1
|
Wang Y, Wu Y, Jiang Y, Tan H, Guragain B, Nguyen T, Zhao J, Zhou Y, Nakada Y, Zhang J. Cardiomyocyte-Specific Overexpression of Activated Yes-Associated Protein Modified-RNA Promotes Cardiomyocyte Proliferation and Myocardial Regeneration. J Am Heart Assoc 2024; 13:e037120. [PMID: 39470057 DOI: 10.1161/jaha.124.037120] [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] [Received: 06/12/2024] [Accepted: 09/13/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND The proliferative capacity of cardiomyocytes in adult mammalian hearts is far too low to replace the cells that are lost to myocardial infarction. Both cardiomyocyte proliferation and myocardial regeneration can be improved via the overexpression of a constitutively active variant of YAP5SA (Yes-associated protein, 5SA [active] mutant), but persistent overexpression of proliferation-inducing genes could lead to hypertrophy and arrhythmia, whereas off-target expression in fibroblasts and macrophages could increase fibrosis and inflammation. METHODS AND RESULTS Transient overexpression of YAP5SA or GFP (green fluorescent protein; control) was targeted to cardiomyocytes via our cardiomyocyte-specific modified mRNA translation system (YAP5SACM-SMRTs or GFPCM-SMRTs, respectively). YAP5SA-cardiomyocyte specificity was confirmed via in vitro experiments in cardiomyocytes and cardiac fibroblasts that had been differentiated from human induced- pluripotent stem cells and in human umbilical-vein endothelial cells, and the regenerative potency of YAP5SACM-SMRTs was evaluated in a mouse myocardial infarction model. In cultured human induced-pluripotent stem cells-cardiomyocytes, YAP was abundantly expressed for 3 days after YAP5SACM-SMRTs administration and was accompanied by increases in the expression of markers for proliferation, before declining to near-background levels after day 7, whereas GFP fluorescence remained high from days 1 to 3 after GFPCM-SMRTs treatment and then slowly declined. GFP fluorescence was also observed in human induced-pluripotent stem cells-cardiac fibroblasts and human umbilical-vein endothelial cells on day 1 after GFPCM-SMRTs administration but declined substantially by day 3. In the mouse myocardial infarction model, echocardiographic assessments of left-ventricular ejection fraction and fractional shortening were significantly greater, whereas infarct sizes were significantly smaller in YAP5SACM-SMRTs-treated mice than in vehicle-treated control animals, and YAP5SACM-SMRTs appeared to promote cardiomyocyte proliferation. CONCLUSIONS The CM-SMRTs can be used to transiently and specifically overexpress YAP5SA in cardiomyocytes, and this treatment strategy significantly promoted cardiomyocyte proliferation and myocardial regeneration in a mouse myocardial infarction model.
Collapse
Affiliation(s)
- Yongyu Wang
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Yalin Wu
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Yu Jiang
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Huilan Tan
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Bijay Guragain
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Thanh Nguyen
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Jianli Zhao
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Yang Zhou
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Yuji Nakada
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
| | - Jianyi Zhang
- Department of Biomedical Engineering University of Alabama at Birmingham Birmingham AL USA
- Department of Medicine, Cardiovascular Disease University of Alabama at Birmingham Birmingham AL USA
| |
Collapse
|
2
|
Saddique MN, Qadri M, Ain NU, Farhan E, Shahid F, Benyamin J, Bashir MA, Jain H, Iqbal J. Safety and effectiveness of interference RNA (RNAi) based therapeutics in cardiac failure: A systematic review. Heart Lung 2024; 68:298-304. [PMID: 39214039 DOI: 10.1016/j.hrtlng.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Heart failure is a major worldwide health concern and leading cause of mortality. RNAi interventions hold promise for patients resistant to conventional drugs due to their off-target effects and lack of specificity. OBJECTIVES To examine the safety and effectiveness of RNAi therapeutics in treating heart failure. METHODS The PubMed, Embase, Scopus and Cochrane databases were searched using appropriate keyword from inception until December 31, 2023. A total of 14 studies fulfilling predefined selection criteria were included for qualitative synthesis. RESULTS We found that in patients with cardiac amyloidosis, patisiran and revusiran showed considerable improvements in cardiac output and left ventricular wall thickness. In animal studies, Nox2-siRNA showed effectiveness in regaining heart function. Furthermore, cardiomyocyte count and left ventricular function were improved by DUSP5 siRNA + T3 therapy and meg3 inhibition after myocardial infarction (MI). RNAi showed minimal adverse effects like peripheral neuropathy, hepatotoxicity, urinary tract infection, vaginal infection, diarrhea, abdominal pain arrhythmias, conduction disorders, and cardiotoxicity (LV wall thinning, heart failure) and improved cardiac biomarkers. CONCLUSION RNAi therapeutics are novel treatment option for improving cardiac function because their high target specificity, ability to target genes that conventional drugs struggle to reach and potential for long-lasting effects. Further research on optimizing delivery methods, improving target specificity, evaluating long-term safety profiles and cost-effectiveness to fully realize their potential.
Collapse
Affiliation(s)
| | - Maria Qadri
- Jinnah Sindh Medical University, Karachi 75510, Pakistan
| | - Noor Ul Ain
- FMH College of Medicine and Dentistry, Lahore 54000, Pakistan
| | - Eesha Farhan
- Karachi Institute of Medical Sciences, Karachi 75510, Pakistan
| | - Fatima Shahid
- King Edward Medical University, Lahore 54000, Pakistan
| | | | | | - Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Javed Iqbal
- Nursing Department, Communicable Disease Center-Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
3
|
Gao T, Xu G, Ma T, Lu X, Chen K, Luo H, Chen G, Song J, Ma X, Fu W, Zheng C, Xia X, Jiang J. ROS-Responsive Injectable Hydrogel Loaded with SLC7A11-modRNA Inhibits Ferroptosis and Mitigates Intervertebral Disc Degeneration in Rats. Adv Healthc Mater 2024; 13:e2401103. [PMID: 38691848 DOI: 10.1002/adhm.202401103] [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: 03/26/2024] [Revised: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Intervertebral disc degeneration (IVDD) is the primary cause of low back pain, with oxidative stress being a recognized factor that causes its development. Presently, low back pain imposes a significant global economic burden. However, the effectiveness of treatments for IVDD remains extremely limited. Therefore, this study aims to explore innovative and effective IVDD treatments by focusing on oxidative stress as a starting point. In this study, an injectable reactive oxygen species-responsive hydrogel (PVA-tsPBA@SLC7A11 modRNA) is developed, designed to achieve rapid loading and selective release of chemically synthesized modified mRNA (modRNA). SLC7A11 modRNA is specifically used to upregulate the expression of the ferroptosis marker SLC7A11. The local injection of PVA-tsPBA@SLC7A11 modRNA into the degenerated intervertebral disc (IVD) results in the cleavage of PVA-tsPBA, leading to the release of enclosed SLC7A11 modRNA. The extent of SLC7A11 modRNA release is directly proportional to the severity of IVDD, ultimately ameliorating IVDD by inhibiting ferroptosis in nucleus pulposus cells (NPCs). This study proposes an innovative system of PVA-tsPBA hydrogel-encapsulated modRNA, representing a potential novel treatment strategy for patients with early-stage IVDD.
Collapse
Affiliation(s)
- Tian Gao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Guangyu Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Tiancong Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Xiao Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Kaiwen Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Huanhuan Luo
- Department of Orthopaedics, The Second Hospital of Jiaxing, Jiaxing, Zhejiang Province, 314000, P. R. China
| | - Gang Chen
- Department of Orthopaedics, The Second Hospital of Jiaxing, Jiaxing, Zhejiang Province, 314000, P. R. China
| | - Jian Song
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Chaojun Zheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Fudan University, Shanghai, 200082, P. R. China
| |
Collapse
|
4
|
Soroudi S, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in cardiovascular diseases: Advances in genome editing and CAR T cell therapy. J Control Release 2024; 372:113-140. [PMID: 38876358 DOI: 10.1016/j.jconrel.2024.06.023] [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: 01/09/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality among non-communicable diseases. Current cardiac regeneration treatments have limitations and may lead to adverse reactions. Hence, innovative technologies are needed to address these shortcomings. Messenger RNA (mRNA) emerges as a promising therapeutic agent due to its versatility in encoding therapeutic proteins and targeting "undruggable" conditions. It offers low toxicity, high transfection efficiency, and controlled protein production without genome insertion or mutagenesis risk. However, mRNA faces challenges such as immunogenicity, instability, and difficulty in cellular entry and endosomal escape, hindering its clinical application. To overcome these hurdles, lipid nanoparticles (LNPs), notably used in COVID-19 vaccines, have a great potential to deliver mRNA therapeutics for CVDs. This review highlights recent progress in mRNA-LNP therapies for CVDs, including Myocardial Infarction (MI), Heart Failure (HF), and hypercholesterolemia. In addition, LNP-mediated mRNA delivery for CAR T-cell therapy and CRISPR/Cas genome editing in CVDs and the related clinical trials are explored. To enhance the efficiency, safety, and clinical translation of mRNA-LNPs, advanced technologies like artificial intelligence (AGILE platform) in RNA structure design, and optimization of LNP formulation could be integrated. We conclude that the strategies to facilitate the extra-hepatic delivery and targeted organ tropism of mRNA-LNPs (SORT, ASSET, SMRT, and barcoded LNPs) hold great prospects to accelerate the development and translation of mRNA-LNPs in CVD treatment.
Collapse
Affiliation(s)
- Setareh Soroudi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Kishore R, Magadum A. Cell-Specific mRNA Therapeutics for Cardiovascular Diseases and Regeneration. J Cardiovasc Dev Dis 2024; 11:38. [PMID: 38392252 PMCID: PMC10889436 DOI: 10.3390/jcdd11020038] [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: 12/12/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Cardiovascular diseases (CVDs) represent a significant global health burden, demanding innovative therapeutic approaches. In recent years, mRNA therapeutics have emerged as a promising strategy to combat CVDs effectively. Unlike conventional small-molecule drugs, mRNA therapeutics enable the direct modulation of cellular functions by delivering specific mRNA molecules to target cells. This approach offers unprecedented advantages, including the ability to harness endogenous cellular machinery for protein synthesis, thus allowing precise control over gene expression without insertion into the genome. This review summarizes the current status of the potential of cell-specific mRNA therapeutics in the context of cardiovascular diseases. First, it outlines the challenges associated with traditional CVD treatments and emphasizes the need for targeted therapies. Subsequently, it elucidates the underlying principles of mRNA therapeutics and the development of advanced delivery systems to ensure cell-specificity and enhanced efficacy. Notably, innovative delivery methods such as lipid nanoparticles and exosomes have shown promise in improving the targeted delivery of mRNA to cardiac cells, activated fibroblasts, and other relevant cell types. Furthermore, the review highlights the diverse applications of cell-specific mRNA therapeutics in addressing various aspects of cardiovascular diseases, including atherosclerosis, myocardial infarction, heart failure, and arrhythmias. By modulating key regulatory genes involved in cardiomyocyte proliferation, inflammation, angiogenesis, tissue repair, and cell survival, mRNA therapeutics hold the potential to intervene at multiple stages of CVD pathogenesis. Despite its immense potential, this abstract acknowledges the challenges in translating cell-specific mRNA therapeutics from preclinical studies to clinical applications like off-target effects and delivery. In conclusion, cell-specific mRNA therapeutics have emerged as a revolutionary gene therapy approach for CVD, offering targeted interventions with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Raj Kishore
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| | - Ajit Magadum
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
6
|
Wang AYL, Chang YC, Chen KH, Loh CYY. Potential Application of Modified mRNA in Cardiac Regeneration. Cell Transplant 2024; 33:9636897241248956. [PMID: 38715279 PMCID: PMC11080755 DOI: 10.1177/09636897241248956] [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: 11/08/2023] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 05/12/2024] Open
Abstract
Heart failure remains the leading cause of human death worldwide. After a heart attack, the formation of scar tissue due to the massive death of cardiomyocytes leads to heart failure and sudden death in most cases. In addition, the regenerative ability of the adult heart is limited after injury, partly due to cell-cycle arrest in cardiomyocytes. In the current post-COVID-19 era, urgently authorized modified mRNA (modRNA) vaccines have been widely used to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Therefore, modRNA-based protein replacement may act as an alternative strategy for improving heart disease. It is a safe, effective, transient, low-immunogenic, and integration-free strategy for in vivo protein expression, in addition to recombinant protein and stem-cell regenerative therapies. In this review, we provide a summary of various cardiac factors that have been utilized with the modRNA method to enhance cardiovascular regeneration, cardiomyocyte proliferation, fibrosis inhibition, and apoptosis inhibition. We further discuss other cardiac factors, modRNA delivery methods, and injection methods using the modRNA approach to explore their application potential in heart disease. Factors for promoting cardiomyocyte proliferation such as a cocktail of three genes comprising FoxM1, Id1, and Jnk3-shRNA (FIJs), gp130, and melatonin have potential to be applied in the modRNA approach. We also discuss the current challenges with respect to modRNA-based cardiac regenerative medicine that need to be overcome to apply this approach to heart disease. This review provides a short description for investigators interested in the development of alternative cardiac regenerative medicines using the modRNA platform.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuan-Hung Chen
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | |
Collapse
|
7
|
Qian B, Shen A, Huang S, Shi H, Long Q, Zhong Y, Qi Z, He X, Zhang Y, Hai W, Wang X, Cui Y, Chen Z, Xuan H, Zhao Q, You Z, Ye X. An Intrinsically Magnetic Epicardial Patch for Rapid Vascular Reconstruction and Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303033. [PMID: 37964406 PMCID: PMC10754083 DOI: 10.1002/advs.202303033] [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: 05/11/2023] [Revised: 08/30/2023] [Indexed: 11/16/2023]
Abstract
Myocardial infarction (MI) is a major cause of mortality worldwide. The major limitation of regenerative therapy for MI is poor cardiac retention of therapeutics, which results from an inefficient vascular network and poor targeting ability. In this study, a two-layer intrinsically magnetic epicardial patch (MagPatch) prepared by 3D printing with biocompatible materials like poly (glycerol sebacate) (PGS) is designed, poly (ε-caprolactone) (PCL), and NdFeB. The two-layer structure ensured that the MagPatch multifariously utilized the magnetic force for rapid vascular reconstruction and targeted drug delivery. MagPatch accumulates superparamagnetic iron oxide (SPION)-labelled endothelial cells, instantly forming a ready-implanted organization, and rapidly reconstructs a vascular network anastomosed with the host. In addition, the prefabricated vascular network within the MagPatch allowed for the efficient accumulation of SPION-labelled therapeutics, amplifying the therapeutic effects of cardiac repair. This study defined an extendable therapeutic platform for vascularization-based targeted drug delivery that is expected to assist in the progress of regenerative therapies in clinical applications.
Collapse
Affiliation(s)
- Bei Qian
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Ao Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Hongpeng Shi
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Xiaojun He
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yanna Cui
- Department of Pharmacology and Chemical BiologyShanghai Jiaotong University School of MedicineShanghai200000China
| | - Ziheng Chen
- School of Mechatronics Engineering and AutomationShanghai UniversityShanghai200000China
| | - Huixia Xuan
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| |
Collapse
|
8
|
Wagner N, Wagner KD. Molecular Mechanisms of Cardiac Development and Disease. Int J Mol Sci 2023; 24:ijms24108784. [PMID: 37240127 DOI: 10.3390/ijms24108784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
During development, the heart is the first organ to form and function [...].
Collapse
Affiliation(s)
- Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d'Azur, 06107 Nice, France
| | | |
Collapse
|