1
|
Guan Y, Wen J, Niu H, Zhai J, Dang Y, Guan J. Targeted delivery of engineered adipose-derived stem cell secretome to promote cardiac repair after myocardial infarction. J Control Release 2025; 383:113765. [PMID: 40274072 PMCID: PMC12145236 DOI: 10.1016/j.jconrel.2025.113765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Stem cell secretome offers a promising alternative to stem cell transplantation for treating myocardial infarction (MI). However, its clinical application faces two major challenges: how to enhance the levels of growth factors within the secretome to promote cardiac cell survival and vascularization, and how to efficiently deliver the secretome to the infarcted heart during the acute MI phase without risking rupture of the weakened myocardium. To address these challenges, we upregulated angiogenic growth factors in the secretome from adipose-derived stem cells (ADSC-secretome) by conditioning the cells under hypoxia and with insulin-like growth factor 1 (IGF-1). Our results show that exposure to 1 % O₂ condition significantly increased the expression of VEGF, bFGF, and PDGF-BB compared to 5 % O₂ condition. Co-treatment with IGF-1 further elevated the levels of these growth factors and, notably, reduced the secretion of pro-inflammatory cytokines such as TNFα, IL-1β, and IL-6 from the ADSCs. To rapidly and specifically deliver the secretome to the infarcted heart during acute MI, we encapsulated it within ischemia-targeting nanoparticles. These nanoparticles, designed for intravenous injection, preferentially accumulated in the infarcted region. The treatment significantly improved cardiac cell survival, tissue vascularization, and cardiac function. These findings suggest that ADSC secretome, enriched with angiogenic growth factors, holds strong potential for facilitating cardiac repair following MI.
Collapse
Affiliation(s)
- Ya Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA; Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jiaxing Wen
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA; Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Hong Niu
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA; Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jin Zhai
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Yu Dang
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA; Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jianjun Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA; Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
2
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [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: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Austria ES, Akhavan B. Polymeric nanoparticle synthesis for biomedical applications: advancing from wet chemistry methods to dry plasma technologies. NANOSCALE 2025. [PMID: 40391562 DOI: 10.1039/d5nr00436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Nanotechnology has introduced a transformative leap in healthcare over recent decades, particularly through nanoparticle-based drug delivery systems. Among these, polymeric nanoparticles (NPs) have gained significant attention due to their tuneable physicochemical properties for overcoming biological barriers. Their surfaces can be engineered with chemical functional groups and biomolecules for a wide range of biomedical applications, ranging from drug delivery to diagnostics. However, despite these advancements, the clinical translation and large-scale commercialization of polymeric NPs face significant challenges. This review uncovers these challenges by examining the interplay between structural design and payload interaction mode. It provides a critical evaluation of the current synthesis methods, beginning with conventional wet chemical techniques, and progressing to emerging dry plasma technologies, such as plasma polymerization. Special attention is given to plasma polymerized nanoparticles (PPNs), highlighting their potential as paradigm-shifting platforms for biomedical applications while identifying key areas for improvement. The review concludes with a forward-looking discussion on strategies to address key challenges, such as achieving regulatory approval and advancing clinical translation of polymeric NP-based therapies, offering unprecedented opportunities for next-generation nanomedicine.
Collapse
Affiliation(s)
- Elmer S Austria
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia.
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Behnam Akhavan
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia.
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
- School of Engineering, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
4
|
Chen H, Hu K, Tang Q, Wang J, Gu Q, Chen J, Hu J, Peng N, Guo M, Jiang Y, Xu Q, Xie J. CD248-targeted BBIR-T cell therapy against late-activated fibroblasts in cardiac repair after myocardial infarction. Nat Commun 2025; 16:2895. [PMID: 40148319 PMCID: PMC11950650 DOI: 10.1038/s41467-025-56703-2] [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: 11/03/2023] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
Excessive cardiac fibrosis is a key cause of heart failure and adverse ventricular remodeling after myocardial infarction. The abnormally activated fibroblasts after scar maturation are the chief culprit. Single-cell RNA sequencing of mouse cardiac interstitial cells after myocardial infarction depicts a late-activated fibroblast subpopulation F-Act and initially identifies its characteristic antigen CD248, which is also verified in human hearts. On this basis, we develop a CD248-targeted biotin-binding immune receptor T cell therapy against F-Act to correct cardiac repair disorders. In our study, the precise removal of F-Act after the scar matured effectively inhibits fibrotic expansion in the peri-infarct zone and improves cardiac function. This therapy provides an idea for the treatment of cardiac fibrosis and also promotes the application of engineered T cells to non-tumor diseases.
Collapse
Affiliation(s)
- Haiting Chen
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Ke Hu
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Qi Tang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, No.101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Junzhuo Wang
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Qianyu Gu
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Jiayu Chen
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No.158 Wuyang Road, Enshi, Hubei, 445099, China
| | - Ningxin Peng
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Meng Guo
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Yaohui Jiang
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Jun Xie
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China.
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
5
|
Cook M, Lal S, Hume RD. Transcriptional, proteomic and metabolic drivers of cardiac regeneration. Heart 2025:heartjnl-2024-325442. [PMID: 40037760 DOI: 10.1136/heartjnl-2024-325442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
Following injury, many organs are capable of rapid regeneration of necrotic tissue to regain normal function. In contrast, the damaged heart typically replaces tissue with a collagen-rich scar, due to the limited regenerative capacity of its functional contractile cardiomyocytes (CMs). However, this regenerative capacity varies dramatically during development and between species. Furthermore, studies have shown that cardiac regeneration can be enhanced to return contractile function to the damaged heart following myocardial infarction (MI). In this review, we outline the proliferative capacity of CMs in utero, postnatally and in adulthood. We also describe the regenerative capacity of the heart following MI injury. Finally, we focus on the various therapeutic strategies that aim to augment cardiac regeneration in preclinical animal models. These include altering transcripts, microRNAs, extracellular matrix proteins and inducing metabolic rewiring. Together, these therapies aim to return function to the damaged heart and potentially improve the lives of the millions of heart failure patients currently suffering worldwide.
Collapse
Affiliation(s)
- Matthew Cook
- School of Biomedical Sciences, Faculty of Health & Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sean Lal
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- School of Medical Sciences, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Robert D Hume
- School of Medical Sciences, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Camperdown, New South Wales, Australia
| |
Collapse
|
6
|
Zhang X, Shao S, Li Q, Wang Y, Kong M, Zhang C. Roles of Autophagy, Mitophagy, and Mitochondria in Left Ventricular Remodeling after Myocardial Infarction. Rev Cardiovasc Med 2025; 26:28195. [PMID: 40160572 PMCID: PMC11951495 DOI: 10.31083/rcm28195] [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: 11/12/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
This review examines the mechanisms of left ventricular dysfunction, focusing on the interplay between ventricular remodeling, autophagy, and mitochondrial dysfunction following myocardial infarction. Left ventricular dysfunction directly affects the heart's pumping efficiency and can lead to severe clinical outcomes, including heart failure. After myocardial infarction, the left ventricle may suffer from weakened contractility, diastolic dysfunction, and cardiac remodeling, progressing to heart failure. Thus, this article discusses the pathophysiological processes involved in ventricular remodeling, including the injury and repair of infarcted and non-infarcted myocardia, adaptive changes, and specific changes in left ventricular systolic and diastolic functions. Furthermore, the role of autophagy in maintaining cellular energy homeostasis, clearing dysfunctional mitochondria, and the key role of mitochondrial dysfunction in heart failure is addressed. Finally, this article discusses therapeutic strategies targeting mitochondrial dysfunction and enhancing mitophagy, providing clinicians and researchers with the latest insights and future research directions.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Shuai Shao
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Qiuting Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yi Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Mowei Kong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
7
|
Zhang XZ, Li QL, Tang TT, Cheng X. Emerging Role of Macrophage-Fibroblast Interactions in Cardiac Homeostasis and Remodeling. JACC Basic Transl Sci 2025; 10:113-127. [PMID: 39958468 PMCID: PMC11830265 DOI: 10.1016/j.jacbts.2024.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 02/18/2025]
Abstract
As major noncardiomyocyte components in cardiac tissues, macrophages and fibroblasts play crucial roles in maintaining cardiac homeostasis, orchestrating reparative responses after cardiac injuries, facilitating adaptive cardiac remodeling, and contributing to adverse cardiac remodeling, owing to their inherent heterogeneity and plasticity. Recent advances in research methods have yielded novel insights into the intricate interactions between macrophages and fibroblasts in the cardiac context. This review aims to comprehensively examine the molecular mechanisms governing macrophage-fibroblast interactions in cardiac homeostasis and remodeling, emphasize recent advancements in the field, and offer an evaluation from a translational standpoint.
Collapse
Affiliation(s)
- Xu-Zhe Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin-Lin Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting-Ting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [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: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
9
|
Adao DMT, Ching C, Fish JE, Simmons CA, Billia F. Endothelial cell-cardiomyocyte cross-talk: understanding bidirectional paracrine signaling in cardiovascular homeostasis and disease. Clin Sci (Lond) 2024; 138:1395-1419. [PMID: 39492693 DOI: 10.1042/cs20241084] [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: 06/09/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
To maintain homeostasis in the heart, endothelial cells and cardiomyocytes engage in dynamic cross-talk through paracrine signals that regulate both cardiac development and function. Here, we review the paracrine signals that endothelial cells release to regulate cardiomyocyte growth, hypertrophy and contractility, and the factors that cardiomyocytes release to influence angiogenesis and vascular tone. Dysregulated communication between these cell types can drive pathophysiology of disease, as seen in ischemia-reperfusion injury, diabetes, maladaptive hypertrophy, and chemotherapy-induced cardiotoxicity. Investingating the role of cross-talk is critical in developing an understanding of tissue homeostasis, regeneration, and disease pathogenesis, with the potential to identify novel targets for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Doris M T Adao
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario, Canada, M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave., Toronto, Ontario, Canada, M5G 1M1
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Peter Munk Cardiac Centre, University Health Network, 585 University Ave., Toronto, Ontario, Canada, M5G 2N2
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario, Canada, M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave., Toronto, Ontario, Canada, M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Rd., Toronto, Ontario, Canada, M5S 3G8
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Peter Munk Cardiac Centre, University Health Network, 585 University Ave., Toronto, Ontario, Canada, M5G 2N2
| |
Collapse
|
10
|
Waldmann M, Bohner M, Baghnavi A, Riedel B, Seidenstuecker M. Release kinetics of growth factors loaded into β-TCP ceramics in an in vitro model. Front Bioeng Biotechnol 2024; 12:1441547. [PMID: 39398641 PMCID: PMC11466813 DOI: 10.3389/fbioe.2024.1441547] [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: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction β-TCP ceramics are bone replacement materials that have recently been tested as a drug delivery system that can potentially be applied to endogenous substances like growth factors found in blood platelets to facilitate positive attributes. Methods In this work, we used flow chamber loading to load β-TCP dowels with blood suspensions of platelet-rich plasma (PRP), platelet-poor plasma (PPP), or buffy coat (BC) character. PRP and BC platelet counts were adjusted to the same level by dilution. Concentrations of TGF-β1, PDGF-AB, and IGF-1 from dowel-surrounding culture medium were subsequently determined using ELISA over 5 days. The influence of alginate was additionally tested to modify the release. Results Concentrations of TGF-β1 and PDGF-AB increased and conclusively showed a release from platelets in PRP and BC compared to PPP. The alginate coating reduced the PDGF-AB release but did not reduce TGF-β1 and instead even increased TGF-β1 in the BC samples. IGF-1 concentrations were highest in PPP, suggesting circulating levels rather than platelet release as the driving factor. Alginate samples tended to have lower IGF-1 concentrations, but the difference was not shown to be significant. Discussion The release of growth factors from different blood suspensions was successfully demonstrated for β-TCP as a drug delivery system with release patterns that correspond to PRP activation after Ca2+-triggered activation. The release pattern was partially modified by alginate coating.
Collapse
Affiliation(s)
- Marco Waldmann
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Anna Baghnavi
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Bianca Riedel
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Michael Seidenstuecker
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
11
|
Gao J, Guo H, Li J, Zhan M, You Y, Xin G, Liu Z, Fan X, Gao Q, Liu J, Zhang Y, Fu J. Buyang Huanwu decoction ameliorates myocardial injury and attenuates platelet activation by regulating the PI3 kinase/Rap1/integrin α(IIb)β(3) pathway. Chin Med 2024; 19:109. [PMID: 39160598 PMCID: PMC11331649 DOI: 10.1186/s13020-024-00976-0] [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: 03/07/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Buyang Huanwu Decoction (BYHWD) is a traditional Chinese medicine to treat the syndrome of qi deficiency and blood stasis. Platelets play an important role in regulating thrombus and inflammation after ischemic injury, studies have shown that BYHWD regulate myocardial fibrosis and exert anti-inflammatory effects through IL-17 and TLR4 pathways, but the mechanism of platelet activation by BYHWD in stable coronary heart disease is still unknown. In the present study, model of left anterior descending coronary artery ligation was applied to investigate the mechanisms of BYHWD on modulating platelets hyperreactivity and heart function after fibrosis of ischemic myocardial infarction (MI). METHODS Myocardial infarction model was constructed by ligation of the left anterior descending coronary artery. The rats were randomly divided into five groups: sham, model, MI with aspirin (positive), MI with a low dosage of BYHWD (BYHWD-ld) and MI with a high dosage of BYHWD (BYHWD-hd) for 28 days. RESULTS Coronary artery ligation prominently induced left ventricle dysfunction, increased cardiomyocyte fibrosis, which was accompanied by platelets with hyperreactivity, and high levels of inflammatory factors. BYHWD obviously reversed cardiac dysfunction and fibrosis, increased the thickness of the left ventricular wall, and inhibited aggregation ratio and CD62p expression. BYHWD restored the mitochondrial respiration of platelets after MI, concomitant with an increased telomere expression and decreased inflammation. According to the result of transcriptome sequencing, we found that 106 differentially expressed genes compared model with BYHWD treatment. Enrichment analysis screened out the Ras-related protein Rap-1 (Rap1) signaling pathway and platelet activation biological function. Quantitative real-time PCR and Western blotting were applied to found that BYHWD reduced the expression of Rap1/PI3K-Akt/Src-CDC42 genes and attenuated the overactivity of PI3 kinase/Rap1/integrin α(IIb)β(3) pathway. CONCLUSION BYHWD reduced inflammation and platelet activation via the PI3 kinase/Rap1/integrin α(IIb)β(3) pathway and improved heart function after MI.
Collapse
Affiliation(s)
- Jiaming Gao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Hao Guo
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Junmei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Min Zhan
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Yue You
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Gaojie Xin
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Zixin Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Xiaodi Fan
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Qinghe Gao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China.
| | - Yehao Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China.
| | - Jianhua Fu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Chinese Materia, Courtyard No. 1, Xiyuan Playground, Haidian District, Beijing, China.
| |
Collapse
|
12
|
Voronkov NS, Maslov LN, Vyshlov EV, Mukhomedzyanov AV, Ryabov VV, Derkachev IA, Kan A, Gusakova SV, Gombozhapova AE, Panteleev OO. Do platelets protect the heart against ischemia/reperfusion injury or exacerbate cardiac ischemia/reperfusion injury? The role of PDGF, VEGF, and PAF. Life Sci 2024; 347:122617. [PMID: 38608835 DOI: 10.1016/j.lfs.2024.122617] [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: 01/25/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is one of the main causes of death. It is quite obvious that there is an urgent need to develop new approaches for treatment of AMI. OBJECTIVE This review analyzes data on the role of platelets in the regulation of cardiac tolerance to ischemia/reperfusion (I/R). METHODS It was performed a search of topical articles using PubMed databases. FINDINGS Platelets activated by a cholesterol-enriched diet, thrombin, and myocardial ischemia exacerbate I/R injury of the heart. The P2Y12 receptor antagonists, remote ischemic postconditioning and conditioning alter the properties of platelets. Platelets acquire the ability to increase cardiac tolerance to I/R. Platelet-derived growth factors (PDGFs) increase tolerance of cardiomyocytes and endothelial cells to I/R. PDGF receptors (PDGFRs) were found in cardiomyocytes and endothelial cells. PDGFs decrease infarct size and partially abrogate adverse postinfarction remodeling. Protein kinase C, phosphoinositide 3-kinase, and Akt involved in the cytoprotective effect of PDGFs. Vascular endothelial growth factor increased cardiac tolerance to I/R and alleviated adverse postinfarction remodeling. The platelet-activating factor (PAF) receptor inhibitors increase cardiac tolerance to I/R in vivo. PAF enhances cardiac tolerance to I/R in vitro. It is possible that PAF receptor inhibitors could protect the heart by blocking PAF receptor localized outside the heart. PAF protects the heart through activation of PAF receptor localized in cardiomyocytes or endothelial cells. Reactive oxygen species and kinases are involved in the cardioprotective effect of PAF. CONCLUSION Platelets play an important role in the regulation of cardiac tolerance to I/R.
Collapse
Affiliation(s)
- Nikita S Voronkov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Leonid N Maslov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia.
| | - Evgeniy V Vyshlov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Alexander V Mukhomedzyanov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Vyacheslav V Ryabov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Ivan A Derkachev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Artur Kan
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Svetlana V Gusakova
- Department of Biophysics and Functional Diagnostics, Siberian State Medical University, 634050 Tomsk, Russia
| | - Alexandra E Gombozhapova
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Oleg O Panteleev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| |
Collapse
|
13
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
14
|
Deshmukh T, Selvakumar D, Thavapalachandran S, Archer O, Figtree GA, Feneley M, Grieve SM, Thomas L, Pathan F, Chong JJH. Correlation of Noninvasive Cardiac MRI Measures of Left Ventricular Myocardial Function and Invasive Pressure-Volume Parameters in a Porcine Ischemia-Reperfusion Model. Radiol Cardiothorac Imaging 2024; 6:e230252. [PMID: 38842454 PMCID: PMC11211950 DOI: 10.1148/ryct.230252] [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: 08/22/2023] [Revised: 03/24/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
Purpose To assess the correlation between noninvasive cardiac MRI-derived parameters with pressure-volume (PV) loop data and evaluate changes in left ventricular function after myocardial infarction (MI). Materials and Methods Sixteen adult female swine were induced with MI, with six swine used as controls and 10 receiving platelet-derived growth factor-AB (PDGF-AB). Load-independent measures of cardiac function, including slopes of end-systolic pressure-volume relationship (ESPVR) and preload recruitable stroke work (PRSW), were obtained on day 28 after MI. Cardiac MRI was performed on day 2 and day 28 after infarct. Global longitudinal strain (GLS) and global circumferential strain (GCS) were measured. Ventriculo-arterial coupling (VAC) was derived from PV loop and cardiac MRI data. Pearson correlation analysis was performed. Results GCS (r = 0.60, P = .01), left ventricular ejection fraction (LVEF) (r = 0.60, P = .01), and cardiac MRI-derived VAC (r = 0.61, P = .01) had a significant linear relationship with ESPVR. GCS (r = 0.75, P < .001) had the strongest significant linear relationship with PRSW, followed by LVEF (r = 0.67, P = .005) and cardiac MRI-derived VAC (r = 0.60, P = .01). GLS was not significantly correlated with ESPVR or PRSW. There was a linear correlation (r = 0.82, P < .001) between VAC derived from cardiac MRI and from PV loop data. GCS (-3.5% ± 2.3 vs 0.5% ± 1.4, P = .007) and cardiac MRI-derived VAC (-0.6 ± 0.6 vs 0.3 ± 0.3, P = .001) significantly improved in the animals treated with PDGF-AB 28 days after MI compared with controls. Conclusion Cardiac MRI-derived parameters of MI correlated with invasive PV measures, with GCS showing the strongest correlation. Cardiac MRI-derived measures also demonstrated utility in assessing therapeutic benefit using PDGF-AB. Keywords: Cardiac MRI, Myocardial Infarction, Pressure Volume Loop, Strain Imaging, Ventriculo-arterial Coupling Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
- Tejas Deshmukh
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Dinesh Selvakumar
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Sujitha Thavapalachandran
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Oliver Archer
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Gemma A. Figtree
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Michael Feneley
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Stuart M. Grieve
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Liza Thomas
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | | | | |
Collapse
|
15
|
Selvakumar D, Clayton ZE, Prowse A, Dingwall S, Kim SK, Reyes L, George J, Shah H, Chen S, Leung HHL, Hume RD, Tjahjadi L, Igoor S, Skelton RJP, Hing A, Paterson H, Foster SL, Pearson L, Wilkie E, Marcus AD, Jeyaprakash P, Wu Z, Chiu HS, Ongtengco CFJ, Mulay O, McArthur JR, Barry T, Lu J, Tran V, Bennett R, Kotake Y, Campbell T, Turnbull S, Gupta A, Nguyen Q, Ni G, Grieve SM, Palpant NJ, Pathan F, Kizana E, Kumar S, Gray PP, Chong JJH. Cellular heterogeneity of pluripotent stem cell-derived cardiomyocyte grafts is mechanistically linked to treatable arrhythmias. NATURE CARDIOVASCULAR RESEARCH 2024; 3:145-165. [PMID: 39196193 PMCID: PMC11358004 DOI: 10.1038/s44161-023-00419-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/26/2023] [Indexed: 08/29/2024]
Abstract
Preclinical data have confirmed that human pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can remuscularize the injured or diseased heart, with several clinical trials now in planning or recruitment stages. However, because ventricular arrhythmias represent a complication following engraftment of intramyocardially injected PSC-CMs, it is necessary to provide treatment strategies to control or prevent engraftment arrhythmias (EAs). Here, we show in a porcine model of myocardial infarction and PSC-CM transplantation that EAs are mechanistically linked to cellular heterogeneity in the input PSC-CM and resultant graft. Specifically, we identify atrial and pacemaker-like cardiomyocytes as culprit arrhythmogenic subpopulations. Two unique surface marker signatures, signal regulatory protein α (SIRPA)+CD90-CD200+ and SIRPA+CD90-CD200-, identify arrhythmogenic and non-arrhythmogenic cardiomyocytes, respectively. Our data suggest that modifications to current PSC-CM-production and/or PSC-CM-selection protocols could potentially prevent EAs. We further show that pharmacologic and interventional anti-arrhythmic strategies can control and potentially abolish these arrhythmias.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Andrew Prowse
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, Queensland, Australia
| | - Steve Dingwall
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, Queensland, Australia
| | - Sul Ki Kim
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Leila Reyes
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Jacob George
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Haisam Shah
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Siqi Chen
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Halina H L Leung
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Robert D Hume
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Laurentius Tjahjadi
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Sindhu Igoor
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Rhys J P Skelton
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Alfred Hing
- Department of Cardiothoracic Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | - Hugh Paterson
- Sydney Imaging, Core Research Facility, the University of Sydney, Sydney, New South Wales, Australia
| | - Sheryl L Foster
- Department of Radiology, Westmead Hospital, Westmead, New South Wales, Australia
- Sydney School of Health Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia
| | - Lachlan Pearson
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Emma Wilkie
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Alan D Marcus
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
| | - Prajith Jeyaprakash
- Department of Cardiology, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Zhixuan Wu
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Han Shen Chiu
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Cherica Felize J Ongtengco
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, Queensland, Australia
| | - Onkar Mulay
- Genomics and Machine Learning Lab, Division of Genetics and Genomics, Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Jeffrey R McArthur
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, UNSW, Darlinghurst, New South Wales, Australia
| | - Tony Barry
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Juntang Lu
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Vu Tran
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Richard Bennett
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Yasuhito Kotake
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Timothy Campbell
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Samual Turnbull
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Anunay Gupta
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Quan Nguyen
- Genomics and Machine Learning Lab, Division of Genetics and Genomics, Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Guiyan Ni
- Genomics and Machine Learning Lab, Division of Genetics and Genomics, Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Stuart M Grieve
- Imaging and Phenotyping Laboratory, Faculty of Medicine and Health, Charles Perkins Centre, the University of Sydney, Sydney, New South Wales, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, Queensland, Australia
| | - Faraz Pathan
- Department of Cardiology, Nepean Hospital, Kingswood, New South Wales, Australia
- Sydney Medical School, Charles Perkins Centre Nepean, Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia
| | - Eddy Kizana
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Saurabh Kumar
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Peter P Gray
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, Queensland, Australia
| | - James J H Chong
- Centre for Heart Research, the Westmead Institute for Medical Research, the University of Sydney, Westmead, New South Wales, Australia.
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
16
|
Zhang B, He Z, Zhao H, Gao H, Zhang Z, Gao Z, Ke K. Evaluating the efficacy of recombinant human growth factors in scar remodelling for patients with facial soft tissue injuries. Int Wound J 2024; 21:e14649. [PMID: 38272796 PMCID: PMC10789918 DOI: 10.1111/iwj.14649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Facial soft tissue injuries, often resulting in scarring, pose a challenge in reconstructive and aesthetic surgery due to the need for functional and aesthetic restoration. This study evaluates the efficacy of recombinant human growth factors (rhGFs) in scar remodelling for such injuries. A retrospective evaluation was conducted from January 2020 to January 2023, involving 100 patients with facial soft tissue injuries. Participants were divided equally into a control group, receiving standard cosmetic surgical repair, and an observation group, treated with rhGFs supplemented cosmetic surgery. The study assessed scar characteristics (pigmentation, pliability, vascularity, height), hospital stay duration, tissue healing time, complication rates and patient satisfaction. The observation group demonstrated significant improvements in all scar characteristics, with notably better pigmentation, pliability, vascularity and height compared with the control group. The rhGF treatment also resulted in reduced hospital stay duration and faster tissue healing. Notably, the total complication rate was significantly lower in the observation group (10%) compared with the control group (34%). Additionally, patient satisfaction levels were higher in the observation group, with 98% combined satisfaction compared with 76% in the control group. The application of rhGFs in treating facial soft tissue injuries significantly enhances scar remodelling, expedites healing, reduces complications and improves patient satisfaction. These findings establish rhGFs as a valuable tool in the management of facial soft tissue injuries, highlighting their potential in improving both functional and aesthetic outcomes.
Collapse
Affiliation(s)
- Baiyu Zhang
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Yunnan Province Clinical Research Center for Chronic Kidney DiseaseKunmingChina
| | - Zonghai He
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Hui Zhao
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Hongbin Gao
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Zhiying Zhang
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Zhenhua Gao
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Yunnan Province Clinical Research Center for Chronic Kidney DiseaseKunmingChina
| | - Kunbin Ke
- The First Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
17
|
Murtha LA, Hardy SA, Mabotuwana NS, Bigland MJ, Bailey T, Raguram K, Liu S, Ngo DT, Sverdlov AL, Tomin T, Birner-Gruenberger R, Hume RD, Iismaa SE, Humphreys DT, Patrick R, Chong JJH, Lee RJ, Harvey RP, Graham RM, Rainer PP, Boyle AJ. Fibulin-3 is necessary to prevent cardiac rupture following myocardial infarction. Sci Rep 2023; 13:14995. [PMID: 37696945 PMCID: PMC10495317 DOI: 10.1038/s41598-023-41894-9] [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: 06/13/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023] Open
Abstract
Despite the high prevalence of heart failure in the western world, there are few effective treatments. Fibulin-3 is a protein involved in extracellular matrix (ECM) structural integrity, however its role in the heart is unknown. We have demonstrated, using single cell RNA-seq, that fibulin-3 was highly expressed in quiescent murine cardiac fibroblasts, with expression highest prior to injury and late post-infarct (from ~ day-28 to week-8). In humans, fibulin-3 was upregulated in left ventricular tissue and plasma of heart failure patients. Fibulin-3 knockout (Efemp1-/-) and wildtype mice were subjected to experimental myocardial infarction. Fibulin-3 deletion resulted in significantly higher rate of cardiac rupture days 3-6 post-infarct, indicating a weak and poorly formed scar, with severe ventricular remodelling in surviving mice at day-28 post-infarct. Fibulin-3 knockout mice demonstrated less collagen deposition at day-3 post-infarct, with abnormal collagen fibre-alignment. RNA-seq on day-3 infarct tissue revealed upregulation of ECM degradation and inflammatory genes, but downregulation of ECM assembly/structure/organisation genes in fibulin-3 knockout mice. GSEA pathway analysis showed enrichment of inflammatory pathways and a depletion of ECM organisation pathways. Fibulin-3 originates from cardiac fibroblasts, is upregulated in human heart failure, and is necessary for correct ECM organisation/structural integrity of fibrotic tissue to prevent cardiac rupture post-infarct.
Collapse
Affiliation(s)
- Lucy A Murtha
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Sean A Hardy
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Nishani S Mabotuwana
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Mark J Bigland
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Taleah Bailey
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Kalyan Raguram
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Saifei Liu
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Doan T Ngo
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Aaron L Sverdlov
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Robert D Hume
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - David T Humphreys
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Randall J Lee
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Edyth and Eli Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
- School of Biotechnology and Molecular Bioscience, UNSW, Sydney, Kensington, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J Boyle
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia.
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia.
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
18
|
Clayton ZE, Santos M, Shah H, Lu J, Chen S, Shi H, Kanagalingam S, Michael PL, Wise SG, Chong JJH. Plasma polymerized nanoparticles are a safe platform for direct delivery of growth factor therapy to the injured heart. Front Bioeng Biotechnol 2023; 11:1127996. [PMID: 37409168 PMCID: PMC10319252 DOI: 10.3389/fbioe.2023.1127996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction: Heart failure due to myocardial infarction is a progressive and debilitating condition, affecting millions worldwide. Novel treatment strategies are desperately needed to minimise cardiomyocyte damage after myocardial infarction and to promote repair and regeneration of the injured heart muscle. Plasma polymerized nanoparticles (PPN) are a new class of nanocarriers which allow for a facile, one-step functionalization with molecular cargo. Methods: Here, we conjugated platelet-derived growth factor AB (PDGF-AB) to PPN, engineering a stable nano-formulation, as demonstrated by optimal hydrodynamic parameters, including hydrodynamic size distribution, polydisperse index (PDI) and zeta potential, and further demonstrated safety and bioactivity in vitro and in vivo. We delivered PPN-PDGF-AB to human cardiac cells and directly to the injured rodent heart. Results: We found no evidence of cytotoxicity after delivery of PPN or PPN-PDGFAB to cardiomyocytes in vitro, as determined through viability and mitochondrial membrane potential assays. We then measured contractile amplitude of human stem cell derived cardiomyocytes and found no detrimental effect of PPN on cardiomyocyte contractility. We also confirmed that PDGF-AB remains functional when bound to PPN, with PDGF receptor alpha positive human coronary artery vascular smooth muscle cells and cardiac fibroblasts demonstrating migratory and phenotypic responses to PPN-PDGF-AB in the same manner as to unbound PDGF-AB. In our rodent model of PPN-PDGF-AB treatment after myocardial infarction, we found a modest improvement in cardiac function in PPN-PDGF-AB treated hearts compared to those treated with PPN, although this was not accompanied by changes in infarct scar size, scar composition, or border zone vessel density. Discussion: These results demonstrate safety and feasibility of the PPN platform for delivery of therapeutics directly to the myocardium. Future work will optimize PPN-PDGF-AB formulations for systemic delivery, including effective dosage and timing to enhance efficacy and bioavailability, and ultimately improve the therapeutic benefits of PDGF-AB in the treatment of heart failure cause by myocardial infarction.
Collapse
Affiliation(s)
- Zoë E. Clayton
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Miguel Santos
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Haisam Shah
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Juntang Lu
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - Siqi Chen
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Han Shi
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | | | - Praveesuda L. Michael
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Steven G. Wise
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - James J. H. Chong
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| |
Collapse
|
19
|
Mouton AJ. Platelet-Derived Growth Factors: A New Therapeutic Opportunity for Treating Cardiac Fibrosis? JACC Basic Transl Sci 2023; 8:675-676. [PMID: 37426528 PMCID: PMC10322860 DOI: 10.1016/j.jacbts.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
20
|
Cao Y, Redd MA, Fang C, Mizikovsky D, Li X, Macdonald PS, King GF, Palpant NJ. New Drug Targets and Preclinical Modelling Recommendations for Treating Acute Myocardial Infarction. Heart Lung Circ 2023:S1443-9506(23)00139-7. [PMID: 37230806 DOI: 10.1016/j.hlc.2022.12.015] [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: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 05/27/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide and the primary underlying risk factor for heart failure. Despite decades of research and clinical trials, there are no drugs currently available to prevent organ damage from acute ischaemic injuries of the heart. In order to address the increasing global burden of heart failure, drug, gene, and cell-based regeneration technologies are advancing into clinical testing. In this review we highlight the burden of disease associated with AMI and the therapeutic landscape based on market analyses. New studies revealing the role of acid-sensitive cardiac ion channels and other proton-gated ion channels in cardiac ischaemia are providing renewed interest in pre- and post-conditioning agents with novel mechanisms of action that may also have implications for gene- and cell-based therapeutics. Furthermore, we present guidelines that couple new cell technologies and data resources with traditional animal modelling pipelines to help de-risk drug candidates aimed at treating AMI. We propose that improved preclinical pipelines and increased investment in drug target identification for AMI is critical to stem the increasing global health burden of heart failure.
Collapse
Affiliation(s)
- Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Chen Fang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Peter S Macdonald
- Cardiopulmonary Transplant Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
21
|
Voges HK, Foster SR, Reynolds L, Parker BL, Devilée L, Quaife-Ryan GA, Fortuna PRJ, Mathieson E, Fitzsimmons R, Lor M, Batho C, Reid J, Pocock M, Friedman CE, Mizikovsky D, Francois M, Palpant NJ, Needham EJ, Peralta M, Monte-Nieto GD, Jones LK, Smyth IM, Mehdiabadi NR, Bolk F, Janbandhu V, Yao E, Harvey RP, Chong JJH, Elliott DA, Stanley EG, Wiszniak S, Schwarz Q, James DE, Mills RJ, Porrello ER, Hudson JE. Vascular cells improve functionality of human cardiac organoids. Cell Rep 2023:112322. [PMID: 37105170 DOI: 10.1016/j.celrep.2023.112322] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/13/2023] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
Crosstalk between cardiac cells is critical for heart performance. Here we show that vascular cells within human cardiac organoids (hCOs) enhance their maturation, force of contraction, and utility in disease modeling. Herein we optimize our protocol to generate vascular populations in addition to epicardial, fibroblast, and cardiomyocyte cells that self-organize into in-vivo-like structures in hCOs. We identify mechanisms of communication between endothelial cells, pericytes, fibroblasts, and cardiomyocytes that ultimately contribute to cardiac organoid maturation. In particular, (1) endothelial-derived LAMA5 regulates expression of mature sarcomeric proteins and contractility, and (2) paracrine platelet-derived growth factor receptor β (PDGFRβ) signaling from vascular cells upregulates matrix deposition to augment hCO contractile force. Finally, we demonstrate that vascular cells determine the magnitude of diastolic dysfunction caused by inflammatory factors and identify a paracrine role of endothelin driving dysfunction. Together this study highlights the importance and role of vascular cells in organoid models.
Collapse
Affiliation(s)
- Holly K Voges
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Simon R Foster
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Liam Reynolds
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lynn Devilée
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Gregory A Quaife-Ryan
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Ellen Mathieson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Mary Lor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Christopher Batho
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Janice Reid
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mark Pocock
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Clayton E Friedman
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Mathias Francois
- The Centenary Institute, David Richmond Program for Cardiovascular Research: Gene Regulation and Editing, Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Elise J Needham
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Marina Peralta
- Australian Regenerative Medicine Institute. Monash University, Clayton, VIC 3800, Australia
| | | | - Lynelle K Jones
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedical Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedical Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Francesca Bolk
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ernestene Yao
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia; School of Biotechnology and Biomolecular Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia; Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Sophie Wiszniak
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Medical School, The University of Sydney, Sydney, 2010 NSW, Australia
| | - Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, The Royal Children's Hospital, Melbourne, VIC 3052, Australia.
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
22
|
Yin X, Yin X, Pan X, Zhang J, Fan X, Li J, Zhai X, Jiang L, Hao P, Wang J, Chen Y. Post-myocardial infarction fibrosis: Pathophysiology, examination, and intervention. Front Pharmacol 2023; 14:1070973. [PMID: 37056987 PMCID: PMC10086160 DOI: 10.3389/fphar.2023.1070973] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiac fibrosis plays an indispensable role in cardiac tissue homeostasis and repair after myocardial infarction (MI). The cardiac fibroblast-to-myofibroblast differentiation and extracellular matrix collagen deposition are the hallmarks of cardiac fibrosis, which are modulated by multiple signaling pathways and various types of cells in time-dependent manners. Our understanding of the development of cardiac fibrosis after MI has evolved in basic and clinical researches, and the regulation of fibrotic remodeling may facilitate novel diagnostic and therapeutic strategies, and finally improve outcomes. Here, we aim to elaborate pathophysiology, examination and intervention of cardiac fibrosis after MI.
Collapse
Affiliation(s)
- Xiaoying Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Pan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyu Zhang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinhui Fan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiaxin Li
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lijun Jiang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Panpan Hao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
23
|
Sun Y, Ma M, Cao D, Zheng A, Zhang Y, Su Y, Wang J, Xu Y, Zhou M, Tang Y, Liu Y, Ma T, Fan A, Zhang X, Zhu Q, Qin J, Mo C, Xu Y, Zhang L, Xu D, Yue R. Inhibition of Fap Promotes Cardiac Repair by Stabilizing BNP. Circ Res 2023; 132:586-600. [PMID: 36756875 DOI: 10.1161/circresaha.122.320781] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Myocardial infarction (MI) elicits cardiac fibroblast activation and extracellular matrix (ECM) deposition to maintain the structural integrity of the heart. Recent studies demonstrate that Fap (fibroblast activation protein)-a prolyl-specific serine protease-is an important marker of activated cardiac fibroblasts after MI. METHODS Left ventricle and plasma samples from patients and healthy donors were used to analyze the expression level of FAP and its prognostic value. Echocardiography and histological analysis of heart sections were used to analyze cardiac functions, scar formation, ECM deposition and angiogenesis after MI. RNA-Sequencing, biochemical analysis, cardiac fibroblasts (CFs) and endothelial cells co-culture were used to reveal the molecular and cellular mechanisms by which Fap regulates angiogenesis. RESULTS We found that Fap is upregulated in patient cardiac fibroblasts after cardiac injuries, while plasma Fap is downregulated and functions as a prognostic marker for cardiac repair. Genetic or pharmacological inhibition of Fap in mice significantly improved cardiac function after MI. Histological and transcriptomic analyses showed that Fap inhibition leads to increased angiogenesis in the peri-infarct zone, which promotes ECM deposition and alignment by cardiac fibroblasts and prevents their overactivation, thereby limiting scar expansion. Mechanistically, we found that BNP (brain natriuretic peptide) is a novel substrate of Fap that mediates postischemic angiogenesis. Fap degrades BNP to inhibit vascular endothelial cell migration and tube formation. Pharmacological inhibition of Fap in Nppb (encoding pre-proBNP) or Npr1 (encoding the BNP receptor)-deficient mice showed no cardioprotective effects, suggesting that BNP is a physiological substrate of Fap. CONCLUSIONS This study identifies Fap as a negative regulator of cardiac repair and a potential drug target to treat MI. Inhibition of Fap stabilizes BNP to promote angiogenesis and cardiac repair.
Collapse
Affiliation(s)
- Yuxi Sun
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.).,Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (Y. Sun, A.Z., M.Z., L.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Mengqiu Ma
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Dandan Cao
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Ancheng Zheng
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (Y. Sun, A.Z., M.Z., L.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Yiying Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Yang Su
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Yanhua Xu
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.).,Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Mi Zhou
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (Y. Sun, A.Z., M.Z., L.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Yansong Tang
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Yifan Liu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Teng Ma
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Aoyuan Fan
- Department of Cardiac Surgery, Ruijin Hospital (A.F.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaoying Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Qiaoling Zhu
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Jiachen Qin
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Chunyang Mo
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.)
| | - Yawei Xu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Li Zhang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (Y. Sun, A.Z., M.Z., L.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Dachun Xu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y. Sun, M.M., Y. Su, Yanhua Xu, Y.T., Y.L., T.M., Yawei Xu, D.X.)
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, China (D.C., Y.Z., J.W., Yanhua Xu, X.Z., Q.Z., J.Q., C.M., R.Y.).,Shanghai Institute of Stem Cell Research and Clinical Translation, China (R.Y.)
| |
Collapse
|
24
|
Hume RD, Deshmukh T, Doan T, Shim WJ, Kanagalingam S, Tallapragada V, Rashid F, Marcuello M, Blessing D, Selvakumar D, Raguram K, Pathan F, Graham D, Ounzain S, Kizana E, Harvey RP, Palpant NJ, Chong JJ. PDGF-AB Reduces Myofibroblast Differentiation Without Increasing Proliferation After Myocardial Infarction. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
25
|
Kim JJH, Parajuli S, Sinha A, Mahamdeh M, van den Boomen M, Coll-Font J, Chen LS, Fan Y, Eder RA, Phipps K, Yuan S, Nguyen C. Pocket CLARITY enables distortion-mitigated cardiac microstructural tissue characterization of large-scale specimens. Front Cardiovasc Med 2022; 9:1037500. [PMID: 36451924 PMCID: PMC9701703 DOI: 10.3389/fcvm.2022.1037500] [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: 09/06/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
Molecular phenotyping by imaging of intact tissues has been used to reveal 3D molecular and structural coherence in tissue samples using tissue clearing techniques. However, clearing and imaging of cardiac tissue remains challenging for large-scale (>100 mm3) specimens due to sample distortion. Thus, directly assessing tissue microstructural geometric properties confounded by distortion such as cardiac helicity has been limited. To combat sample distortion, we developed a passive CLARITY technique (Pocket CLARITY) that utilizes a permeable cotton mesh pocket to encapsulate the sample to clear large-scale cardiac swine samples with minimal tissue deformation and protein loss. Combined with light sheet auto-fluorescent and scattering microscopy, Pocket CLARITY enabled the characterization of myocardial microstructural helicity of cardiac tissue from control, heart failure, and myocardial infarction in swine. Pocket CLARITY revealed with high fidelity that transmural microstructural helicity of the heart is significantly depressed in cardiovascular disease (CVD), thereby revealing new insights at the tissue level associated with impaired cardiac function.
Collapse
Affiliation(s)
- Joan J. H. Kim
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Shestruma Parajuli
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Aman Sinha
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Mohammed Mahamdeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Maaike van den Boomen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States,Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Jaume Coll-Font
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Lily Shi Chen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Yiling Fan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Robert A. Eder
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Kellie Phipps
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Christopher Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, United States,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States,Department of Medicine, Harvard Medical School, Boston, MA, United States,Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, United States,Cardiovascular Innovation Research Center, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States,*Correspondence: Christopher Nguyen,
| |
Collapse
|
26
|
Comparative assessment of motion averaged free-breathing or breath-held cardiac magnetic resonance imaging protocols in a porcine myocardial infarction model. Sci Rep 2022; 12:3727. [PMID: 35260600 PMCID: PMC8904807 DOI: 10.1038/s41598-022-07566-w] [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: 07/18/2021] [Accepted: 02/11/2022] [Indexed: 11/08/2022] Open
Abstract
Breath-held (BH) cardiac magnetic resonance imaging (CMR) is the gold standard for volumetric quantification. However, large animals for pre-clinical research are unable to voluntarily breath-hold, necessitating general anaesthesia and mechanical ventilation, increasing research costs and affecting cardiovascular physiology. Conducting CMR in lightly sedated, free-breathing (FB) animal subjects is an alternative strategy which can overcome these constraints, however, may result in poorer image quality due to breathing motion artefact. We sought to assess the reproducibility of CMR metrics between FB and BH CMR in a porcine model of ischaemic cardiomyopathy. FB or BH CMR was performed in 38 porcine subjects following percutaneous induction of myocardial infarction. Analysis was performed by two independent, blinded observers according to standard reporting guidelines. Subjective and objective image quality was significantly improved in the BH cohort (image quality score: 3.9/5 vs. 2.4/5; p < 0.0001 and myocardium:blood pool intensity ratio: 2.6-3.3 vs. 1.9-2.3; p < 0.001), along with scan acquisition time (4 min 06 s ± 1 min 55 s vs. 8 min 53 s ± 2 min 39 s; p < 0.000). Intra- and inter-observer reproducibility of volumetric analysis was substantially improved in BH scans (correlation coefficients: 0.94-0.99 vs. 0.76-0.91; coefficients of variation: < 5% in BH and > 5% in FB; Bland-Altman limits of agreement: < 10 in BH and > 10 in FB). Interstudy variation between approaches was used to calculate sample sizes, with BH CMR resulting in greater than 85% reduction in animal numbers required to show clinically significant treatment effects. In summary, BH porcine CMR produces superior image quality, shorter scan acquisition, greater reproducibility, and requires smaller sample sizes for pre-clinical trials as compared to FB acquisition.
Collapse
|
27
|
Yao Y, Li A, Wang S, Lu Y, Xie J, Zhang H, Zhang D, Ding J, Wang Z, Tu C, Shen L, Zhuang L, Zhu Y, Gao C. Multifunctional elastomer cardiac patches for preventing left ventricle remodeling after myocardial infarction in vivo. Biomaterials 2022; 282:121382. [DOI: 10.1016/j.biomaterials.2022.121382] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/18/2022] [Indexed: 01/10/2023]
|
28
|
Zhao P, Li B, Li Y, Chen L, Wang H, Ye L. DNA-Templated ultrasmall bismuth sulfide nanoparticles for photoacoustic imaging of myocardial infarction. J Colloid Interface Sci 2022; 615:475-484. [PMID: 35150955 DOI: 10.1016/j.jcis.2022.01.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
Abstract
Photoacoustic imaging (PAI) has shown great clinical potential in diagnosing various diseases due to its noninvasive, cost-effective, and real-time imaging properties but is limited by the lack of contrast agents with high sensitivity for deep tissue imaging. Here, DNA-templated ultrasmall bismuth sulfide (Bi2S3) nanoparticles (NPs) were reported as a photoacoustic (PA) probe for imaging myocardial infarction. We present a simple synthesis strategy of ultrasmall NPs via self-assembly of single-stranded DNA (ssDNA)/metal ion complexes. The in vivo imaging results showed a dramatically enhanced PA signal in the region of myocardial infarction after intravenous injection of DNA-Bi2S3 NPs in the myocardial ischaemia/reperfusion (I/R) mouse model. Further near infrared fluorescence imaging indicated that Bi2S3 NPs mainly accumulated in the infarcted area, leading to enhancement of PA signals. Moreover, such hybrid NPs possess a well-defined nanostructure, superior photobleaching resistance, excellent water dispersibility and negligible acute toxicity. These results not only demonstrate that ultrasmall DNA-Bi2S3 NPs are a potent PA probe for imaging the infarcted region but also provide a new avenue for preparing ultrasmall-sized PA probes by using ssDNA as a template.
Collapse
Affiliation(s)
- Peng Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR. China
| | - Bing Li
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR. China
| | - Yingxu Li
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR. China
| | - Leshan Chen
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR. China
| | - Hao Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR. China.
| | - Ling Ye
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR. China
| |
Collapse
|
29
|
Bone morphogenetic protein 1.3 inhibition decreases scar formation and supports cardiomyocyte survival after myocardial infarction. Nat Commun 2022; 13:81. [PMID: 35013172 PMCID: PMC8748453 DOI: 10.1038/s41467-021-27622-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022] Open
Abstract
Despite the high prevalence of ischemic heart diseases worldwide, no antibody-based treatment currently exists. Starting from the evidence that a specific isoform of the Bone Morphogenetic Protein 1 (BMP1.3) is particularly elevated in both patients and animal models of myocardial infarction, here we assess whether its inhibition by a specific monoclonal antibody reduces cardiac fibrosis. We find that this treatment reduces collagen deposition and cross-linking, paralleled by enhanced cardiomyocyte survival, both in vivo and in primary cultures of cardiac cells. Mechanistically, we show that the anti-BMP1.3 monoclonal antibody inhibits Transforming Growth Factor β pathway, thus reducing myofibroblast activation and inducing cardioprotection through BMP5. Collectively, these data support the therapeutic use of anti-BMP1.3 antibodies to prevent cardiomyocyte apoptosis, reduce collagen deposition and preserve cardiac function after ischemia. Here the authors show that a monoclonal antibody against a soluble isoform of Bone Morphogenetic Protein 1 prevents cardiac cell death, reducing fibrosis and preserving cardiac function after myocardial ischemia.
Collapse
|
30
|
Wei X, Chen S, Xie T, Chen H, Jin X, Yang J, Sahar S, Huang H, Zhu S, Liu N, Yu C, Zhu P, Wang W, Zhang W. An MMP-degradable and conductive hydrogel to stabilize HIF-1α for recovering cardiac functions. Am J Cancer Res 2022; 12:127-142. [PMID: 34987638 PMCID: PMC8690911 DOI: 10.7150/thno.63481] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/24/2021] [Indexed: 01/12/2023] Open
Abstract
Rationale: Although a few injectable hydrogels have shown a reliable biosafety and a moderate promise in treating myocardial infarction (MI), the updated hydrogel systems with an on-demand biodegradation and multi-biofunctions to deliver therapeutic drug would achieve more prominent efficacy in the future applications. In this report, a conductive and injectable hydrogel crosslinked by matrix metalloproteinase-sensitive peptides (MMP-SP) was rationally constructed to stabilize hypoxia-inducible factor-1α (HIF-1α) to recover heart functions after MI. Methods: Firstly, tetraaniline (TA) was incorporated into partially oxidized alginate (ALG-CHO) to endow the hydrogels with conductivity. The 1,4-dihydrophenonthrolin-4-one-3-carboxylic acid (DPCA) nanodrug was manufactured with high drug loading capacity and decorated with polymerized dopamine (PDA) to achieve a stable release of the drug. Both ALG-CHO and DPCA@PDA can be cross-linked by thiolated hyaluronic acid (HA-SH) and thiolated MMP-SP to construct a MMP-degradable and conductive hydrogel. After administration in the infarcted heart of rats, echocardiographic assessments, histological evaluation, and RT-PCR were used to evaluate therapeutic effects of hydrogels. Results: The cell viability and the results of subcutaneous implantation verify a good cytocompatibility and biocompatibility of the resulting hydrogels. The hydrogel shows remarkable strength in decreasing the expression of inflammatory factors, maintaining a high level of HIF-1α to promote the vascularization, and promoting the expression of junctional protein connexin 43. Meanwhile, the multifunctional hydrogels greatly reduce the infarcted area (by 33.8%) and improve cardiac functions dramatically with ejection fraction (EF) and fractional shortening (FS) being increased by 31.3% and 19.0%, respectively. Conclusion: The as-prepared hydrogels in this report achieve a favorable therapeutic effect, offering a promising therapeutic strategy for treating heart injury.
Collapse
|
31
|
Li J, Zhao Y, Zhu W. Targeting angiogenesis in myocardial infarction: Novel therapeutics (Review). Exp Ther Med 2022; 23:64. [PMID: 34934435 PMCID: PMC8649855 DOI: 10.3892/etm.2021.10986] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myocardial infarction (AMI) remains the main cause of mortality worldwide. Despite surgery and medical treatment, the non-regeneration of dead cardiomyocytes and the limited contractile ability of scar tissue can lead to heart failure. Therefore, restoring blood flow in the infarcted area is important for the repair of myocardial injury. The objective of the present review was to summarize the factors influencing angiogenesis after AMI, and to describe the application of angiogenesis for cardiac repair. Collectively, this review may be helpful for relevant studies and to provide insight into future therapeutic applications in clinical practice.
Collapse
Affiliation(s)
- Jiejie Li
- Jiangsu Key Laboratory of Medical Science and Laboratory of Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yuanyuan Zhao
- Jiangsu Key Laboratory of Medical Science and Laboratory of Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wei Zhu
- Jiangsu Key Laboratory of Medical Science and Laboratory of Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
32
|
Herrera-Vargas AK, García-Rodríguez E, Olea-Flores M, Mendoza-Catalán MA, Flores-Alfaro E, Navarro-Tito N. Pro-angiogenic activity and vasculogenic mimicry in the tumor microenvironment by leptin in cancer. Cytokine Growth Factor Rev 2021; 62:23-41. [PMID: 34736827 DOI: 10.1016/j.cytogfr.2021.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
The acquired ability to induce the formation of a functional vasculature is a hallmark of cancer. Blood vessels in tumors are formed through various mechanisms, among the most important in cancer biology, angiogenesis, and vasculogenic mimicry have been described. Leptin is one of the main adipokines secreted by adipocytes in normal breast tissue and the tumor microenvironment. Here, we provide information on the relationship between leptin and the development of angiogenesis and vasculogenic mimicry in different types of cancer. Here, we report that leptin activates different pathways such as JAK-STAT3, MAPK/ERK, PKC, JNK, p38, and PI3K-Akt to induce the expression of various angiogenic factors and vasculogenic mimicry. In vivo models, leptin induces blood vessel formation through the PI3K-Akt-mTOR pathway. Interestingly, the relationship between leptin and vasculogenic mimicry was more significant in breast cancer. The information obtained suggests that leptin could be playing an essential role in tumor survival and metastasis through the induction of vascular mechanisms such as angiogenesis and vasculogenic mimicry; thus, leptin-induced pathways could be suggested as a promising therapeutic target.
Collapse
Affiliation(s)
- Ana K Herrera-Vargas
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Eduardo García-Rodríguez
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Miguel A Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO, 39090, Mexico.
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO 39087, Mexico.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| |
Collapse
|
33
|
Pluripotent stem cell-derived mesenchymal stromal cells improve cardiac function and vascularity after myocardial infarction. Cytotherapy 2021; 23:1074-1084. [PMID: 34588150 DOI: 10.1016/j.jcyt.2021.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have been shown to improve cardiac function after injury and are the subject of ongoing clinical trials. In this study, the authors tested the cardiac regenerative potential of an induced pluripotent stem cell-derived MSC (iPSC-MSC) population (Cymerus MSCs) in a rat model of myocardial ischemia-reperfusion (I/R). Furthermore, the authors compared this efficacy with bone marrow-derived MSCs (BM-MSCs), which are the predominant cell type in clinical trials. METHODS Four days after myocardial I/R injury, rats were randomly assigned to (i) a Cymerus MSC group (n = 15), (ii) a BM-MSC group (n = 15) or (iii) a vehicle control group (n = 14). For cell-treated animals, a total of 5 × 106 cells were injected at three sites within the infarcted left ventricular (LV) wall. RESULTS One month after cell transplantation, Cymerus MSCs improved LV function (assessed by echocardiography) compared with vehicle and BM-MSCs. Interestingly, Cymerus MSCs enhanced angiogenesis without sustained engraftment or significant impact on infarct scar size. Suggesting safety, Cymerus MSCs had no effect on inducible tachycardia or the ventricular scar heterogeneity that provides a substrate for cardiac re-entrant circuits. CONCLUSIONS The authors here demonstrate that intra-myocardial administration of iPSC-MSCs (Cymerus MSCs) provide better therapeutic effects compared with conventional BM-MSCs in a rodent model of myocardial I/R. Because of its manufacturing scalability, iPSC-MSC therapy offers an exciting opportunity for an "off-the-shelf" stem cell therapy for cardiac repair.
Collapse
|
34
|
Kalra K, Eberhard J, Farbehi N, Chong JJ, Xaymardan M. Role of PDGF-A/B Ligands in Cardiac Repair After Myocardial Infarction. Front Cell Dev Biol 2021; 9:669188. [PMID: 34513823 PMCID: PMC8424099 DOI: 10.3389/fcell.2021.669188] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/20/2021] [Indexed: 01/06/2023] Open
Abstract
Platelet-derived growth factors (PDGFs) are powerful inducers of cellular mitosis, migration, angiogenesis, and matrix modulation that play pivotal roles in the development, homeostasis, and healing of cardiac tissues. PDGFs are key signaling molecules and important drug targets in the treatment of cardiovascular disease as multiple researchers have shown that delivery of recombinant PDGF ligands during or after myocardial infarction can reduce mortality and improve cardiac function in both rodents and porcine models. The mechanism involved cannot be easily elucidated due to the complexity of PDGF regulatory activities, crosstalk with other protein tyrosine kinase activators, and diversity of the pathological milieu. This review outlines the possible roles of PDGF ligands A and B in the healing of cardiac tissues including reduced cell death, improved vascularization, and improved extracellular matrix remodeling to improve cardiac architecture and function after acute myocardial injury. This review may highlight the use of recombinant PDGF-A and PDGF-B as a potential therapeutic modality in the treatment of cardiac injury.
Collapse
Affiliation(s)
- Kunal Kalra
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Joerg Eberhard
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nona Farbehi
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - James J Chong
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Munira Xaymardan
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
35
|
Rashid FN, Clayton ZE, Ogawa M, Perdomo J, Hume RD, Kizana E, Chong JJH. Platelet derived growth factor-A (Pdgf-a) gene transfer modulates scar composition and improves left ventricular function after myocardial infarction. Int J Cardiol 2021; 341:24-30. [PMID: 34265313 DOI: 10.1016/j.ijcard.2021.07.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/18/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Novel therapies that can limit or reverse damage caused by myocardial infarction (MI) could ease the increasing burden of heart failure. In this regard Platelet Derived Growth Factor (PDGF) has been previously shown to contribute to cardiac repair after MI. Here, we use a rodent model of MI and recombinant adeno-associated virus 9 (rAAV9)-mediated gene transfer to overexpress Pdgf-a in the injured heart and assess its therapeutic potential. METHODS AND RESULTS Sprague Dawley rats underwent temporary occlusion of the left anterior descending coronary artery, followed immediately by systemic delivery of 1 × 10^11 vector genomes of either rAAV9 Pdgf-a or rAAV9 Empty vector (control). At day 28 post-MI echocardiography showed significantly improved left ventricular (LV) function (fractional shortening) after rAAV9 Pdgf-a (0.394 ± 0.019%) treatment vs control (0.304 ± 0.018%). Immunohistochemical analysis demonstrated significantly increased capillary and arteriolar density in the infarct border zone of rAAV9 Pdgf-a treated hearts together with a significant reduction in infarct scar size (rAAV9 Pdgf-a 6.09 ± 0.94% vs Empty 12.45 ± 0.92%). Western blot and qPCR analyses confirmed overexpression of PDGF-A and showed upregulation of smooth muscle alpha actin (Acta2), collagen type III alpha 1 (Col3a1) and lysyl oxidase (Lox) genes in rAAV9 Pdgf-a treated infarcts. CONCLUSION Overexpression of Pdgf-a in the post-MI heart can modulate scar composition and improve LV function. Our study highlights the potential of rAAV gene transfer of Pdgf-a as a cardio-reparative therapy.
Collapse
Affiliation(s)
- Fairooj N Rashid
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia
| | - Zoë E Clayton
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia
| | - Masahito Ogawa
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia
| | - Jose Perdomo
- Haematology Research Unit, St George and Sutherland Clinical School, University of New South Wales, NSW, Australia
| | - Robert D Hume
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia
| | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia; Department of Cardiology, Westmead Hospital, Westmead 2145, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead 2145, NSW, Australia; The University of Sydney, Australia; Department of Cardiology, Westmead Hospital, Westmead 2145, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|
36
|
Wang D, Li T, Xu Y, Yang X, He M, Zhang Z, Wu W, Yan Y. [Platelet-rich plasma alleviates myocardial ischemia-reperfusion injury in rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:775-782. [PMID: 34134967 DOI: 10.12122/j.issn.1673-4254.2021.05.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the protective effect of platelet-rich plasma (PRP) against acute myocardial ischemiareperfusion (IR) injury and the possible mechanism. OBJECTIVE Aortic blood samples were collected from 10 SD rats to prepare PRP, in which the concentrations of platelet-derived growth factor-BB (PDGF-BB) and transforming growth factor-β1 (TGF-β1) were measured. Cell models of IR injury were established in primary cultures of neonatal SD rat cardiomyocytes by exposing the cells to 3 h of hypoxia. The cells were then reoxygenated and co-cultured with 1%, 5%, 10%, and 20% volume of PRP for 12 h, and the changes in cell viability was assessed. Immunofluorescence staining of the cardiomyocytes was performed, and the cellular expression of AMPK and its phosphorylation level were detected. The effects of PRP on the proliferation and migration of rat aortic endothelial cells (RAOECs) were examined. In a SD rat model of myocardial IR injury, 100 μL of PRP (n= 20) or normal saline (n=20) was injected at 4 sites around the ligation site immediately after cardiac reperfusion. One day after the injection, 6 rats were selected from each group for TTC staining of the myocardial tissues and measurement of troponin Ⅰ content. One week later, the cardiac function of the remaining rats was assessed by echocardiography, and HE staining of the myocardial tissues was performed. The effect of PRP treatment for 24 h on polarization of M1 and M2 macrophages was also examined by flow cytometry in RAW264.7 cells after hypoxic exposure for 3 h. OBJECTIVE The concentrations of PDGF-BB and TGF-β1 were significantly higher in PRP than in whole blood. Addition of 1% volume of PRP significantly reduced death of the cardiomyocytes following reoxygenation, and this effect was closely related with the activation of AMPK. Treatment with PRP obviously promoted the proliferation and migration of RAOECs. In rat models of acute myocardial IR injury, injections of PRP significantly reduced the infarct size and troponin Ⅰ concentration as compared with saline injection (P < 0.001). One week after PRP injection, the rats showed significantly improved cardiac function with a lowered level of inflammatory response in comparison with the rats with saline injection. In RAW264.7 cells with hypoxic exposure, treatment with PRP obviously decreased the number of M1 macrophages and increase the number of M2 macrophages. OBJECTIVE PRP can improve acute myocardial IR injury in rats by phosphorylating AMPK and regulating macrophage polarization, which produces a protective immunomodulatory effect on the ischemic myocardial tissues.
Collapse
Affiliation(s)
- D Wang
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.,Translational Research Centre of Regenerative Medicine and 3D Printing, Guangzhou Medical University, Guangzhou 510150, China.,State Key Laboratory of Organ Failure Research, Department of Pathophysiology, Guangzhou 510515, China
| | - T Li
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.,State Key Laboratory of Organ Failure Research, Department of Pathophysiology, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory for Shock and Microcirculation Research, Southern Medical University, Guangzhou 510515, China
| | - Y Xu
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.,Guangdong Provincial Key Laboratory for Shock and Microcirculation Research, Southern Medical University, Guangzhou 510515, China
| | - X Yang
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.,Guangdong Provincial Key Laboratory for Shock and Microcirculation Research, Southern Medical University, Guangzhou 510515, China
| | - M He
- State Key Laboratory of Organ Failure Research, Department of Pathophysiology, Guangzhou 510515, China
| | - Z Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Guangzhou Medical University, Guangzhou 510150, China
| | - W Wu
- Guangdong Provincial Key Laboratory for Shock and Microcirculation Research, Southern Medical University, Guangzhou 510515, China
| | - Y Yan
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.,Translational Research Centre of Regenerative Medicine and 3D Printing, Guangzhou Medical University, Guangzhou 510150, China.,State Key Laboratory of Organ Failure Research, Department of Pathophysiology, Guangzhou 510515, China
| |
Collapse
|
37
|
Nguyen PD, de Bakker DEM, Bakkers J. Cardiac regenerative capacity: an evolutionary afterthought? Cell Mol Life Sci 2021; 78:5107-5122. [PMID: 33950316 PMCID: PMC8254703 DOI: 10.1007/s00018-021-03831-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
Cardiac regeneration is the outcome of the highly regulated interplay of multiple processes, including the inflammatory response, cardiomyocyte dedifferentiation and proliferation, neovascularization and extracellular matrix turnover. Species-specific traits affect these injury-induced processes, resulting in a wide variety of cardiac regenerative potential between species. Indeed, while mammals are generally considered poor regenerators, certain amphibian and fish species like the zebrafish display robust regenerative capacity post heart injury. The species-specific traits underlying these differential injury responses are poorly understood. In this review, we will compare the injury induced processes of the mammalian and zebrafish heart, describing where these processes overlap and diverge. Additionally, by examining multiple species across the animal kingdom, we will highlight particular traits that either positively or negatively affect heart regeneration. Last, we will discuss the possibility of overcoming regeneration-limiting traits to induce heart regeneration in mammals.
Collapse
Affiliation(s)
- Phong D Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
38
|
Contreras O, Córdova-Casanova A, Brandan E. PDGF-PDGFR network differentially regulates the fate, migration, proliferation, and cell cycle progression of myogenic cells. Cell Signal 2021; 84:110036. [PMID: 33971280 DOI: 10.1016/j.cellsig.2021.110036] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022]
Abstract
Platelet-derived growth factors (PDGFs) regulate embryonic development, tissue regeneration, and wound healing through their binding to PDGF receptors, PDGFRα and PDGFRβ. However, the role of PDGF signaling in regulating muscle development and regeneration remains elusive, and the cellular and molecular responses of myogenic cells are understudied. Here, we explore the PDGF-PDGFR gene expression changes and their involvement in skeletal muscle myogenesis and myogenic fate. By surveying bulk RNA sequencing and single-cell profiling data of skeletal muscle stem cells, we show that myogenic progenitors and muscle stem cells differentially express PDGF ligands and PDGF receptors during myogenesis. Quiescent adult muscle stem cells and myoblasts preferentially express PDGFRβ over PDGFRα. Remarkably, cell culture- and injury-induced muscle stem cell activation altered PDGF family gene expression. In myoblasts, PDGF-AB and PDGF-BB treatments activate two pro-chemotactic and pro-mitogenic downstream transducers, RAS-ERK1/2 and PI3K-AKT. PDGFRs inhibitor AG1296 inhibited ERK1/2 and AKT activation, myoblast migration, proliferation, and cell cycle progression induced by PDGF-AB and PDGF-BB. We also found that AG1296 causes myoblast G0/G1 cell cycle arrest. Remarkably, PDGF-AA did not promote a noticeable ERK1/2 or AKT activation, myoblast migration, or expansion. Also, myogenic differentiation reduced the expression of both PDGFRα and PDGFRβ, whereas forced PDGFRα expression impaired myogenesis. Thus, our data highlight PDGF signaling pathway to stimulate satellite cell proliferation aiming to enhance skeletal muscle regeneration and provide a deeper understanding of the role of PDGF signaling in non-fibroblastic cells.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington 2052, Australia; Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; Fundación Ciencia & Vida, 7780272 Santiago, Chile
| |
Collapse
|
39
|
Youssef ME, El-Mas MM, Abdelrazek HM, El-Azab MF. α7-nAChRs-mediated therapeutic angiogenesis accounts for the advantageous effect of low nicotine doses against myocardial infarction in rats. Eur J Pharmacol 2021; 898:173996. [PMID: 33684450 DOI: 10.1016/j.ejphar.2021.173996] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 12/20/2022]
Abstract
Angiogenesis accelerates tissue regeneration in a variety of ischemic conditions including myocardial infarction (MI). Here we tested the hypothesis that angiogenesis induced by α7-nicotinic acetylcholine receptors (α7-nAChRs) mitigates histopathological, electrocardiographic, and molecular consequences of MI in rats. These profiles were evaluated in the isoprenaline (85 mg/kg/day i. p. For 2 days) MI rat model treated with or without nicotine or PHA-543613 (PHA, selective α7-nAChR agonist). Isoprenaline-insulted rats showed (i) ECG signs of MI such as significant ST-segment elevations and prolonged QT-intervals, (ii) deteriorated left ventricular histopathological scoring and elevated inflammatory cell infiltration, (iii) reduced immunohistochemical expression of cardiac CD34, a surrogate marker of capillary density, (iv) decreased cardiac expression of iNOS and α7-nAChRs, and (v) adaptive increases in cardiac HO-1 expression and plasma angiogenic markers such as vascular endothelial growth factor (VEGF) and nitric oxide (NO). These effects of isoprenaline, except cardiac iNOS and α7-nAChRs downregulation, were ameliorated in rats treated with a low dose (20 μg/kg/day s. c. For 16 days) of nicotine or PHA. We also show that concurrent α7-nAChR blockade by methyllycaconitine (MLA, 40 μg/kg/day, for 16 days) reversed the ECG, histopathological, and capillary density effects of nicotine, thereby reinforcing the advantageous cardioprotective and anti-ischemic roles of α7-nAChRs in this setting. The observed results showed promising effects on isoprenaline induced myocardial damage. In conclusion, the activation of α7-nAChRs by doses of nicotine or PHA in the microgram scale promotes neovascularization and offers a promising therapeutic strategy for MI. CATEGORY: Cardiovascular Pharmacology.
Collapse
Affiliation(s)
- Mahmoud E Youssef
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait
| | - Heba M Abdelrazek
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Mona F El-Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
40
|
Sharma P, Wang X, Ming CLC, Vettori L, Figtree G, Boyle A, Gentile C. Considerations for the Bioengineering of Advanced Cardiac In Vitro Models of Myocardial Infarction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2003765. [PMID: 33464713 DOI: 10.1002/smll.202003765] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Indexed: 06/12/2023]
Abstract
Despite the latest advances in cardiovascular biology and medicine, myocardial infarction (MI) remains one of the major causes of deaths worldwide. While reperfusion of the myocardium is critical to limit the ischemic damage typical of a MI event, it causes detrimental morphological and functional changes known as "reperfusion injury." This complex scenario is poorly represented in currently available models of ischemia/reperfusion injury, leading to a poor translation of findings from the bench to the bedside. However, more recent bioengineered in vitro models of the human heart represent more clinically relevant tools to prevent and treat MI in patients. These include 3D cultures of cardiac cells, the use of patient-derived stem cells, and 3D bioprinting technology. This review aims at highlighting the major features typical of a heart attack while comparing current in vitro, ex vivo, and in vivo models. This information has the potential to further guide in developing novel advanced in vitro cardiac models of ischemia/reperfusion injury. It may pave the way for the generation of advanced pathophysiological cardiac models with the potential to develop personalized therapies.
Collapse
Affiliation(s)
- Poonam Sharma
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Laura Vettori
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Gemma Figtree
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Andrew Boyle
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Carmine Gentile
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| |
Collapse
|
41
|
Roostalu U, Thisted L, Skytte JL, Salinas CG, Pedersen PJ, Hecksher-Sørensen J, Rolin B, Hansen HH, MacKrell JG, Christie RM, Vrang N, Jelsing J, Zois NE. Effect of captopril on post-infarction remodelling visualized by light sheet microscopy and echocardiography. Sci Rep 2021; 11:5241. [PMID: 33664407 PMCID: PMC7933438 DOI: 10.1038/s41598-021-84812-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
Angiotensin converting enzyme inhibitors, among them captopril, improve survival following myocardial infarction (MI). The mechanisms of captopril action remain inadequately understood due to its diverse effects on multiple signalling pathways at different time periods following MI. Here we aimed to establish the role of captopril in late-stage post-MI remodelling. Left anterior descending artery (LAD) ligation or sham surgery was carried out in male C57BL/6J mice. Seven days post-surgery LAD ligated mice were allocated to daily vehicle or captopril treatment continued over four weeks. To provide comprehensive characterization of the changes in mouse heart following MI a 3D light sheet imaging method was established together with automated image analysis workflow. The combination of echocardiography and light sheet imaging enabled to assess cardiac function and the underlying morphological changes. We show that delayed captopril treatment does not affect infarct size but prevents left ventricle dilation and hypertrophy, resulting in improved ejection fraction. Quantification of lectin perfused blood vessels showed improved vascular density in the infarct border zone in captopril treated mice in comparison to vehicle dosed control mice. These results validate the applicability of combined echocardiographic and light sheet assessment of drug mode of action in preclinical cardiovascular research.
Collapse
Affiliation(s)
- Urmas Roostalu
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark.
| | | | | | | | | | | | - Bidda Rolin
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
- Novo Nordisk, 2760, Maaloev, Denmark
| | | | - James G MacKrell
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Robert M Christie
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Niels Vrang
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | - Jacob Jelsing
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | | |
Collapse
|
42
|
Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair. Nat Commun 2021; 12:1412. [PMID: 33658506 PMCID: PMC7930285 DOI: 10.1038/s41467-021-21682-7] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac patches are an effective way to deliver therapeutics to the heart. However, such procedures are normally invasive and difficult to perform. Here, we develop and test a method to utilize the pericardial cavity as a natural "mold" for in situ cardiac patch formation after intrapericardial injection of therapeutics in biocompatible hydrogels. In rodent models of myocardial infarction, we demonstrate that intrapericardial injection is an effective and safe method to deliver hydrogels containing induced pluripotent stem cells-derived cardiac progenitor cells or mesenchymal stem cells-derived exosomes. After injection, the hydrogels form a cardiac patch-like structure in the pericardial cavity, mitigating immune response and increasing the cardiac retention of the therapeutics. With robust cardiovascular repair and stimulation of epicardium-derived cells, the delivered therapeutics mitigate cardiac remodeling and improve cardiac functions post myocardial infarction. Furthermore, we demonstrate the feasibility of minimally-invasive intrapericardial injection in a clinically-relevant porcine model. Collectively, our study establishes intrapericardial injection as a safe and effective method to deliver therapeutic-bearing hydrogels to the heart for cardiac repair.
Collapse
|
43
|
Deshmukh T, Selvakumar D, Thavapalachandran S, Archer O, Grieve S, Chong J. Improvement in Cardiac Strain Metrics With Platelet Derived Growth Factor-AB Administration in a Porcine Ischaemia Heart Failure Model. Heart Lung Circ 2021. [DOI: 10.1016/j.hlc.2021.06.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Large Animal Models of Cell-Free Cardiac Regeneration. Biomolecules 2020; 10:biom10101392. [PMID: 33003617 PMCID: PMC7600588 DOI: 10.3390/biom10101392] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022] Open
Abstract
The adult mammalian heart lacks the ability to sufficiently regenerate itself, leading to the progressive deterioration of function and heart failure after ischemic injuries such as myocardial infarction. Thus far, cell-based therapies have delivered unsatisfactory results, prompting the search for cell-free alternatives that can induce the heart to repair itself through cardiomyocyte proliferation, angiogenesis, and advantageous remodeling. Large animal models are an invaluable step toward translating basic research into clinical applications. In this review, we give an overview of the state-of-the-art in cell-free cardiac regeneration therapies that have been tested in large animal models, mainly pigs. Cell-free cardiac regeneration therapies involve stem cell secretome- and extracellular vesicles (including exosomes)-induced cardiac repair, RNA-based therapies, mainly regarding microRNAs, but also modified mRNA (modRNA) as well as other molecules including growth factors and extracellular matrix components. Various methods for the delivery of regenerative substances are used, including adenoviral vectors (AAVs), microencapsulation, and microparticles. Physical stimulation methods and direct cardiac reprogramming approaches are also discussed.
Collapse
|
45
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
46
|
White SJ, Chong JJH. Growth factor therapy for cardiac repair: an overview of recent advances and future directions. Biophys Rev 2020; 12:805-815. [PMID: 32691300 DOI: 10.1007/s12551-020-00734-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
Heart disease represents a significant public health burden and is associated with considerable morbidity and mortality at the level of the individual. Current therapies for pathologies such as myocardial infarction, cardiomyopathy and heart failure are unable to repair damaged tissue to an extent that provides restoration of function approaching that of the pre-diseased state. Novel approaches to repair and regenerate the injured heart include cell therapy and the use of exogenous factors. Improved understanding of the role of growth factors in endogenous cardiac repair processes has motivated the investigation of their potential as therapeutic agents for cardiac pathology. Despite the disappointing performance of other growth factors in historical clinical trials, insulin-like growth factor 1 (IGF-1), neuregulin and platelet-derived growth factor (PDGF) have recently emerged as new candidate therapies. These growth factors elicit tissue repair through anti-apoptotic, pro-angiogenic and fibrosis-modulating mechanisms and have produced clinically significant functional improvement in preclinical studies. Early human trials suggest that IGF-1 and neuregulin are well tolerated and yield dose-dependent benefit, warranting progression to later phase studies. However, outstanding challenges such as short growth factor serum half-life and insufficient target-organ specificity currently necessitate the development of novel delivery strategies.
Collapse
Affiliation(s)
- Samuel J White
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Department of Cardiology, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
47
|
Platelet-derived growth factor AB promotes wound repair after MI. Nat Rev Cardiol 2020; 17:133. [DOI: 10.1038/s41569-020-0338-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|