiomyocyte death strategy, activation of neovascularization, antioxidants strategy, immunomodulation, anti-cardiac remodeling, and cardiac repair.
Collapse
Affiliation(s)
- Lin Song
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Kangwei Jia
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Fuqing Yang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
8
|
Gao M, Cai Q, Bian Y, Wang Z, Xu L, Peng J. Protective effect of esculentoside A against myocardial infarction via targeting C-X-C motif chemokine receptor 2. Biomed Pharmacother 2024; 174:116529. [PMID: 38569275 DOI: 10.1016/j.biopha.2024.116529] [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: 01/28/2024] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024] Open
Abstract
Myocardial infarction (MI) is the primary cause of cardiac mortality. Esculentoside A (EsA), a triterpenoid saponin, has anti-inflammatory and antioxidant activities. However, its effect on MI remains unknown. In this study, the protective effect and mechanisms of EsA against MI were investigated. EsA significantly alleviated hypoxia-induced HL-1 cell injury, including increasing cell viability, inhibiting reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) and lactate dehydrogenase (LDH) leakage. In mouse MI model by left coronary artery (LAD) ligating, EsA obviously restored serum levels of creatine kinase isoenzymes (CK-MB), cardiac troponin I (cTnI), superoxide dismutase (SOD) and malondialdehyde (MDA). In addition, the cardioprotective effect of EsA was further confirmed by infarct size, electrocardiogram and echocardiography. Mechanistically, the targeted binding relationship between EsA and C-X-C motif chemokine receptor 2 (CXCR2) was predicted by molecular docking and dynamics, and validated by small molecule pull-down and surface plasmon resonance tests. EsA inhibited CXCR2 level both in vitro and in vivo, correspondingly alleviated oxidative stress by suppressing NOX1 and NOX2 and relieved inflammation through inhibiting p65 and p-p65. It demonstrated that EsA could play a cardioprotective role by targeting CXCR2. However, the effect of EsA against MI was abolished in combination with CXCR2 overexpression both in vitro and in vivo. This study revealed that EsA showed excellent cardioprotective activities by targeting CXCR2 to alleviate oxidative stress and inflammation in MI. EsA may function as a novel CXCR2 inhibitor and a potent candidate for the prevention and intervention of MI in the future.
Collapse
Affiliation(s)
- Meng Gao
- Institute of Intergrative Medicine, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Qing Cai
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yehua Bian
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Zhuoya Wang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Department of Traditional Chinese Medicine Pharmacology, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Hubei Shizhen Laboratory, Wuhan 430065, China.
| |
Collapse
|
9
|
Hua Y, He Z, Ni Y, Sun L, Wang R, Li Y, Li X, Jiang G. Silk fibroin and hydroxypropyl cellulose composite injectable hydrogel-containing extracellular vesicles for myocardial infarction repair. Biomed Phys Eng Express 2024; 10:045001. [PMID: 38640908 DOI: 10.1088/2057-1976/ad40b2] [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: 12/16/2023] [Accepted: 04/19/2024] [Indexed: 04/21/2024]
Abstract
Extracellular vesicles (EVs) have been recognized as one of the promising specific drugs for myocardial infarction (MI) prognosis. Nevertheless, low intramyocardial retention of EVs remains a major impediment to their clinical application. In this study, we developed a silk fibroin/hydroxypropyl cellulose (SF/HPC) composite hydrogel combined with AC16 cell-derived EVs targeted modification by folic acid for the treatment of acute myocardial infarction repair. EVs were functionalized by distearoylphosphatidyl ethanolamine-polyethylene glycol (DSPE-PEG-FA) via noncovalent interaction for targeting and accelerating myocardial infarction repair.In vitro, cytocompatibility analyses revealed that the as-prepared hydrogels had excellent cell viability by MTT assay and the functionalized EVs had higher cell migration by scratch assay.In vivo, the composite hydrogels can promote myocardial tissue repair effects by delaying the process of myocardial fibrosis and promoting angiogenesis of infarct area in MI rat model.
Collapse
Affiliation(s)
- Yinjian Hua
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Zhengfei He
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Yunjie Ni
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Linggang Sun
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Rui Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Yan Li
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Xintong Li
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, People's Republic of China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| |
Collapse
|
10
|
Zhou G, Xu R, Groth T, Wang Y, Yuan X, Ye H, Dou X. The Combination of Bioactive Herbal Compounds with Biomaterials for Regenerative Medicine. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38481114 DOI: 10.1089/ten.teb.2024.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Regenerative medicine aims to restore the function of diseased or damaged tissues and organs by cell therapy, gene therapy, and tissue engineering, along with the adjunctive application of bioactive molecules. Traditional bioactive molecules, such as growth factors and cytokines, have shown great potential in the regulation of cellular and tissue behavior, but have the disadvantages of limited source, high cost, short half-life, and side effects. In recent years, herbal compounds extracted from natural plants/herbs have gained increasing attention. This is not only because herbal compounds are easily obtained, inexpensive, mostly safe, and reliable, but also owing to their excellent effects, including anti-inflammatory, antibacterial, antioxidative, proangiogenic behavior and ability to promote stem cell differentiation. Such effects also play important roles in the processes related to tissue regeneration. Furthermore, the moieties of the herbal compounds can form physical or chemical bonds with the scaffolds, which contributes to improved mechanical strength and stability of the scaffolds. Thus, the incorporation of herbal compounds as bioactive molecules in biomaterials is a promising direction for future regenerative medicine applications. Herein, an overview on the use of bioactive herbal compounds combined with different biomaterial scaffolds for regenerative medicine application is presented. We first introduce the classification, structures, and properties of different herbal bioactive components and then provide a comprehensive survey on the use of bioactive herbal compounds to engineer scaffolds for tissue repair/regeneration of skin, cartilage, bone, neural, and heart tissues. Finally, we highlight the challenges and prospects for the future development of herbal scaffolds toward clinical translation. Overall, it is believed that the combination of bioactive herbal compounds with biomaterials could be a promising perspective for the next generation of regenerative medicine.
Collapse
Affiliation(s)
- Guoying Zhou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruojiao Xu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingyu Yuan
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hua Ye
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
- Oxford Suzhou Centre for Advanced Research, University of Oxford, Suzhou, China
| | - Xiaobing Dou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Zhou G, Cao Y, Yan Y, Xu H, Zhang X, Yan T, Wan H. Injectable Hydrogels Based on Hyaluronic Acid and Gelatin Combined with Salvianolic Acid B and Vascular Endothelial Growth Factor for Treatment of Traumatic Brain Injury in Mice. Molecules 2024; 29:1705. [PMID: 38675525 PMCID: PMC11052029 DOI: 10.3390/molecules29081705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Traumatic brain injury (TBI) leads to structural damage in the brain, and is one of the major causes of disability and death in the world. Herein, we developed a composite injectable hydrogel (HA/Gel) composed of hyaluronic acid (HA) and gelatin (Gel), loaded with vascular endothelial growth factor (VEGF) and salvianolic acid B (SAB) for treatment of TBI. The HA/Gel hydrogels were formed by the coupling of phenol-rich tyramine-modified HA (HA-TA) and tyramine-modified Gel (Gel-TA) catalyzed by horseradish peroxidase (HRP) in the presence of hydrogen peroxide (H2O2). SEM results showed that HA/Gel hydrogel had a porous structure. Rheological test results showed that the hydrogel possessed appropriate rheological properties, and UV spectrophotometry results showed that the hydrogel exhibited excellent SAB release performance. The results of LIVE/DEAD staining, CCK-8 and Phalloidin/DAPI fluorescence staining showed that the HA/Gel hydrogel possessed good cell biocompatibility. Moreover, the hydrogels loaded with SAB and VEGF (HA/Gel/SAB/VEGF) could effectively promote the proliferation of bone marrow mesenchymal stem cells (BMSCs). In addition, the results of H&E staining, CD31 and α-SMA immunofluorescence staining showed that the HA/Gel/SAB/VEGF hydrogel possessed good in vivo biocompatibility and pro-angiogenic ability. Furthermore, immunohistochemical results showed that the injection of HA/Gel/SAB/VEGF hydrogel to the injury site could effectively reduce the volume of defective tissues in traumatic brain injured mice. Our results suggest that the injection of HA/Gel hydrogel loaded with SAB and VEGF might provide a new approach for therapeutic brain tissue repair after traumatic brain injury.
Collapse
Affiliation(s)
- Guoying Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Yajie Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Yujia Yan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Haibo Xu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Xiao Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Tingzi Yan
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Haitong Wan
- Institute of Cardio-Cerebrovascular Disease, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
12
|
Li Z, Song P, Li G, Han Y, Ren X, Bai L, Su J. AI energized hydrogel design, optimization and application in biomedicine. Mater Today Bio 2024; 25:101014. [PMID: 38464497 PMCID: PMC10924066 DOI: 10.1016/j.mtbio.2024.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
Traditional hydrogel design and optimization methods usually rely on repeated experiments, which is time-consuming and expensive, resulting in a slow-moving of advanced hydrogel development. With the rapid development of artificial intelligence (AI) technology and increasing material data, AI-energized design and optimization of hydrogels for biomedical applications has emerged as a revolutionary breakthrough in materials science. This review begins by outlining the history of AI and the potential advantages of using AI in the design and optimization of hydrogels, such as prediction and optimization of properties, multi-attribute optimization, high-throughput screening, automated material discovery, optimizing experimental design, and etc. Then, we focus on the various applications of hydrogels supported by AI technology in biomedicine, including drug delivery, bio-inks for advanced manufacturing, tissue repair, and biosensors, so as to provide a clear and comprehensive understanding of researchers in this field. Finally, we discuss the future directions and prospects, and provide a new perspective for the research and development of novel hydrogel materials for biomedical applications.
Collapse
Affiliation(s)
- Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yafei Han
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiaoxiang Ren
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
13
|
Han XS, Li PC, Song HT, Chen YM, Li JH, Yang Y, Li HP, Miyatake H, Ito Y. Mussel inspired sequential protein delivery based on self-healing injectable nanocomposite hydrogel. Int J Biol Macromol 2024; 264:130568. [PMID: 38447822 DOI: 10.1016/j.ijbiomac.2024.130568] [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: 12/28/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Polysaccharide based self-healing and injectable hydrogels with reversible characteristics have widespread potential in protein drug delivery. However, it is a challenge to design the dynamic hydrogel for sequential release of protein drugs. Herein, we developed a novel mussel inspired sequential protein delivery dynamic polysaccharide hydrogel. The nanocomposite hydrogel can be fabricated through doping polydopamine nanoparticles (PDA NPs) into reversible covalent bond (imine bonds) crosslinked polymer networks of oxidized hyaluronic acid (OHA) and carboxymethyl chitosan (CEC), named PDA NPs@OHA-l-CEC. Besides multiple capabilities (i.e., injection, self-healing, and biodegradability), the nanocomposite hydrogel can achieve sustained and sequential protein delivery of vascular endothelial growth factor (VEGF) and bovine serum albumin (BSA). PDA NPs doped in hydrogel matrix serve dual roles, acting as secondary protein release structures and form dynamic non-covalent interactions (i.e., hydrogen bonds) with polysaccharides. Moreover, by adjusting the oxidation degree of OHA, the hydrogels with different crosslinking density could control overall protein release rate. Analysis of different release kinetic models revealed that Fickian diffusion drove rapid VEGF release, while the slower BSA release followed a Super Case II transport mechanism. The novel biocompatible system achieved sequential release of protein drugs has potentials in multi-stage synergistic drug deliver based on dynamic hydrogel.
Collapse
Affiliation(s)
- Xiao Shuai Han
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Peng Cheng Li
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Heng Tao Song
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, PR China
| | - Yong Mei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, PR China.
| | - Jian Hui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, PR China.
| | - Yang Yang
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Hao Peng Li
- Second Affiliated Hospital of Xi'an Jiaotong University, College of Chemistry, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hideyuki Miyatake
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Emergent Bioengineering Materials Research Team, RIKEN Center formergent Matter Science, 2-1 Hirosawa, Wako, Saitama 3510198, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Emergent Bioengineering Materials Research Team, RIKEN Center formergent Matter Science, 2-1 Hirosawa, Wako, Saitama 3510198, Japan
| |
Collapse
|
14
|
Li M, Jin M, Yang H. Remodelers of the vascular microenvironment: The effect of biopolymeric hydrogels on vascular diseases. Int J Biol Macromol 2024; 264:130764. [PMID: 38462100 DOI: 10.1016/j.ijbiomac.2024.130764] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Vascular disease is the leading health problem worldwide. Vascular microenvironment encompasses diverse cell types, including those within the vascular wall, blood cells, stromal cells, and immune cells. Initiation of the inflammatory state of the vascular microenvironment and changes in its mechanics can profoundly affect vascular homeostasis. Biomedical materials play a crucial role in modern medicine, hydrogels, characterized by their high-water content, have been increasingly utilized as a three-dimensional interaction network. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular microenvironment have enabled the treatment of vascular diseases. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the various vascular diseases including atherosclerosis, aneurysm, vascular ulcers of the lower limbs and myocardial infarction. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments.
Collapse
Affiliation(s)
- Minhao Li
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China.
| |
Collapse
|
15
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [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
|
16
|
Vetter VC, Bouten CVC, van der Pol A. Hydrogels for Cardiac Restorative Support: Relevance of Gelation Mechanisms for Prospective Clinical Use. Curr Heart Fail Rep 2023; 20:519-529. [PMID: 37812347 PMCID: PMC10746579 DOI: 10.1007/s11897-023-00630-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Cardiac tissue regenerative strategies have gained much traction over the years, in particular those utilizing hydrogels. With our review, and with special focus on supporting post-myocardial infarcted tissue, we aim to provide insights in determining crucial design considerations of a hydrogel and the implications these could have for future clinical use. RECENT FINDINGS To date, two hydrogel delivery strategies are being explored, cardiac injection or patch, to treat myocardial infarction. Recent advances have demonstrated that the mechanism by which a hydrogel is gelated (i.e., physically or chemically cross-linked) not only impacts the biocompatibility, mechanical properties, and chemical structure, but also the route of delivery of the hydrogel and thus its effect on cardiac repair. With regard to cardiac regeneration, various hydrogels have been developed with the ability to function as a delivery system for therapeutic strategies (e.g., drug and stem cells treatments), as well as a scaffold to guide cardiac tissue regeneration following myocardial infarction. However, these developments remain within the experimental and pre-clinical realm and have yet to transition towards the clinical setting.
Collapse
Affiliation(s)
- Valentine C Vetter
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Atze van der Pol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
17
|
Feng K, Ruan Y, Zhang X, Wu X, Liu Z, Sun X. Photothermal-Ionic-Pharmacotherapy of Myocardial Infarction with Enhanced Angiogenesis and Antiapoptosis. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38031235 DOI: 10.1021/acsami.3c14109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Promoting angiogenesis is an effective therapeutic strategy to repair damaged hearts after myocardial infarction (MI). Copper ions and mild heat (41-42 °C) have been shown to promote angiogenesis, but their efficacy in MI is unknown. Here, a multicomponent hydrogel (EDR@PHCuS HG) is developed by encapsulating edaravone (EDR, a free radical scavenger) loaded porous hollow copper sulfide nanoparticles (PHCuS NPs) in a hyaluronic acid hydrogel (HG). Exposed to 808 nm near-infrared (NIR) light irradiation, the EDR@PHCuS HG exhibits controlled copper-ion release and mild photothermal effect to synergistically promote angiogenesis. In addition, released EDR inhibits cardiomyocyte apoptosis to further repair hearts. In the mouse model of MI, treatment with the EDR@PHCuS HG under an 808 nm laser significantly recovers the cardiac function and inhibits ventricular remodeling. This platform elucidates the cardioprotective effects of copper ions and mild heat and will provide a highly efficient treatment for MI.
Collapse
Affiliation(s)
- Kai Feng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yiling Ruan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xinmiao Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaojing Wu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zixuan Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
18
|
Amini H, Namjoo AR, Narmi MT, Mardi N, Narimani S, Naturi O, Khosrowshahi ND, Rahbarghazi R, Saghebasl S, Hashemzadeh S, Nouri M. Exosome-bearing hydrogels and cardiac tissue regeneration. Biomater Res 2023; 27:99. [PMID: 37803483 PMCID: PMC10559618 DOI: 10.1186/s40824-023-00433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND In recent years, cardiovascular disease in particular myocardial infarction (MI) has become the predominant cause of human disability and mortality in the clinical setting. The restricted capacity of adult cardiomyocytes to proliferate and restore the function of infarcted sites is a challenging issue after the occurrence of MI. The application of stem cells and byproducts such as exosomes (Exos) has paved the way for the alleviation of cardiac tissue injury along with conventional medications in clinics. However, the short lifespan and activation of alloreactive immune cells in response to Exos and stem cells are the main issues in patients with MI. Therefore, there is an urgent demand to develop therapeutic approaches with minimum invasion for the restoration of cardiac function. MAIN BODY Here, we focused on recent data associated with the application of Exo-loaded hydrogels in ischemic cardiac tissue. Whether and how the advances in tissue engineering modalities have increased the efficiency of whole-based and byproducts (Exos) therapies under ischemic conditions. The integration of nanotechnology and nanobiology for designing novel smart biomaterials with therapeutic outcomes was highlighted. CONCLUSION Hydrogels can provide suitable platforms for the transfer of Exos, small molecules, drugs, and other bioactive factors for direct injection into the damaged myocardium. Future studies should focus on the improvement of physicochemical properties of Exo-bearing hydrogel to translate for the standard treatment options.
Collapse
Affiliation(s)
- Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi Narmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Narimani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Shahriar Hashemzadeh
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
He G, Chen G, Liu W, Ye D, Liu X, Liang X, Song J. Salvianolic Acid B: A Review of Pharmacological Effects, Safety, Combination Therapy, New Dosage Forms, and Novel Drug Delivery Routes. Pharmaceutics 2023; 15:2235. [PMID: 37765204 PMCID: PMC10538146 DOI: 10.3390/pharmaceutics15092235] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Salvianolic acid B is extracted from the roots and rhizomes of Danshen (Salvia miltiorrhiza Bge., family Labiatae). It is a water-soluble, weakly acidic drug that has demonstrated antitumor and anti-inflammatory effects on various organs and tissues such as the lung, heart, kidney, intestine, bone, liver, and skin and protective effects in diseases such as depression and spinal cord injury. The mechanisms underlying the protective effects of salvianolic acid B are mainly related to its anti-inflammatory, antioxidant, anti- or pro-apoptotic, anti- or pro-autophagy, anti-fibrotic, and metabolism-regulating functions. Salvianolic acid B can regulate various signaling pathways, cells, and molecules to achieve maximum therapeutic effects. This review summarizes the safety profile, combination therapy potential, and new dosage forms and delivery routes of salvianolic acid B. Although significant research progress has been made, more in-depth pharmacological studies are warranted to identify the mechanism of action, related signaling pathways, more suitable combination drugs, more effective dosage forms, and novel routes of administration of salvianolic acid B.
Collapse
Affiliation(s)
- Guannan He
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Guangfeng Chen
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Weidong Liu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Dongxue Ye
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Xuehuan Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Xiaodong Liang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Jing Song
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
- Shandong Yuze Pharmaceutical Industry Technology Research Institute Co., Ltd., Dezhou 251200, China
| |
Collapse
|
20
|
Mavridi-Printezi A, Menichetti A, Mordini D, Montalti M. Functionalization of and through Melanin: Strategies and Bio-Applications. Int J Mol Sci 2023; 24:9689. [PMID: 37298641 PMCID: PMC10253489 DOI: 10.3390/ijms24119689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
A unique feature of nanoparticles for bio-application is the ease of achieving multi-functionality through covalent and non-covalent functionalization. In this way, multiple therapeutic actions, including chemical, photothermal and photodynamic activity, can be combined with different bio-imaging modalities, such as magnetic resonance, photoacoustic, and fluorescence imaging, in a theragnostic approach. In this context, melanin-related nanomaterials possess unique features since they are intrinsically biocompatible and, due to their optical and electronic properties, are themselves very efficient photothermal agents, efficient antioxidants, and photoacoustic contrast agents. Moreover, these materials present a unique versatility of functionalization, which makes them ideal for the design of multifunctional platforms for nanomedicine integrating new functions such as drug delivery and controlled release, gene therapy, or contrast ability in magnetic resonance and fluorescence imaging. In this review, the most relevant and recent examples of melanin-based multi-functionalized nanosystems are discussed, highlighting the different methods of functionalization and, in particular, distinguishing pre-functionalization and post-functionalization. In the meantime, the properties of melanin coatings employable for the functionalization of a variety of material substrates are also briefly introduced, especially in order to explain the origin of the versatility of melanin functionalization. In the final part, the most relevant critical issues related to melanin functionalization that may arise during the design of multifunctional melanin-like nanoplatforms for nanomedicine and bio-application are listed and discussed.
Collapse
Affiliation(s)
| | | | | | - Marco Montalti
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.-P.); (A.M.); (D.M.)
| |
Collapse
|
21
|
Wang Q, Zhang Y, Ma Y, Wang M, Pan G. Nano-crosslinked dynamic hydrogels for biomedical applications. Mater Today Bio 2023; 20:100640. [PMID: 37179534 PMCID: PMC10173301 DOI: 10.1016/j.mtbio.2023.100640] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Hydrogels resemble natural extracellular matrices and have been widely studied for biomedical applications. Nano-crosslinked dynamic hydrogels combine the injectability and self-healing property of dynamic hydrogels with the versatility of nanomaterials and exhibit unique advantages. The incorporation of nanomaterials as crosslinkers can improve the mechanical properties (strength, injectability, and shear-thinning properties) of hydrogels by reinforcing the skeleton and endowing them with multifunctionality. Nano-crosslinked functional hydrogels that can respond to external stimuli (such as pH, heat, light, and electromagnetic stimuli) and have photothermal properties, antimicrobial properties, stone regeneration abilities, or tissue repair abilities have been constructed through reversible covalent crosslinking strategies and physical crosslinking strategies. The possible cytotoxicity of the incorporated nanomaterials can be reduced. Nanomaterial hydrogels show excellent biocompatibility and can facilitate cell proliferation and differentiation for biomedical applications. This review introduces different nano-crosslinked dynamic hydrogels in the medical field, from fabrication to application. In this review, nanomaterials for dynamic hydrogel fabrication, such as metals and metallic oxides, nanoclays, carbon-based nanomaterials, black phosphorus (BP), polymers, and liposomes, are discussed. We also introduce the dynamic crosslinking method commonly used for nanodynamic hydrogels. Finally, the medical applications of nano-crosslinked hydrogels are presented. We hope that this summary will help researchers in the related research fields quickly understand nano-crosslinked dynamic hydrogels to develop more preparation strategies and promote their development and application.
Collapse
Affiliation(s)
- Qinghe Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
| | - Yan Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
| | - Yue Ma
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
| | - Miao Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
- Corresponding author.
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
- Corresponding author.
| |
Collapse
|
22
|
Carvalho T, Bártolo R, Pedro SN, Valente BFA, Pinto RJB, Vilela C, Shahbazi MA, Santos HA, Freire CSR. Injectable Nanocomposite Hydrogels of Gelatin-Hyaluronic Acid Reinforced with Hybrid Lysozyme Nanofibrils-Gold Nanoparticles for the Regeneration of Damaged Myocardium. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37200222 DOI: 10.1021/acsami.3c03874] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Biopolymeric injectable hydrogels are promising biomaterials for myocardial regeneration applications. Besides being biocompatible, they adjust themselves, perfectly fitting the surrounding tissue. However, due to their nature, biopolymeric hydrogels usually lack desirable functionalities, such as antioxidant activity and electrical conductivity, and in some cases, mechanical performance. Protein nanofibrils (NFs), such as lysozyme nanofibrils (LNFs), are proteic nanostructures with excellent mechanical performance and antioxidant activity, which can work as nanotemplates to produce metallic nanoparticles. Here, gold nanoparticles (AuNPs) were synthesized in situ in the presence of LNFs, and the obtained hybrid AuNPs@LNFs were incorporated into gelatin-hyaluronic acid (HA) hydrogels for myocardial regeneration applications. The resulting nanocomposite hydrogels showed improved rheological properties, mechanical resilience, antioxidant activity, and electrical conductivity, especially for the hydrogels containing AuNPs@LNFs. The swelling and bioresorbability ratios of these hydrogels are favorably adjusted at lower pH levels, which correspond to the ones in inflamed tissues. These improvements were observed while maintaining important properties, namely, injectability, biocompatibility, and the ability to release a model drug. Additionally, the presence of AuNPs allowed the hydrogels to be monitorable through computer tomography. This work demonstrates that LNFs and AuNPs@LNFs are excellent functional nanostructures to formulate injectable biopolymeric nanocomposite hydrogels for myocardial regeneration applications.
Collapse
Affiliation(s)
- Tiago Carvalho
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Raquel Bártolo
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sónia N Pedro
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Bruno F A Valente
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Ricardo J B Pinto
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Carla Vilela
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Carmen S R Freire
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
23
|
Kulkarni N, Rao P, Jadhav GS, Kulkarni B, Kanakavalli N, Kirad S, Salunke S, Tanpure V, Sahu B. Emerging Role of Injectable Dipeptide Hydrogels in Biomedical Applications. ACS OMEGA 2023; 8:3551-3570. [PMID: 36743055 PMCID: PMC9893456 DOI: 10.1021/acsomega.2c05601] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/30/2022] [Indexed: 06/18/2023]
Abstract
Owing to their properties such as biocompatibility, tunable mechanical properties, permeability toward oxygen, nutrients, and the ability to hold a significant amount of water, hydrogels have wide applications in biomedical research. They have been engaged in drug delivery systems, 3D cell culture, imaging, and extracellular matrix (ECM) mimetics. Injectable hydrogels represent a major subset of hydrogels possessing advantages of site-specific conformation with minimal invasive techniques. It preserves the inherent properties of drug/biomolecules and is devoid of any side effects associated with surgery. Various polymeric materials utilized in developing injectable hydrogels are associated with the limitations of toxicity, immunogenicity, tedious manufacturing processes, and lack of easy synthetic tunability. Peptides are an important class of biomaterials that have interesting properties such as biocompatibility, stimuli responsiveness, shear thinning, self-healing, and biosignaling. They lack immunogenicity and toxicity. Therefore, numerous peptide-based injectable hydrogels have been explored in the past, and a few of them have reached the market. In recent years, minimalistic dipeptides have shown their ability to form stable hydrogels through cooperative noncovalent interactions. In addition to inherent properties of lengthy peptide-based injectable hydrogels, dipeptides have the unique advantages of low production cost, high synthetic accessibility, and higher stability. Given the instances of expanding significance of injectable peptide hydrogels in biomedical research and an emerging recent trend of dipeptide-based injectable hydrogels, a timely review on dipeptide-based injectable hydrogels shall highlight various aspects of this interesting class of biomaterials. This concise review that focuses on the dipeptide injectable hydrogel may stimulate the current trends of research on this class of biomaterial to translate its significance as interesting products for biomedical applications.
Collapse
Affiliation(s)
- Neeraj Kulkarni
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| | - Prajakta Rao
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
- Quality
Operations, Novartis Healthcare Pvt. Ltd., Knowledge City, Raidurg, Hyderabad 500081, Telangana, India
| | - Govinda Shivaji Jadhav
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| | - Bhakti Kulkarni
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
- Springer
Nature Technology and Publishing Solutions, Hadapsar, Pune 411013, Maharashtra, India
| | - Nagaraju Kanakavalli
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
- Aragen
Life Sciences Pvt, Ltd., Madhapur, Hyderabad 500076, Telangana, India
| | - Shivani Kirad
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| | - Sujit Salunke
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| | - Vrushali Tanpure
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| | - Bichismita Sahu
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar 382355, India
| |
Collapse
|
24
|
Chen X, Zhu L, Wang X, Xiao J. Insight into Heart-Tailored Architectures of Hydrogel to Restore Cardiac Functions after Myocardial Infarction. Mol Pharm 2023; 20:57-81. [PMID: 36413809 DOI: 10.1021/acs.molpharmaceut.2c00650] [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: 11/23/2022]
Abstract
With permanent heart muscle injury or death, myocardial infarction (MI) is complicated by inflammatory, proliferation and remodeling phases from both the early ischemic period and subsequent infarct expansion. Though in situ re-establishment of blood flow to the infarct zone and delays of the ventricular remodeling process are current treatment options of MI, they fail to address massive loss of viable cardiomyocytes while transplanting stem cells to regenerate heart is hindered by their poor retention in the infarct bed. Equipped with heart-specific mimicry and extracellular matrix (ECM)-like functionality on the network structure, hydrogels leveraging tissue-matching biomechanics and biocompatibility can mechanically constrain the infarct and act as localized transport of bioactive ingredients to refresh the dysfunctional heart under the constant cyclic stress. Given diverse characteristics of hydrogel including conductivity, anisotropy, adhesiveness, biodegradability, self-healing and mechanical properties driving local cardiac repair, we aim to investigate and conclude the dynamic balance between ordered architectures of hydrogels and the post-MI pathological milieu. Additionally, our review summarizes advantages of heart-tailored architectures of hydrogels in cardiac repair following MI. Finally, we propose challenges and prospects in clinical translation of hydrogels to draw theoretical guidance on cardiac repair and regeneration after MI.
Collapse
Affiliation(s)
- Xuerui Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Liyun Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xu Wang
- Hangzhou Medical College, Binjiang Higher Education Park, Binwen Road 481, Hangzhou 310053, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
25
|
Bai X, Fan W, Luo Y, Liu Y, Zhang Y, Liao X. Fast Screening of Protein Tyrosine Phosphatase 1B Inhibitor from Salvia miltiorrhiza Bge by Cell Display-Based Ligand Fishing. Molecules 2022; 27:molecules27227896. [PMID: 36431993 PMCID: PMC9693971 DOI: 10.3390/molecules27227896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
Salvia miltiorrhiza Bge is a medicinal plant (Chinese name "Danshen") widely used for the treatment of hyperglycemia in traditional Chinese medicine. Protein tyrosine phosphatase 1B (PTP1B) has been recognized as a potential target for insulin sensitizing for the treatment of diabetes. In this work, PTP1B was displayed at the surface of E. coli cells (EC-PTP1B) to be used as a bait for fishing of the enzyme's inhibitors present in the aqueous extract of S. miltiorrhiza. Salvianolic acid B, a polyphenolic compound, was fished out by EC-PTP1B, which was found to inhibit PTP1B with an IC50 value of 23.35 µM. The inhibitory mechanism of salvianolic acid B was further investigated by enzyme kinetic experiments and molecular docking, indicating salvianolic acid B was a non-competitive inhibitor for PTP1B (with Ki and Kis values of 31.71 µM and 20.08 µM, respectively) and its binding energy was -7.89 kcal/mol. It is interesting that in the comparative work using a traditional ligand fishing bait of PTP1B-immobilized magnetic nanoparticles (MNPs-PTP1B), no ligands were extracted at all. This study not only discovered a new PTP1B inhibitor from S. miltiorrhiza which is significant to understand the chemical basis for the hypoglycemic activity of this plant, but also indicated the effectiveness of cell display-based ligand fishing in screening of active compounds from complex herbal extracts.
Collapse
Affiliation(s)
- Xiaolin Bai
- Chinese Academy of Sciences, Chengdu Institute of Biology, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqin Fan
- Chinese Academy of Sciences, Chengdu Institute of Biology, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingjie Luo
- Department of Molecular Science, The University of Western Australia, Perth, WA 6000, Australia
| | - Yipei Liu
- Polus International College, Chengdu 610103, China
| | - Yongmei Zhang
- Chinese Academy of Sciences, Chengdu Institute of Biology, Chengdu 610041, China
- Correspondence: (Y.Z.); (X.L.); Tel.: +86-28-82890756 (Y.Z.); +86-28-828290402 (X.L.)
| | - Xun Liao
- Chinese Academy of Sciences, Chengdu Institute of Biology, Chengdu 610041, China
- Correspondence: (Y.Z.); (X.L.); Tel.: +86-28-82890756 (Y.Z.); +86-28-828290402 (X.L.)
| |
Collapse
|
26
|
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: 37] [Impact Index Per Article: 18.5] [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
|
27
|
Shen P, Jia Y, Shi S, Sun J, Han X. Analytical and biomedical applications of microfluidics in traditional Chinese medicine research. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Strategies for Improving Peptide Stability and Delivery. Pharmaceuticals (Basel) 2022; 15:ph15101283. [PMID: 36297395 PMCID: PMC9610364 DOI: 10.3390/ph15101283] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Peptides play an important role in many fields, including immunology, medical diagnostics, and drug discovery, due to their high specificity and positive safety profile. However, for their delivery as active pharmaceutical ingredients, delivery vectors, or diagnostic imaging molecules, they suffer from two serious shortcomings: their poor metabolic stability and short half-life. Major research efforts are being invested to tackle those drawbacks, where structural modifications and novel delivery tactics have been developed to boost their ability to reach their targets as fully functional species. The benefit of selected technologies for enhancing the resistance of peptides against enzymatic degradation pathways and maximizing their therapeutic impact are also reviewed. Special note of cell-penetrating peptides as delivery vectors, as well as stapled modified peptides, which have demonstrated superior stability from their parent peptides, are reported.
Collapse
|
29
|
Yue T, Xiong S, Zheng D, Wang Y, Long P, Yang J, Danzeng D, Gao H, Wen X, Li X, Hou J. Multifunctional biomaterial platforms for blocking the fibrosis process and promoting cellular restoring effects in myocardial fibrosis therapy. Front Bioeng Biotechnol 2022; 10:988683. [PMID: 36185428 PMCID: PMC9520723 DOI: 10.3389/fbioe.2022.988683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Myocardial fibrosis is the result of abnormal healing after acute and chronic myocardial damage and is a direct cause of heart failure and cardiac insufficiency. The clinical approach is to preserve cardiac function and inhibit fibrosis through surgery aimed at dredging blood vessels. However, this strategy does not adequately address the deterioration of fibrosis and cardiac function recovery. Therefore, numerous biomaterial platforms have been developed to address the above issues. In this review, we summarize the existing biomaterial delivery and restoring platforms, In addition, we also clarify the therapeutic strategies based on biomaterial platforms, including general strategies to block the fibrosis process and new strategies to promote cellular restoring effects. The development of structures with the ability to block further fibrosis progression as well as to promote cardiomyocytes viability should be the main research interests in myocardial fibrosis, and the reestablishment of structures necessary for normal cardiac function is central to the treatment of myocardial fibrosis. Finally, the future application of biomaterials for myocardial fibrosis is also highlighted.
Collapse
Affiliation(s)
- Tian Yue
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Shiqiang Xiong
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
| | - Dezhi Zheng
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Pan Long
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jiali Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Dunzhu Danzeng
- Department of Basic Medicine, Medical College, Tibet University, Lhasa, China
| | - Han Gao
- Department of Basic Medicine, Medical College, Tibet University, Lhasa, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People’s Hospital, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| | - Xin Li
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| | - Jun Hou
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| |
Collapse
|
30
|
Protective Effect of Natural Medicinal Plants on Cardiomyocyte Injury in Heart Failure: Targeting the Dysregulation of Mitochondrial Homeostasis and Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3617086. [PMID: 36132224 PMCID: PMC9484955 DOI: 10.1155/2022/3617086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/16/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Heart failure occurs because of various cardiovascular pathologies, such as coronary artery disease or cardiorenal syndrome, eventually reaching end-stage disease. Various factors contribute to cardiac structural or functional changes that result in systolic or diastolic dysfunction. Several studies have confirmed that the key factor in heart failure progression is myocardial cell death, and mitophagy is the major mechanism regulating myocardial cell death in heart failure. The clinical mechanisms of heart failure are well understood in practice. However, the essential role of mitophagic regulation in heart failure has only recently received widespread attention. Receptor-mediated mitophagy is involved in various mitochondrial processes like oxidative stress injury, energy metabolism disorders, and calcium homeostasis, which are also the main causes of heart failure. Understanding of the diverse regulatory mechanisms in mitophagy and the complexity of its pathophysiology in heart failure remains incomplete. Related studies have found that various natural medicinal plants and active ingredients, such as flavonoids and saponins, can regulate mitophagy to a certain extent, improve myocardial function, and protect myocardial cells. This review comprehensively covers the relevant mechanisms of different types of mitophagy in regulating heart failure pathology and controlling mitochondrial adaptability to stress injury. Further, it explores the relationship between mitophagy and cardiac ejection dysfunction. Natural medicinal plant-targeted regulation strategies and scientific evidence on mitophagy were provided to elucidate current and potential strategies to apply mitophagy-targeted therapy for heart failure.
Collapse
|
31
|
Tapeinos C, Gao H, Bauleth-Ramos T, Santos HA. Progress in Stimuli-Responsive Biomaterials for Treating Cardiovascular and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200291. [PMID: 35306751 DOI: 10.1002/smll.202200291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) describe abnormal vascular system conditions affecting the brain and heart. Among these, ischemic heart disease and ischemic stroke are the leading causes of death worldwide, resulting in 16% and 11% of deaths globally. Although several therapeutic approaches are presented over the years, the continuously increasing mortality rates suggest the need for more advanced strategies for their treatment. One of these strategies lies in the use of stimuli-responsive biomaterials. These "smart" biomaterials can specifically target the diseased tissue, and after "reading" the altered environmental cues, they can respond by altering their physicochemical properties and/or their morphology. In this review, the progress in the field of stimuli-responsive biomaterials for CCVDs in the last five years, aiming at highlighting their potential as early-stage therapeutics in the preclinical scenery, is described.
Collapse
Affiliation(s)
- Christos Tapeinos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Tomás Bauleth-Ramos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
32
|
Wang D, Cao H, Hua W, Gao L, Yuan Y, Zhou X, Zeng Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Bone Defect Repair. MEMBRANES 2022; 12:membranes12070716. [PMID: 35877919 PMCID: PMC9315966 DOI: 10.3390/membranes12070716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022]
Abstract
The repair of critical bone defects is a hotspot of orthopedic research. With the development of bone tissue engineering (BTE), there is increasing evidence showing that the combined application of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) (MSC-EVs), especially exosomes, with hydrogels, scaffolds, and other bioactive materials has made great progress, exhibiting a good potential for bone regeneration. Recent studies have found that miRNAs, proteins, and other cargo loaded in EVs are key factors in promoting osteogenesis and angiogenesis. In BTE, the expression profile of the intrinsic cargo of EVs can be changed by modifying the gene expression of MSCs to obtain EVs with enhanced osteogenic activity and ultimately enhance the osteoinductive ability of bone graft materials. However, the current research on MSC-EVs for repairing bone defects is still in its infancy, and the underlying mechanism remains unclear. Therefore, in this review, the effect of bioactive materials such as hydrogels and scaffolds combined with MSC-EVs in repairing bone defects is summarized, and the mechanism of MSC-EVs promoting bone defect repair by delivering active molecules such as internal miRNAs is further elucidated, which provides a theoretical basis and reference for the clinical application of MSC-EVs in repairing bone defects.
Collapse
Affiliation(s)
- Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
| | - Weizhong Hua
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Lu Gao
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Yu Yuan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
- Correspondence: (X.Z.); (Z.Z.)
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
- Correspondence: (X.Z.); (Z.Z.)
| |
Collapse
|
33
|
Hou Y, Tan T, Guo Z, Ji Y, Hu J, Zhang Y. Gram-selective antibacterial peptide hydrogels. Biomater Sci 2022; 10:3831-3844. [PMID: 35678287 DOI: 10.1039/d2bm00558a] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The human microbiome plays fundamental roles in human health and disease. However, widely used broad-spectrum antibiotics severely disrupt human-related microbial communities, eventually leading to resistant bacteria, posing a growing threat to global medical health. Antimicrobial peptides (AMPs) are promising antimicrobial agents that barely cause bacterial resistance. Excellent broad-spectrum antimicrobial activities have been achieved using hydrogels self-assembled from AMPs, but there is still a lack of AMP hydrogels that can target Gram-positive and Gram-negative bacteria. Herein, several hydrogels self-assembled from AMPs, termed IK1, IK3, and IK4, were designed and synthesized. In vitro antibacterial results indicated that the IK1 and IK4 hydrogels specifically targeted Gram-positive and Gram-negative bacteria, respectively, while the IK3 hydrogel targeted both Gram-positive and Gram-negative bacteria. The desired broad-spectrum or Gram-selective AMP hydrogels are believed to be obtained through the rational design of the hydrophilicity, hydrophobicity, and charge properties of the peptide molecules. Good in vivo Gram-selective antibacterial properties and the ability to promote wound healing have been demonstrated via treating mouse wound models with these AMP hydrogels. We believe that these Gram-selective AMP hydrogels could potentially have important applications in treating common recurring infections.
Collapse
Affiliation(s)
- Yangqian Hou
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingyuan Tan
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Guo
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuwen Ji
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Hu
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yi Zhang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China. .,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
34
|
Hu W, Yang C, Guo X, Wu Y, Loh XJ, Li Z, Wu YL, Wu C. Research Advances of Injectable Functional Hydrogel Materials in the Treatment of Myocardial Infarction. Gels 2022; 8:423. [PMID: 35877508 PMCID: PMC9316750 DOI: 10.3390/gels8070423] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 12/10/2022] Open
Abstract
Myocardial infarction (MI) has become one of the serious diseases threatening human life and health. However, traditional treatment methods for MI have some limitations, such as irreversible myocardial necrosis and cardiac dysfunction. Fortunately, recent endeavors have shown that hydrogel materials can effectively prevent negative remodeling of the heart and improve the heart function and long-term prognosis of patients with MI due to their good biocompatibility, mechanical properties, and electrical conductivity. Therefore, this review aims to summarize the research progress of injectable hydrogel in the treatment of MI in recent years and to introduce the rational design of injectable hydrogels in myocardial repair. Finally, the potential challenges and perspectives of injectable hydrogel in this field will be discussed, in order to provide theoretical guidance for the development of new and effective treatment strategies for MI.
Collapse
Affiliation(s)
- Wei Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Cui Yang
- School of Medicine, Xiamen University, Xiamen 361003, China;
| | - Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Yihong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE) Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| |
Collapse
|
35
|
Zhu S, Yu C, Liu N, Zhao M, Chen Z, Liu J, Li G, Huang H, Guo H, Sun T, Chen J, Zhuang J, Zhu P. Injectable conductive gelatin methacrylate / oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact Mater 2022; 13:119-134. [PMID: 35224296 PMCID: PMC8844712 DOI: 10.1016/j.bioactmat.2021.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 02/03/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) transplantation has been proposed as a promising treatment modality for myocardial infarction (MI), but the low retention rate remains a considerable challenge. Injectable natural polymer hydrogels with conductivity ability are highly desirable as cell delivery vehicles to repair infarct myocardium and restore the cardiac function. In this work, we developed a hydrogel system based on gelatin methacrylate (GelMA) and oxidized dextran (ODEX) as cell delivery vehicles for MI. And dopamine could be used as a reductant of graphene oxide (GO) to form reductive GO (rGO). By adjusting the amount of rGO, the conductivity of hydrogels with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. In vitro cell experiments showed that the prepared hydrogels had excellent biocompatibility and cell delivery ability of UCMSCs. More importantly, GelMA-O5/rGO hydrogel could promote UCMSCs growth and proliferation, improve the myocardial differentiation ability of UCMSCs, and up-regulate the expression of cTnI and Cx43. Further in vivo experiments demonstrated that GelMA-O5/rGO/UCMSCs Hydrogel could significantly improve the ejection fraction (EF) of rats and significantly reduce myocardial infarct area compared to PBS group, promote the survival of UCMSCs, enhance the expression level of cTnI and Cx43, and decrease the expression level of caspase-3. The findings of this study suggested that the injectable conductive GelMA-O5/rGO hydrogel encapsulating UCMSCs could improve damaged myocardial tissue and reconstruct myocardial function, which will be a promising therapeutic strategy for cardiac repair. Conducting interpenetrating polymer network (IPN) hydrogels were synthesized for myocardial infarction treatment. The conductivity of hydrogel with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. The hydrogel could promote the growth and proliferation of UCMSCs, and improve the myocardial differentiation ability of UCMSCs. The hydrogel could reduce infarct size and cardiac fibrosis in the infarct zone, increase ventricular ejection fraction. The hydrogel could promote the survival of UCMSCs, up-regulate the expression level of cTnI and Cx43, down-regulate the expression level of caspase-3.
Collapse
|
36
|
An injectable co-assembled hydrogel blocks reactive oxygen species and inflammation cycle resisting myocardial ischemia-reperfusion injury. Acta Biomater 2022; 149:82-95. [PMID: 35777549 DOI: 10.1016/j.actbio.2022.06.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/02/2022] [Accepted: 06/22/2022] [Indexed: 12/23/2022]
Abstract
The overproduction of reactive oxygen species (ROS) and burst of inflammation following cardiac ischemia-reperfusion (I/R) are the leading causes of cardiomyocyte injury. Monotherapeutic strategies designed to enhance anti-inflammatory or anti-ROS activity explicitly for treating I/R injury have demonstrated limited success because of the complex mechanisms of ROS production and induction of inflammation. Intense oxidative stress leads to sustained injury, necrosis, and apoptosis of cardiomyocytes. The damaged and necrotic cells can release danger-associated molecular patterns (DAMPs) that can cause the aggregation of immune cells by activating Toll-like receptor 4 (TLR4). These immune cells also promote ROS production by expressing NADPH oxidase. Finally, ROS production and inflammation form a vicious cycle, and ROS and TLR4 are critical nodes of this cycle. In the present study, we designed and prepared an injectable hydrogel system of EGCG@Rh-gel by co-assembling epigallocatechin-3-gallate (EGCG) and the rhein-peptide hydrogel (Rh-gel). The co-assembled hydrogel efficiently blocked the ROS-inflammation cycle by ROS scavenging and TLR4 inhibition. Benefited by the abundant noncovalent interactions of π-π stacking and hydrogen bonding between EGCG and Rh-gel, the co-assembled hydrogel had good mechanical strength and injectable property. Following the injection EGCG@Rh-gel into the damaged region of the mice's heart after I/R, the hydrogel enabled to achieve long-term sustained release and treatment, improve cardiac function, and significantly reduce the formation of scarring. Further studies demonstrated that these beneficial outcomes arise from the reduction of ROS production, inhibition of inflammation, and induction of anti-apoptosis in cardiomyocytes. Therefore, EGCG@Rh-gel is a promising drug delivery system to block the ROS-inflammation cycle for resisting myocardial I/R injury. STATEMENT OF SIGNIFICANCE: 1. Monotherapeutic strategies designed to enhance anti-inflammatory or anti-ROS effects for treating I/R injury have demonstrated limited success because of the complex mechanisms of ROS and inflammation. 2. ROS production and inflammation form a vicious cycle, and ROS and TLR4 are critical nodes of this cycle. 3. Here, we designed an injectable hydrogel system of EGCG@Rh-gel by co-assembling epigallocatechin-3-gallate (EGCG) and a rhein-peptide hydrogel (Rh-gel). EGCG@Rh-gel efficiently blocked the ROS-inflammation cycle by ROS scavenging and TLR4 inhibition. 4. EGCG@Rh-gel achieved long-term sustained release and treatment, improved cardiac function, and significantly reduced the formation of scarring after I/R. 5. The beneficial outcomes arise from reducing ROS production, inhibiting inflammation, and inducing anti-apoptosis in cardiomyocytes.
Collapse
|
37
|
Quan L, Xin Y, Wu X, Ao Q. Mechanism of Self-Healing Hydrogels and Application in Tissue Engineering. Polymers (Basel) 2022; 14:2184. [PMID: 35683857 PMCID: PMC9183126 DOI: 10.3390/polym14112184] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 12/26/2022] Open
Abstract
Self-healing hydrogels and traditional hydrogels both have three-dimensional polymeric networks that are capable of absorbing and retaining a large amount of water. Self-healing hydrogels can heal and restore damage automatically, and they can avoid premature failure of hydrogels caused by mechanical damage after implantation. The formation mechanism of self-healing hydrogels and the factors that hydrogels can load are various. Researchers can design hydrogels to meet the needs of different tissues through the diversity of hydrogels Therefore, it is necessary to summarize different self-healing mechanisms and different factors to achieve different functions. Here, we briefly reviewed the hydrogels designed by researchers in recent years according to the self-healing mechanism of water coagulation. Then, the factors for different functions of self-healing hydrogels in different tissues were statistically analyzed. We hope our work can provide effective support for researchers in the design process of self-healing hydrogel.
Collapse
Affiliation(s)
| | | | | | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (L.Q.); (Y.X.); (X.W.)
| |
Collapse
|
38
|
Wang B, Wu C, He S, Wang Y, Wang D, Tao H, Wang C, Pang X, Li F, Yuan Y, Gross ER, Liang G, Zhang Y. V1-Cal hydrogelation enhances its effects on ventricular remodeling reduction and cardiac function improvement post myocardial infarction. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2022; 433:134450. [PMID: 36338580 PMCID: PMC9634955 DOI: 10.1016/j.cej.2021.134450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Myocardial infarction (MI) is a major cause of disability and mortality worldwide. A cell permeable peptide V1-Cal has shown remarkable therapeutic effects on ML However, using V1-Cal to improve long-term cardiac function after MI is presently limited by its short half-life. Herein, we co-assembled V1-Cal with a well-known hydrogelator Nap-Phe-Phe-Tyr (NapFFY) to obtain a new supramolecular hydrogel V1-Cal/NapFFY. We found that the hydrogel could significantly enhance the therapeutic effects of V1-Cal on ventricular remodeling reduction and cardiac function improvement in a myocardial infarction rat model. In vitro experiments indicated that co-assembly of V1-Cal with NapFFY significantly increased mechanic strength of the hydrogel, enabling a sustained release of V1-Cal for more than two weeks. In vivo experiments supported that sustained release of V1-Cal from V1-Cal/NapFFY hydrogel could effectively decrease the expression and activation of TRPV1, reduce apoptosis and the release of inflammatory factors in a MI rat model. In particular, V1-Cal/NapFFY hydrogel significantly decreased infarct size and fibrosis, while improved cardiac function 28 days post MI. We anticipate that V1-Cal/NapFFY hydrogel could be used clinically to treat MI in the near future.
Collapse
Affiliation(s)
- Bin Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Chengfan Wu
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, PR China
| | - Shufang He
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Yaguang Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Di Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Hui Tao
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Chenchen Wang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, PR China
| | - Xiaoxi Pang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Fei Li
- Department of Nuclear Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| | - Yue Yuan
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, PR China
| | - Eric R. Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Gaolin Liang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, PR China
- State Key Laboratory of Bioelectronics School of Biological Sciences and Medical Engineering Southeast University, 2 Sipailou Road, Nanjing 210096, PR China
| | - Ye Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, PR China
| |
Collapse
|
39
|
Xie J, Yu P, Wang Z, Li J. Recent Advances of Self-Healing Polymer Materials via Supramolecular Forces for Biomedical Applications. Biomacromolecules 2022; 23:641-660. [PMID: 35199999 DOI: 10.1021/acs.biomac.1c01647] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Noncovalent interactions can maintain the three-dimensional structures of biomacromolecules (e.g., polysaccharides and proteins) and control specific recognition in biological systems. Supramolecular chemistry was gradually developed as a result, and this led to design and application of self-healing materials. Self-healing materials have attracted attention in many fields, such as coatings, bionic materials, elastomers, and flexible electronic devices. Nevertheless, self-healing materials for biomedical applications have not been comprehensively summarized, even though many reports have been focused on specific areas. In this Review, we first introduce the different categories of supramolecular forces used in preparing self-healing materials and then describe biological applications developed in the last 5 years, including antibiofouling, smart drug/protein delivery, wound healing, electronic skin, cartilage lubrication protection, and tissue engineering scaffolds. Finally, the limitations of current biomedical applications are indicated, key design points are offered for new biological self-healing materials, and potential directions for biological applications are highlighted.
Collapse
Affiliation(s)
- Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P.R. China
| | - Peng Yu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P.R. China
| | - Zhanhua Wang
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, Sichuan University, Chengdu 610065, P.R. China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
40
|
Zhang Y, Yu Y, Gao J. Supramolecular Nanomedicines of In-Situ Self-Assembling Peptides. Front Chem 2022; 10:815551. [PMID: 35186883 PMCID: PMC8854645 DOI: 10.3389/fchem.2022.815551] [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: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Nanomedicines provide distinct clinical advantages over traditional monomolecular therapeutic and diagnostic agents. Supramolecular nanomedicines made from in-situ self-assembling peptides have emerged as a promising strategy in designing and fabricating nanomedicines. In-situ self-assambly (SA) allows the combination of nanomedicines approach with prodrug approach, which exhibited both advantages of these strategies while addressed the problems of both and thus receiving more and more research attention. In this review, we summarized recently designed supramolecular nanomedicines of in-situ SA peptides in the manner of applications and design principles, and the interaction between the materials and biological environments was also discussed.
Collapse
|
41
|
Liang B, Zhang XX, Li R, Gu N. Guanxin V protects against ventricular remodeling after acute myocardial infarction through the interaction of TGF-β1 and Vimentin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153866. [PMID: 34883417 DOI: 10.1016/j.phymed.2021.153866] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Our previous study demonstrated that Guanxin V (GXV), a traditional Chinese herbal medicine, has a significant therapeutic effect on ventricular remodeling. However, the mechanistic action of GXV in ventricular remodeling warrants clarification. PURPOSE Here, we aimed to explore the anti-ventricular remodeling contribution of GXV and to provide an experimental basis for clinical generalization. METHODS A ventricular remodeling model after acute myocardial infarction was constructed in Syrian hamsters. The echocardiography and biochemical indices of cardiac function and remodeling were evaluated in different groups. Moreover, we built a remodeling model in cardiomyocytes and further explored the mechanism. Transmission electron microscopy was used to observe the ultrastructure of cardiomyocytes. The vital markers involved in the signaling pathway were detected by RT-qPCR and immunoblotting. Transforming growth factor beta 1 (TGF-β1) was overexpressed with lentivirus to verify the necessity of TGF-β1 in GXV's anti-ventricular remodeling effect. Finally, co-immunoprecipitation was conducted to test the interaction of TGF-β1 and Vimentin. RESULTS In hamster cardiac remodeling induced by acute myocardial infarction, GXV alleviated apoptosis, cardiac hypertrophy, and cardiac remodeling, and even improved cardiac function. Mechanistically, GXV inhibited the remodeling process by directly targeting TGF-β1. Overexpression of TGF-β1 exacerbated the ventricular remodeling, whereas GXV reversed this dysregulation. GXV also decreased the up-regulated Vimentin level in pathological ventricular remodeling. Moreover, the interaction of Vimentin and TGF-β1 was confirmed by co-immunoprecipitation, and GXV impeded this interaction. CONCLUSION We showed that the interaction of Vimentin and TGF-β1 may be a novel target for ventricular remodeling and that GXV might be a new agent to fight against ventricular remodeling by targeting TGF-β1 and impeding its interaction with Vimentin.
Collapse
Affiliation(s)
- Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Rui Li
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
42
|
Zhu YC, Liang B, Gu N. Cellular and Molecular Mechanism of Traditional Chinese Medicine on Ventricular Remodeling. Front Cardiovasc Med 2021; 8:753095. [PMID: 34926607 PMCID: PMC8671630 DOI: 10.3389/fcvm.2021.753095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Ventricular remodeling is related to the renin-angiotensin-aldosterone system, immune system, and various cytokines involved in inflammation, apoptosis, and cell signal regulation. Accumulated studies have shown that traditional Chinese medicine can significantly inhibit the process of ventricular remodeling, which may be related to the mechanism mentioned above. Here, we conducted a system overview to critically review the cellular and molecular mechanism of traditional Chinese medicine on ventricular remodeling. We mainly searched PubMed for basic research about the anti-ventricular remodeling of traditional Chinese medicine in 5 recent years, and then objectively summarized these researches. We included more than 25 kinds of Chinese herbal medicines including Qi-Li-Qian-Xin, Qi-Shen-Yi-Qi Pill, Xin-Ji-Er-Kang Formula, and Yi-Qi-Wen-Yang Decoction, and found that they can inhibit ventricular remodeling effectively through multi-components and multi-action targets, which are promoting the clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Yong-Chun Zhu
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
43
|
Perveen S, Rossin D, Vitale E, Rosso R, Vanni R, Cristallini C, Rastaldo R, Giachino C. Therapeutic Acellular Scaffolds for Limiting Left Ventricular Remodelling-Current Status and Future Directions. Int J Mol Sci 2021; 22:ijms222313054. [PMID: 34884856 PMCID: PMC8658014 DOI: 10.3390/ijms222313054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of heart-related deaths worldwide. Following MI, the hypoxic microenvironment triggers apoptosis, disrupts the extracellular matrix and forms a non-functional scar that leads towards adverse left ventricular (LV) remodelling. If left untreated this eventually leads to heart failure. Besides extensive advancement in medical therapy, complete functional recovery is never accomplished, as the heart possesses limited regenerative ability. In recent decades, the focus has shifted towards tissue engineering and regenerative strategies that provide an attractive option to improve cardiac regeneration, limit adverse LV remodelling and restore function in an infarcted heart. Acellular scaffolds possess attractive features that have made them a promising therapeutic candidate. Their application in infarcted areas has been shown to improve LV remodelling and enhance functional recovery in post-MI hearts. This review will summarise the updates on acellular scaffolds developed and tested in pre-clinical and clinical scenarios in the past five years with a focus on their ability to overcome damage caused by MI. It will also describe how acellular scaffolds alone or in combination with biomolecules have been employed for MI treatment. A better understanding of acellular scaffolds potentialities may guide the development of customised and optimised therapeutic strategies for MI treatment.
Collapse
Affiliation(s)
- Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Emanuela Vitale
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Rachele Rosso
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
- Correspondence:
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| |
Collapse
|