1
|
Ni W, Ge X, Liu Y, Chen J, Wang L, Chen L, Li Z, Zhang P, Huang S, Xu J, Zhang L, Fan X, Wang G, Huang W, Ye Y, Zhou J, Dai C, Liu B. CD163 + macrophages attenuate pressure overload-induced left ventricular systolic dysfunction and cardiac mitochondrial dysfunction via interleukin-10. Basic Res Cardiol 2025:10.1007/s00395-025-01114-z. [PMID: 40343453 DOI: 10.1007/s00395-025-01114-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/27/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
Macrophage depletion exacerbates pressure overload-induced heart failure, but therapeutic translation is hindered by macrophage subset heterogeneity. The functional role of CD163+ macrophages in heart failure remains unclear. Transverse aortic constriction (TAC) was employed to induce pressure overload. Cd163-/- mice exhibited significantly aggravated TAC-induced left ventricular systolic dysfunction, as demonstrated by reduced ejection fraction, fractional shortening, and global longitudinal strain, compared to wild-type (WT) controls. RNA sequencing of cardiac tissues revealed significant differential gene expression between TAC-treated WT and Cd163-/- mice, especially in pathways governing mitochondrial bioenergetics and homeostasis. Transmission electron microscopy confirmed greater accumulation of dysfunctional mitochondria in cardiomyocytes of Cd163-/- mice relative to WT following TAC. Additionally, the proportion of CD163+ macrophages among cardiac macrophages increased post-TAC. Serum IL-10 levels and cardiac macrophage IL-10 expression were significantly diminished in Cd163-/- mice compared to WT after TAC. IL-10 supplementation effectively reversed the TAC-induced impairment in left ventricular systolic function in both WT and Cd163-/- mice, and reduced NADH/NAD+ ratios, reduced mitochondrial dysfunction, and improved mitochondrial membrane potential in Cd163-/- mice. Cross-sectional clinical data supported these findings, showing decreased IL-10 levels as a significant risk factor for heart failure in hypertensive patients (odds ratio: 0.397; 95% CI 0.203-0.775; p = 0.007). Collectively, these results highlight the protective role of CD163+ macrophages against pressure overload-induced left ventricular dysfunction and mitochondrial dysfunction through IL-10-dependent pathways.
Collapse
Affiliation(s)
- Wei Ni
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Qingchun East Road 3, Hangzhou, 310016, China
| | - Xiaofeng Ge
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Yang Liu
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
- Liaoning University of Traditional Chinese Medicine, Chongshan East Road 79, Shenyang, 110032, China
| | - Jingyu Chen
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Lin Wang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Linjian Chen
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Zhaokai Li
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Peng Zhang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Shufen Huang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Junhui Xu
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Le Zhang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Xiabin Fan
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Gang Wang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Wei Huang
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Yuanchao Ye
- Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Jiancang Zhou
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Qingchun East Road 3, Hangzhou, 310016, China
| | - Cuilian Dai
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China.
| | - Binbin Liu
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China.
| |
Collapse
|
2
|
Ramadoss S, Qin J, Tao B, Thomas NE, Cao E, Wu R, Sandoval DR, Piermatteo A, Grunddal KV, Ma F, Li S, Sun B, Zhou Y, Wan J, Pellegrini M, Holst B, Lusis AJ, Gordts PLSM, Deb A. Bone-marrow macrophage-derived GPNMB protein binds to orphan receptor GPR39 and plays a critical role in cardiac repair. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1356-1373. [PMID: 39455836 DOI: 10.1038/s44161-024-00555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein initially identified in nonmetastatic melanomas and has been associated with human heart failure; however, its role in cardiac injury and function remains unclear. Here we show that GPNMB expression is elevated in failing human and mouse hearts after myocardial infarction (MI). Lineage tracing and bone-marrow transplantation reveal that bone-marrow-derived macrophages are the main source of GPNMB in injured hearts. Using genetic loss-of-function models, we demonstrate that GPNMB deficiency leads to increased mortality, cardiac rupture and rapid post-MI left ventricular dysfunction. Conversely, increasing circulating GPNMB levels through viral delivery improves heart function after MI. Single-cell transcriptomics show that GPNMB enhances myocyte contraction and reduces fibroblast activation. Additionally, we identified GPR39 as a receptor for circulating GPNMB, with its absence negating the beneficial effects. These findings highlight a pivotal role of macrophage-derived GPNMBs in post-MI cardiac repair through GPR39 signaling.
Collapse
MESH Headings
- Animals
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Humans
- Macrophages/metabolism
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/genetics
- Mice, Knockout
- Disease Models, Animal
- Myocytes, Cardiac/metabolism
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Ventricular Function, Left
- Heart Failure/metabolism
- Heart Failure/genetics
- Female
- Mice
- Cells, Cultured
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/genetics
- Bone Marrow Transplantation
- Protein Binding
- Regeneration
- Eye Proteins
Collapse
Affiliation(s)
- Sivakumar Ramadoss
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Juan Qin
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nathan E Thomas
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
- Glycobiology Research and Training Center, University of California-San Diego, La Jolla, CA, USA
| | - Edward Cao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Daniel R Sandoval
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Ann Piermatteo
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Kaare V Grunddal
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Baiming Sun
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Yonggang Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Jijun Wan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
- Glycobiology Research and Training Center, University of California-San Diego, La Jolla, CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA.
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Li X, Luo W, Tang Y, Wu J, Zhang J, Chen S, Zhou L, Tao Y, Tang Y, Wang F, Huang Y, Jose PA, Guo L, Zeng C. Semaglutide attenuates doxorubicin-induced cardiotoxicity by ameliorating BNIP3-Mediated mitochondrial dysfunction. Redox Biol 2024; 72:103129. [PMID: 38574433 PMCID: PMC11000183 DOI: 10.1016/j.redox.2024.103129] [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: 01/12/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
AIMS Doxorubicin is a powerful chemotherapeutic agent for cancer, whose use is limited due to its potential cardiotoxicity. Semaglutide (SEMA), a novel analog of glucagon-like peptide-1 (GLP-1), has received widespread attention for the treatment of diabetes. However, increasing evidence has highlighted its potential therapeutic benefits on cardiac function. Therefore, the objective of this study was to examine the efficacy of semaglutide in ameliorating doxorubicin-induced cardiotoxicity. METHODS AND RESULTS Doxorubicin-induced cardiotoxicity is an established model to study cardiac function. Cardiac function was studied by transthoracic echocardiography and invasive hemodynamic monitoring. The results showed that semaglutide significantly ameliorated doxorubicin-induced cardiac dysfunction. RNA sequencing suggested that Bnip3 is the candidate gene that impaired the protective effect of semaglutide in doxorubicin-induced cardiotoxicity. To determine the role of BNIP3 on the effect of semaglutide in doxorubicin-induced cardiotoxicity, BNIP3 with adeno-associated virus serotype 9 (AAV9) expressing cardiac troponin T (cTnT) promoter was injected into tail vein of C57/BL6J mice to overexpress BNIP3, specifically in the heart. Overexpression of BNIP3 prevented the improvement in cardiac function caused by semaglutide. In vitro experiments showed that semaglutide, via PI3K/AKT pathway, reduced BNIP3 expression in the mitochondria, improving mitochondrial function. CONCLUSION Semaglutide ameliorates doxorubicin-induced mitochondrial and cardiac dysfunction via PI3K/AKT pathway, by reducing BNIP3 expression in mitochondria. The improvement in mitochondrial function reduces doxorubicin-mediated cardiac injury and improves cardiac function. Therefore, semaglutide is a potential therapy to reduce doxorubicin-induced acute cardiotoxicity.
Collapse
Affiliation(s)
- Xiaoping Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Wenbin Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yang Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; College of Bioengineering, Chongqing University, Chongqing, China
| | - Jiangjiao Wu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Junkai Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Shengnan Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Lu Zhou
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yu Tao
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yuanjuan Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China; Department of Cardiology, The Third People's Hospital of Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Li Guo
- Endocrinology Department, The First Affiliated Hospital of the Third Military Medical University (Army Medical University), Chongqing, China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, China; Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, China; Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
4
|
Fu W, Liao Q, Shi Y, Liu W, Ren H, Xu C, Zeng C. Transient induction of actin cytoskeletal remodeling associated with dedifferentiation, proliferation, and redifferentiation stimulates cardiac regeneration. Acta Pharm Sin B 2024; 14:2537-2553. [PMID: 38828141 PMCID: PMC11143747 DOI: 10.1016/j.apsb.2024.01.021] [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: 11/02/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 06/05/2024] Open
Abstract
The formation of new and functional cardiomyocytes requires a 3-step process: dedifferentiation, proliferation, and redifferentiation, but the critical genes required for efficient dedifferentiation, proliferation, and redifferentiation remain unknown. In our study, a circular trajectory using single-nucleus RNA sequencing of the pericentriolar material 1 positive (PCM1+) cardiomyocyte nuclei from hearts 1 and 3 days after surgery-induced myocardial infarction (MI) on postnatal Day 1 was reconstructed and demonstrated that actin remodeling contributed to the dedifferentiation, proliferation, and redifferentiation of cardiomyocytes after injury. We identified four top actin-remodeling regulators, namely Tmsb4x, Tmsb10, Dmd, and Ctnna3, which we collectively referred to as 2D2P. Transiently expressed changes of 2D2P, using a polycistronic non-integrating lentivirus driven by Tnnt2 (cardiac-specific troponin T) promoters (Tnnt2-2D2P-NIL), efficiently induced transiently proliferative activation and actin remodeling in postnatal Day 7 cardiomyocytes and adult hearts. Furthermore, the intramyocardial delivery of Tnnt2-2D2P-NIL resulted in a sustained improvement in cardiac function without ventricular dilatation, thickened septum, or fatal arrhythmia for at least 4 months. In conclusion, this study highlights the importance of actin remodeling in cardiac regeneration and provides a foundation for new gene-cocktail-therapy approaches to improve cardiac repair and treat heart failure using a novel transient and cardiomyocyte-specific viral construct.
Collapse
Affiliation(s)
- Wenbin Fu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Qiao Liao
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Yu Shi
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Wujian Liu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Chunmei Xu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, the Third Military Medical University, Chongqing 400042, China
- Cardiovascular Research Center, Chongqing College, University of Chinese Academy of Sciences, Chongqing 400042, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400042, China
| |
Collapse
|
5
|
Wang Y, Li Q, Tao B, Angelini M, Ramadoss S, Sun B, Wang P, Krokhaleva Y, Ma F, Gu Y, Espinoza A, Yamauchi K, Pellegrini M, Novitch B, Olcese R, Qu Z, Song Z, Deb A. Fibroblasts in heart scar tissue directly regulate cardiac excitability and arrhythmogenesis. Science 2023; 381:1480-1487. [PMID: 37769108 PMCID: PMC10768850 DOI: 10.1126/science.adh9925] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/11/2023] [Indexed: 09/30/2023]
Abstract
After heart injury, dead heart muscle is replaced by scar tissue. Fibroblasts can electrically couple with myocytes, and changes in fibroblast membrane potential can lead to myocyte excitability, which suggests that fibroblast-myocyte coupling in scar tissue may be responsible for arrhythmogenesis. However, the physiologic relevance of electrical coupling of myocytes and fibroblasts and its impact on cardiac excitability in vivo have never been demonstrated. We genetically engineered a mouse that expresses the optogenetic cationic channel ChR2 (H134R) exclusively in cardiac fibroblasts. After myocardial infarction, optical stimulation of scar tissue elicited organ-wide cardiac excitation and induced arrhythmias in these animals. Complementing computational modeling with experimental approaches, we showed that gap junctional and ephaptic coupling, in a synergistic yet functionally redundant manner, excited myocytes coupled to fibroblasts.
Collapse
Affiliation(s)
- Yijie Wang
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Qihao Li
- Peng Cheng Laboratory, Shenzhen, Guangdong 518000, China
| | - Bo Tao
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Marina Angelini
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sivakumar Ramadoss
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Baiming Sun
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ping Wang
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yuliya Krokhaleva
- UCLA Cardiac Arrhythmia Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yiqian Gu
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences–The Collaboratory, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences–The Collaboratory, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ken Yamauchi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences–The Collaboratory, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett Novitch
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Zhilin Qu
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Zhen Song
- Peng Cheng Laboratory, Shenzhen, Guangdong 518000, China
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
Ma E, Wu C, Chen J, Wo D, Ren DN, Yan H, Peng L, Zhu W. Resveratrol prevents Ang II-induced cardiac hypertrophy by inhibition of NF-κB signaling. Biomed Pharmacother 2023; 165:115275. [PMID: 37541173 DOI: 10.1016/j.biopha.2023.115275] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Pathological cardiac hypertrophy is a hallmark of various cardiovascular diseases (CVD) including chronic heart failure (HF) and an important target for the treatment of these diseases. Aberrant activation of Angiotensin II (Ang II)/AT1R signaling pathway is one of the main triggers of cardiac hypertrophy, which further gives rise to excessive inflammation that is mediated by the key transcription factor NF-κB. Resveratrol (REV) is a natural polyphenol with multiple anti-inflammatory and anti-oxidative effects, however the ability of REV in preventing Ang II-induced cardiac hypertrophy in combination with NF-κB signaling activation remains unclear. METHODS Murine models of cardiac hypertrophy was conducted via implantation of Ang II osmotic pumps. Primary neonatal rat cardiomyocyte and heart tissues were examined to determine the effect and underlying mechanism of REV in preventing Ang II-induced cardiac hypertrophy. RESULTS Administrations of REV significantly prevented Ang II-induced cardiac hypertrophy, as well as robustly attenuated Ang II-induced cardiac fibrosis, and cardiac dysfunction. Furthermore, REV not only directly prevented Ang II/AT1R signal transductions, but also prevented Ang II-induced expressions of pro-inflammatory cytokines and activation of NF-κB signaling pathway. CONCLUSIONS Our study provides important new mechanistic insight into the cardioprotective effects of REV in preventing Ang II-induced cardiac hypertrophy via inhibiting adverse NF-κB signaling activation. Our findings further suggest the therapeutic potential of REV as a promising drug for the treatment of cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- En Ma
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Celiang Wu
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jinxiao Chen
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Da Wo
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China; Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Dan-Ni Ren
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Hongwei Yan
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Luying Peng
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China.
| | - Weidong Zhu
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China; Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| |
Collapse
|
7
|
Fang G, Li X, Yang F, Huang T, Qiu C, Peng K, Wang Z, Yang Y, Lan C. Amentoflavone mitigates doxorubicin-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation through inhibition of the STING/NLRP3 signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154922. [PMID: 37321078 DOI: 10.1016/j.phymed.2023.154922] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Doxorubicin (DOX) is a potent anticancer chemotherapeutic agent whose clinical application is substantially constrained by its cardiotoxicity. The pathophysiology of DOX-induced cardiotoxicity manifests as cardiomyocyte pyroptosis and inflammation. Amentoflavone (AMF) is a naturally occurring biflavone possessing anti-pyroptotic and anti-inflammatory properties. However, the mechanism through which AMF alleviates DOX-induced cardiotoxicity remains undetermined. PURPOSE This study aimed at investigating the role of AMF in alleviating DOX-induced cardiotoxicity. STUDY DESIGN AND METHODS To assess the in vivo effect of AMF, DOX was intraperitoneally administered into a mouse model to induce cardiotoxicity. To elucidate the underlying mechanisms, the activities of STING/NLRP3 were quantified using the NLRP3 agonist nigericin and the STING agonist amidobenzimidazole (ABZI). Primary cardiomyocytes isolated from neonatal Sprague-Dawley rats were treated with saline (vehicle) or DOX with or without AMF and/or ABZI. The echocardiogram, haemodynamics, cardiac injury markers, heart/body weight ratio, and pathological alterations were monitored; the STING/NLRP3 pathway-associated proteins were detected by western blot and cardiomyocyte pyroptosis was analysed by immunofluorescence staining of cleaved N-terminal GSDMD and scanning electron microscopy. Furthermore, we evaluated the potential of AMF in compromising the anticancer effects of DOX in human breast cancer cell lines. RESULTS AMF substantially alleviated cardiac dysfunction and reduced heart/body weight ratio and myocardial damage in mice models of DOX-induced cardiotoxicity. AMF effectively suppressed DOX-mediated upregulation of IL-1β, IL-18, TNF-α, and pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and cleaved N-terminal GSDMD. The levels of apoptosis-related proteins, namely Bax, cleaved caspase-3, and BCL-2 were not affected. In addition, AMF inhibited STING phosphorylation in DOX-affected hearts. Intriguingly, the administration of nigericin or ABZI dampened the cardioprotective effects of AMF. The in vitro anti-pyroptotic effect of AMF was demonstrated in attenuating the DOX-induced reduction in cardiomyocyte cell viability, upregulation of cleaved N-terminal GSDMD, and pyroptotic morphology alteration at the microstructural level. AMF exhibited a synergistic effect with DOX to reduce the viability of human breast cancer cells. CONCLUSION AMF alleviates DOX-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation via inhibition of the STING/NLRP3 signalling pathway, thereby validating its efficacy as a cardioprotective agent.
Collapse
Affiliation(s)
- Guangyao Fang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Xiuchuan Li
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ting Huang
- Department of Medical Oncology, People's Hospital of Luotian County, Huanggang, Hubei, P.R. China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ke Peng
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ziran Wang
- Department of Orthopedics, 903rd Hospital of PLA, Hangzhou, Zhejiang, P.R. China
| | - Yongjian Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China..
| | - Cong Lan
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China..
| |
Collapse
|
8
|
Paz-Artigas L, Montero-Calle P, Iglesias-García O, Mazo MM, Ochoa I, Ciriza J. Current approaches for the recreation of cardiac ischaemic environment in vitro. Int J Pharm 2023; 632:122589. [PMID: 36623742 DOI: 10.1016/j.ijpharm.2023.122589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Myocardial ischaemia is one of the leading dead causes worldwide. Although animal experiments have historically provided a wealth of information, animal models are time and money consuming, and they usually miss typical human patient's characteristics associated with ischemia prevalence, including aging and comorbidities. Generating reliable in vitro models that recapitulate the human cardiac microenvironment during an ischaemic event can boost the development of new drugs and therapeutic strategies, as well as our understanding of the underlying cellular and molecular events, helping the optimization of therapeutic approaches prior to animal and clinical testing. Although several culture systems have emerged for the recreation of cardiac physiology, mimicking the features of an ischaemic heart tissue in vitro is challenging and certain aspects of the disease process remain poorly addressed. Here, current in vitro cardiac culture systems used for modelling cardiac ischaemia, from self-aggregated organoids to scaffold-based constructs and heart-on-chip platforms are described. The advantages of these models to recreate ischaemic hallmarks such as oxygen gradients, pathological alterations of mechanical strength or fibrotic responses are highlighted. The new models represent a step forward to be considered, but unfortunately, we are far away from recapitulating all complexity of the clinical situations.
Collapse
Affiliation(s)
- Laura Paz-Artigas
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Pilar Montero-Calle
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Manuel M Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Hematology and Cell Therapy, Clínica Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| | - Jesús Ciriza
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| |
Collapse
|