1
|
Shen J, Duan X, Xie T, Zhang X, Cai Y, Pan J, Zhang X, Sun X. Advances in locally administered nucleic acid therapeutics. Bioact Mater 2025; 49:218-254. [PMID: 40144794 PMCID: PMC11938090 DOI: 10.1016/j.bioactmat.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Nucleic acid drugs represent the latest generation of precision therapeutics, holding significant promise for the treatment of a wide range of intractable diseases. Delivery technology is crucial for the clinical application of nucleic acid drugs. However, extrahepatic delivery of nucleic acid drugs remains a significant challenge. Systemic administration often fails to achieve sufficient drug enrichment in target tissues. Localized administration has emerged as the predominant approach to facilitate extrahepatic delivery. While localized administration can significantly enhance drug accumulation at the injection sites, nucleic acid drugs still face biological barriers in reaching the target lesions. This review focuses on non-viral nucleic acid drug delivery techniques utilized in local administration for the treatment of extrahepatic diseases. First, the classification of nucleic acid drugs is described. Second, the current major non-viral delivery technologies for nucleic acid drugs are discussed. Third, the bio-barriers, administration approaches, and recent research advances in the local delivery of nucleic acid drugs for treating lung, brain, eye, skin, joint, and heart-related diseases are highlighted. Finally, the challenges associated with the localized therapeutic application of nucleic acid drugs are addressed.
Collapse
Affiliation(s)
- Jie Shen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xusheng Duan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ting Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xinrui Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yue Cai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Junhao Pan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xin Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
2
|
Yang G, Ren D, Yu T, Fang J. Biodegradable copper-doped calcium phosphate nanoplatform enables tumor microenvironment modulations for amplified ferroptosis in cervical carcinoma treatment. Int J Pharm X 2025; 9:100315. [PMID: 39811248 PMCID: PMC11731240 DOI: 10.1016/j.ijpx.2024.100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
As a recently discovered form of regulated cell death, ferroptosis has attracted much attention in the field cancer therapy. However, achieving considerably enhanced efficacy is often restricted by the overexpression of endogenous glutathione (GSH) in tumor microenvironment (TME). In this work, we report a ferroptosis-inducing strategy of GSH depletion and reactive oxygen species (ROS) generation based on a biodegradable copper-doped calcium phosphate (CaP) with L-buthionine sulfoximine (BSO) loading (denoted as BSO@CuCaP-LOD, BCCL). BCCL was conducted by a biomineralization approach using lactate oxidases (LOD) as a bio-template to obtain Cu-doped CaP nanoparticles. Then, BSO was loaded to form BCCL nanoparticles with pH-responsive biodegradability to endow controlled release of Cu2+ and BSO in response to acidic TME. Benefiting from the catalytic performance of LOD, BCCL efficiently depletes the level of lactate in tumor, which can generate endogenous H2O2 for subsequent Fenton-like reaction. The Cu2+ and BSO intracellular GSH depletion followed by GSH-mediated Cu2+/Cu+ conversion, leading to the inhibition of glutathione peroxidase 4 (GPX4) and generation of •OH radicals via Cu+-mediated Fenton-like reaction. BCCL confers enhanced ferroptosis induction via intracellular LOD-induced H2O2 production, BSO-mediated GSH depletion, and Cu+-mediated ROS generation, leading to cause effective ferroptotic cell damage. As verified by in vitro and in vivo assays, the designed BCCL nanoplatform is highly biocompatible and exhibits superior anticancer therapy on uterine cervical carcinoma U14 tumor xenografts. This study, therefore, provides a biocompatible therapeutic platform that modulating the TME to enable intensive ROS generating efficacy and GSH depleting performance, as well as provides an innovative paradigm for achieving effective ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
| | | | - Tao Yu
- Department of Gynecology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, PR China
| | - Junfeng Fang
- Department of Gynecology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, PR China
| |
Collapse
|
3
|
Alogna A, Lo Muzio FP, Catalucci D. Cardiovascular inhalation for targeted drug delivery in cardiac disease. Heart Fail Rev 2025:10.1007/s10741-025-10527-w. [PMID: 40410529 DOI: 10.1007/s10741-025-10527-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Recombinant proteins, cell, and gene therapies are collectively defined as biological drugs or biologics. These therapies have transformed the lives of millions of patients over the past decades, with the number of FDA-approved biologics increasing exponentially in recent years. However, out of approximately 700 biological therapies approved by the FDA in the last 20 years, less than 1% are indicated for cardiac pathologies. The application of biologics in cardiovascular disease has faced significant challenges, including short plasma half-life, the multifactorial complexity of cardiac disease, and the lack of efficient, non-invasive, and patient-friendly drug-delivery routes. This translational gap is particularly pressing given the immense socioeconomic burden of cardiovascular disease, which remains the leading cause of death globally and accounts for billions in annual healthcare costs and lost productivity. Inhalation-based drug delivery has recently emerged as a promising strategy for treating cardiovascular disease, with several proof-of-concept studies demonstrating its potential in heart failure, the most prevalent cardiac condition. This narrative review summarizes the latest experimental evidence in the novel field of Cardiovascular Inhalation, i.e., the lung-to-heart route for biologics. We discuss translational challenges, preclinical evidence, and future perspectives for bringing this innovative approach to clinical practice.
Collapse
Affiliation(s)
- Alessio Alogna
- Deutsches Herzzentrum Der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785, Berlin, Germany.
| | - Francesco Paolo Lo Muzio
- Deutsches Herzzentrum Der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
| | - Daniele Catalucci
- Institute of Genetic and Biomedical Research (IRGB), National Research Council of Italy, Milan Unit, 20138, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| |
Collapse
|
4
|
Garitano N, Aguado-Alvaro LP, Pelacho B. Emerging Epigenetic Therapies for the Treatment of Cardiac Fibrosis. Biomedicines 2025; 13:1170. [PMID: 40426997 PMCID: PMC12109272 DOI: 10.3390/biomedicines13051170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/27/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Fibrosis is a pathological process characterized by excessive extracellular matrix (ECM) deposition, leading to tissue stiffening and organ dysfunction. It is a major contributor to chronic diseases affecting various organs, with limited therapeutic options available. Among the different forms of fibrosis, cardiac fibrosis is particularly relevant due to its impact on cardiovascular diseases (CVDs), which remain the leading cause of morbidity and mortality worldwide. This process is driven by activated cardiac fibroblasts (CFs), which promote ECM accumulation in response to chronic stressors. Epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, are key regulators of fibroblast activation and fibrotic gene expression. Enzymes such as DNA methyltransferases (DNMTs), histone methyltransferases (HMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs) have emerged as potential therapeutic targets, and epigenetic inhibitors have shown promise in modulating these enzymes to attenuate fibrosis by controlling fibroblast function and ECM deposition. These small-molecule compounds offer advantages such as reversibility and precise temporal control, making them attractive candidates for therapeutic intervention. This review aims to provide a comprehensive overview of the mechanisms by which epigenetic regulators influence cardiac fibrosis and examines the latest advances in preclinical epigenetic therapies. By integrating recent data from functional studies, single-cell profiling, and drug development, it highlights key molecular targets, emerging therapeutic strategies, and current limitations, offering a critical framework to guide future research and clinical translation.
Collapse
Affiliation(s)
- Nerea Garitano
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Laura Pilar Aguado-Alvaro
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Beatriz Pelacho
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
5
|
Tong L, Wang Q, Zhang Y, Lai F, Xu J, Yin W, Zhang S, Wei G, Yin J, Yi H, Storm G, Wang Z, Huang R, Xu T, Wang JW. Myocardial delivery of miR30d with peptide-functionalized milk-derived extracellular vesicles for targeted treatment of hypertrophic heart failure. Biomaterials 2025; 316:122976. [PMID: 39637583 DOI: 10.1016/j.biomaterials.2024.122976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
miR30d has been shown to reverse cardiac hypertrophy. However, effective delivery of miR30d to the heart is challenging. Here, we engineered milk-derived extracellular vesicles (mEVs) by surface functionalization with an ischemic myocardium-targeting peptide (IMTP) and encapsulated miR30d to develop a formulation, the miR30d-mEVsIMTP, enabling targeted delivery of miR30d to the injured heart. In vitro, the miR30d-mEVsIMTP can be effectively internalized by hypoxia-induced H9C2 cells via the endo-lysosomal pathway. In the isoproterenol (ISO)-induced cardiac hypertrophy mice, more miR30d-mEVsIMTP accumulated in cardiac tissue than miR30d-mEVs following intravenous administration. As a result, miR30d-mEVsIMTP alleviated cardiac hypertrophy and rescued cardiac function in three murine models of hypertrophic heart failure. Mechanistically, we identified GRK5 as an unprecedented target of miR30d in cardiac hypertrophy. Taken together, our findings demonstrate that mEVs conjugated with IMTP effectively deliver miR30d to the pathological heart and thereby ameliorating cardiac hypertrophy and dysfunction via targeting GRK5-mediated signaling pathways.
Collapse
Affiliation(s)
- Lingjun Tong
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiyue Wang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yameng Zhang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Fengling Lai
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jiarun Xu
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wenchao Yin
- Department of Cardiology, Shandong First Medical Affiliated Shandong Provincial Hospital, Jinan, 250021, China
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore
| | - Guoyue Wei
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jie Yin
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Department of Cardiology, Shandong First Medical Affiliated Qianfoshan Hospital, Jinan 250013, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore
| | - Zhaoyang Wang
- Department of Cardiology, Shandong First Medical Affiliated Shandong Provincial Hospital, Jinan, 250021, China.
| | - Rong Huang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Tao Xu
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Guangzhou Laboratory, Guangzhou, 510005, China.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore; Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS), Singapore, 117599, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
6
|
Wang X, Mu X, Li X, Yang C, Cai Y, Liu C, Liu Z, He Z. Construction of a deep learning model and identification of the pivotal characteristics of FGF7- and MGST1- positive fibroblasts in heart failure post-myocardial infarction. Int J Biol Macromol 2025; 310:143171. [PMID: 40258553 DOI: 10.1016/j.ijbiomac.2025.143171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/05/2025] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Dysregulation of fibroblast function is closely associated with the occurrence of heart failure after myocardial infarction (post-MI HF). Myocardial fibrosis is a detrimental consequence of aberrant fibroblast activation and extracellular matrix deposition following myocardial infarction (MI). However, the heterogeneity of fibroblasts in normal cardiac tissue and heart failure tissue remains to be further investigated. We discovered that the abundance of FGF7+MGST1+ fibroblasts were down-regulated in post-MI HF according to scRNA-seq analysis. Key gene characteristics of FGF7+MGST1+ fibroblasts were uncovered through both differential expression analysis and hdWGCNA pipeline. Pseudotime analysis revealed that FGF7+MGST1+ fibroblasts were gradually decreased with the occurrence of heart failure. Cell-cell communication analysis indicated an enhanced secretory ability in FGF7+MGST1+ fibroblasts compared to other fibroblasts. Utilizing machine learning algorithms, we identified 17 feature genes of this cell population. A deep learning model capable of predicting heart failure was successfully built based on these feature genes and immune infiltration levels of post-MI HF. FGF7 was highly related to cardioprotective pathway terms, including "PI3K/AKT pathway" and "protein secretion". Parallelly, mendelian randomization analysis was adopted to better understand the causal relationships between feature genes and post-MI HF. Results indicated that MGST1 was causally associated with heart failure, consistent with single cell data. And the post-MI HF mouse model was constructed and qRT-PCR assays supported that both FGF7 and MGST1 were largely down-regulated in myocardial infarction area than other cardiac tissues. These findings provide new insights into the roles of FGF7+MGST1+ fibroblasts in post MI HF.
Collapse
Affiliation(s)
- Xicheng Wang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Xiaolan Mu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Xiuhua Li
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Chao Yang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Yongchao Cai
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Changcheng Liu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Zhongmin Liu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China.
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China.
| |
Collapse
|
7
|
Liu D, Wang X, Liu Z, Ding L, Liu M, Li T, Zeng S, Zheng M, Wang L, Zhang J, Zhang F, Li M, Liu G, Tang Y. Platelet Membrane and miR-181a-5p Doubly Optimized Nanovesicles Enhance Cardiac Repair Post-Myocardial Infarction through Macrophage Polarization. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16520-16532. [PMID: 40064701 PMCID: PMC11931480 DOI: 10.1021/acsami.4c19325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025]
Abstract
Macrophages play a crucial role in cardiac remodeling and prognosis after myocardial infarction (MI). Our previous studies have built a scalable method for preparing scaled stem cell nanovesicles (NVs) and demonstrated their remarkable reparative effects on ischemic heart disease. To further enhance the targeted reparative capabilities of the NVs toward injured myocardium, we employed a dual modification strategy involving platelet membrane coating and miR-181a-5p loading, creating a nanovesicle termed P-181-NV. This study aimed to investigate the efficacy of P-181-NV in targeted reparative interventions for damaged myocardium and to reveal the underlying mechanisms involved. After successful construction and characteristic analysis of P-181-NV, the in vivo tracking techniques demonstrated a significant enhancement in the targeting capacity of P-181-NV toward the injured myocardium. Moreover, P-181-NV showed marked improvements in cardiac function and remodeling as observed through ultrasound echocardiography and Masson's trichrome staining. Furthermore, P-181-NV significantly augmented myocardial cell viability, angiogenic potential, and the polarization ratio of the anti-inflammatory macrophages. The findings of this study underscore the pivotal role of platelet-membrane-coated and miR-181a-5p modified stem cell nanovesicles in facilitating postmyocardial infarction cardiac repair. By modulating macrophage polarization, P-181-NV offers a promising approach for enhancing the efficacy of targeted reparative interventions for damaged myocardium. These results contribute to our understanding of the potential of nanovesicles as therapeutic agents for cardiac repair and regeneration, presenting avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Dongyue Liu
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Xianyun Wang
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Zhao Liu
- Traditional
Chinese Medicine Processing Technology Innovation Center of Hebei
Province, School of Pharmacy, Hebei University
of Chinese Medicine, Shijiazhuang 050091, China
- International
Joint Research Center on Resource Utilization and Quality Evaluation
of Traditional Chinese Medicine of Hebei Province, Shijiazhuang 050091, China
| | - Lini Ding
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Mei Liu
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Tianshuo Li
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Shasha Zeng
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Mingqi Zheng
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Heart and Metabolism, Shijiazhuang 050031, Hebei Province, China
| | - Le Wang
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Heart and Metabolism, Shijiazhuang 050031, Hebei Province, China
| | - Jun Zhang
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Fan Zhang
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Meng Li
- College
of
Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Gang Liu
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Hebei
Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
- Hebei International
Joint Research Center for Structural Heart Disease, Shijiazhuang 050031, Hebei Province, China
| | - Yida Tang
- Department
of Cardiovascular Medicine, The First Hospital
of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
- Department
of Cardiology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
8
|
Kook T, Lee MY, Kwak TH, Jeong D, Sim DS, Jeong MH, Ahn Y, Kook H, Park WJ, Jang SP. Intratracheal Delivery of a Phospholamban Decoy Peptide Attenuates Cardiac Damage Following Myocardial Infarction. Int J Mol Sci 2025; 26:2649. [PMID: 40141290 PMCID: PMC11942360 DOI: 10.3390/ijms26062649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Heart failure (HF) remains a major cause of mortality worldwide. While novel approaches, including gene and cell therapies, show promise, efficient delivery methods for such biologics to the heart are critically needed. One emerging strategy is lung-to-heart delivery using nanoparticle (NP)-encapsulated biologics. This study examines the efficiency of delivering a therapeutic peptide conjugated to a cell-penetrating peptide (CPP) to the heart via the lung-to-heart route through intratracheal (IT) injection in mice. The CPP, a tandem repeat of NP2 (dNP2) derived from the human novel LZAP-binding protein (NLBP), facilitates intracellular delivery of the therapeutic payload. The therapeutic peptide, SE, is a decoy peptide designed to inhibit protein phosphatase 1 (PP1)-mediated dephosphorylation of phospholamban (PLN). Our results demonstrated that IT injection of dNP2-SE facilitated efficient delivery to the heart, with peak accumulation at 3 h post-injection. The administration of dNP2-SE significantly ameliorated morphological and functional deterioration of the heart under myocardial infarction. At the molecular level, dNP2-SE effectively prevented PLN dephosphorylation in the heart. Immunoprecipitation experiments further revealed that dNP2-SE binds strongly to PP1 and disrupts its interaction with PLN. Collectively, our findings suggest that lung-to-heart delivery of a CPP-conjugated therapeutic peptide, dNP2-SE, represents a promising approach for the treatment of HF.
Collapse
Affiliation(s)
- Taewon Kook
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mi-Young Lee
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Tae Hwan Kwak
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan 15588, Republic of Korea;
| | - Doo Sun Sim
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Woo Jin Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Seung Pil Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Center for Gene and Cell Therapy, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
9
|
Zhou X, Wang H, Yan B, Nie X, Chen Q, Yang X, Lei M, Guo X, Ouyang C, Ren Z. Ferroptosis in Cardiovascular Diseases and Ferroptosis-Related Intervention Approaches. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07642-5. [PMID: 39641901 DOI: 10.1007/s10557-024-07642-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Cardiovascular diseases (CVDs) are major public health problems that threaten the lives and health of individuals. The article has reviewed recent progresses about ferroptosis and ferroptosis-related intervention approaches for the treatment of CVDs and provided more references and strategies for targeting ferroptosis to prevent and treat CVDs. METHODS A comprehensive review was conducted using the literature researches. RESULTS AND DISCUSSION Many ferroptosis-targeted compounds and ferroptosis-related genes may be prospective targets for treating CVDs and our review provides a solid foundation for further studies about the detailed pathological mechanisms of CVDs. CONCLUSION There are challenges and limitations about the translation of ferroptosis-targeted potential therapies from experimental research to clinical practice. It warrants further exploration to pursure safer and more effective ferroptosis-targeted thereapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Xianpeng Zhou
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Hao Wang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Biao Yan
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xinwen Nie
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiaosong Yang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Min Lei
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiying Guo
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Changhan Ouyang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|