1
|
Ma X, Hou J, Xiong JW. A cellular triad for linking cardiac niche to regeneration. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:29. [PMID: 39653982 PMCID: PMC11628456 DOI: 10.1186/s13619-024-00213-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Cardiovascular disease is the leading cause of mortality with very limited therapeutic interventions, thus holding great hope for cardiac regenerative medicine. A recent work from Martin's laboratory reports their identification of a fetal-like cardiomyocyte progenitor, adult cardiomyocyte type 2 (aCM2), and its potential interactions with C3+ cardiac fibroblasts and C3ar1+ macrophages to form a regenerative cellular triad, which is only present in the regenerative heart models, YAP5SA-expressing adult hearts and neonatal hearts. The complement signaling is essential for cellular interactions in this regenerative triad. This Highlight summarizes these major findings and provides brief perspectives on the impact of this regenerative niche during cardiac regeneration in the future.
Collapse
Affiliation(s)
- Xiaokai Ma
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, and College of Future Technology, Peking University, Beijing, 100871, China
| | - Junjie Hou
- School of Basic Medical Sciences, Institute of Biomedical Innovation, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, and College of Future Technology, Peking University, Beijing, 100871, China.
- School of Basic Medical Sciences, Institute of Biomedical Innovation, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
2
|
Sada T, Kimura W. Transition from fetal to postnatal state in the heart: Crosstalk between metabolism and regeneration. Dev Growth Differ 2024; 66:438-451. [PMID: 39463005 DOI: 10.1111/dgd.12947] [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: 06/16/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. Myocardial injury resulting from ischemia can be fatal because of the limited regenerative capacity of adult myocardium. Mammalian cardiomyocytes rapidly lose their proliferative capacities, with only a small fraction of adult myocardium remaining proliferative, which is insufficient to support post-injury recovery. Recent investigations have revealed that this decline in myocardial proliferative capacity is closely linked to perinatal metabolic shifts. Predominantly glycolytic fetal myocardial metabolism transitions towards mitochondrial fatty acid oxidation postnatally, which not only enables efficient production of ATP but also causes a dramatic reduction in cardiomyocyte proliferative capacity. Extensive research has elucidated the mechanisms behind this metabolic shift, as well as methods to modulate these metabolic pathways. Some of these methods have been successfully applied to enhance metabolic reprogramming and myocardial regeneration. This review discusses recently acquired insights into the interplay between metabolism and myocardial proliferation, emphasizing postnatal metabolic transitions.
Collapse
Affiliation(s)
- Tai Sada
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Wataru Kimura
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
3
|
Yue T, Zhang W, Pei H, Danzeng D, He J, Yang J, Luo Y, Zhang Z, Xiong S, Yang X, Ji Q, Yang Z, Hou J. Monascus pigment-protected bone marrow-derived stem cells for heart failure treatment. Bioact Mater 2024; 42:270-283. [PMID: 39285916 PMCID: PMC11403898 DOI: 10.1016/j.bioactmat.2024.08.038] [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: 07/27/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in heart failure (HF) treatment. However, their clinical application is impeded by low retention rate and low cellular activity of MSCs caused by high inflammatory and reactive oxygen species (ROS) microenvironment. In this study, monascus pigment (MP) nanoparticle (PPM) was proposed for improving adverse microenvironment and assisting in transplantation of bone marrow-derived MSCs (BMSCs). Meanwhile, in order to load PPM and reduce the mechanical damage of BMSCs, injectable hydrogels based on Schiff base cross-linking were prepared. The PPM displays ROS-scavenging and macrophage phenotype-regulating capabilities, significantly enhancing BMSCs survival and activity in HF microenvironment. This hydrogel demonstrates superior biocompatibility, injectability, and tissue adhesion. With the synergistic effects of injectable, adhesive hydrogel and the microenvironment-modulating properties of MP, cardiac function was effectively improved in the pericardial sac of rats. Our results offer insights into advancing BMSCs-based HF therapies and their clinical applications.
Collapse
Affiliation(s)
- Tian Yue
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Wentai Zhang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Haifeng Pei
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, China
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, 850000, China
| | - Jian He
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Jiali Yang
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Yong Luo
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Shiqiang Xiong
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Xiangbo Yang
- Ya'an Xunkang Pharmaceutical Co., LTD, Ya'an, Sichuan, 625015, China
| | - Qisen Ji
- Ya'an Xunkang Pharmaceutical Co., LTD, Ya'an, Sichuan, 625015, China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Jun Hou
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| |
Collapse
|
4
|
Sharma P, Bal T, Singh SK, Sharma N. Biodegradable polymeric nanocomposite containing phloretin for enhanced oral bioavailability and improved myocardial ischaemic recovery. J Microencapsul 2024; 41:754-769. [PMID: 39431662 DOI: 10.1080/02652048.2024.2418608] [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: 06/18/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
AIM The study aimed to enhance phloretin's oral absorption and systemic availability through nanoencapsulation within biodegradable polymers, improving its anti-oxidant and cardioprotective potential. METHODS Phloretin-loaded polymeric nanocomposites were prepared using ionic gelation and optimised for yield, encapsulation, loading, particle size, PdI and zeta potential. The formulation was characterised by FTIR, XRD, FESEM and MS. In-vitro drug release, stability, pharmacokinetics, biodistribution, anti-oxidant capacity, haemolysis and both in-vitro and in-vivo assessments were conducted in an ischaemia-induced rat model. RESULTS The average particle size, zeta potential, encapsulation and drug loading of the optimised nanoparticles were 105.8 ± 1.92 nm, -41.5 ± 1.10 mV, 92.36 ± 0.01% and 18.47 ± 0.38%, respectively. Nano-phloretin enhanced oral bioavailability, anti-oxidant capacity. In-vivo, it reduced myocardial infarct size by ∼46% versus ∼13% for free phloretin, showing significant cardiomyocyte protection and ROS suppression. CONCLUSION The study demonstrates polymer-based nanoparticles as effective oral drug delivery systems capable of enhancing both systemic bioavailability and therapeutic efficacy of the encapsulated drug.
Collapse
Affiliation(s)
- Prasanti Sharma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Trishna Bal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Sandeep Kumar Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Neelima Sharma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| |
Collapse
|
5
|
Zuo X, Xiao Y, Yang J, He Y, He Y, Liu K, Chen X, Guo J. Engineering collagen-based biomaterials for cardiovascular medicine. COLLAGEN AND LEATHER 2024; 6:33. [DOI: 10.1186/s42825-024-00174-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 01/06/2025]
Abstract
AbstractCardiovascular diseases have been the leading cause of global mortality and disability. In addition to traditional drug and surgical treatment, more and more studies investigate tissue engineering therapeutic strategies in cardiovascular medicine. Collagen interweaves in the form of trimeric chains to form the physiological network framework of the extracellular matrix of cardiac and vascular cells, possessing excellent biological properties (such as low immunogenicity and good biocompatibility) and adjustable mechanical properties, which renders it a vital tissue engineering biomaterial for the treatment of cardiovascular diseases. In recent years, promising advances have been made in the application of collagen materials in blood vessel prostheses, injectable cardiac hydrogels, cardiac patches, and hemostatic materials, although their clinical translation still faces some obstacles. Thus, we reviewed these findings and systematically summarizes the application progress as well as problems of clinical translation of collagen biomaterials in the cardiovascular field. The present review contributes to a comprehensive understanding of the application of collagen biomaterials in cardiovascular medicine.
Graphical abstract
Collapse
|
6
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [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: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Chen ZY, Ji SJ, Huang CW, Tu WZ, Ren XY, Guo R, Xie X. In situ reprogramming of cardiac fibroblasts into cardiomyocytes in mouse heart with chemicals. Acta Pharmacol Sin 2024; 45:2290-2299. [PMID: 38890526 PMCID: PMC11489685 DOI: 10.1038/s41401-024-01308-6] [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: 02/05/2024] [Accepted: 05/07/2024] [Indexed: 06/20/2024] Open
Abstract
Cardiomyocytes are terminal differentiated cells and have limited ability to proliferate or regenerate. Condition like myocardial infarction causes massive death of cardiomyocytes and is the leading cause of death. Previous studies have demonstrated that cardiac fibroblasts can be induced to transdifferentiate into cardiomyocytes in vitro and in vivo by forced expression of cardiac transcription factors and microRNAs. Our previous study have demonstrated that full chemical cocktails could also induce fibroblast to cardiomyocyte transdifferentiation both in vitro and in vivo. With the development of tissue clearing techniques, it is possible to visualize the reprogramming at the whole-organ level. In this study, we investigated the effect of the chemical cocktail CRFVPTM in inducing in situ fibroblast to cardiomyocyte transdifferentiation with two strains of genetic tracing mice, and the reprogramming was observed at whole-heart level with CUBIC tissue clearing technique and 3D imaging. In addition, single-cell RNA sequencing (scRNA-seq) confirmed the generation of cardiomyocytes from cardiac fibroblasts which carries the tracing marker. Our study confirms the use of small molecule cocktails in inducing in situ fibroblast to cardiomyocyte reprogramming at the whole-heart level and proof-of-conceptly providing a new source of naturally incorporated cardiomyocytes to help heart regeneration.
Collapse
Affiliation(s)
- Zi-Yang Chen
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Si-Jia Ji
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Chen-Wen Huang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wan-Zhi Tu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Xin-Yue Ren
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ren Guo
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264119, China
| | - Xin Xie
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264119, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
8
|
Fiorino E, Rossin D, Vanni R, Aubry M, Giachino C, Rastaldo R. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction. Int J Mol Sci 2024; 25:11747. [PMID: 39519298 PMCID: PMC11546116 DOI: 10.3390/ijms252111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial infarction (MI) is a critical global health issue and a leading cause of heart failure. Indeed, while neonatal mammals can regenerate cardiac tissue mainly through cardiomyocyte proliferation, this ability is lost shortly after birth, resulting in the adult heart's inability to regenerate after injury effectively. In adult mammals, the adverse cardiac remodelling, which compensates for the loss of cardiac cells, impairs cardiac function due to the non-contractile nature of fibrotic tissue. Moreover, the neovascularisation after MI is inadequate to restore blood flow to the infarcted myocardium. This review aims to synthesise the most recent insights into the molecular and cellular players involved in endogenous myocardial and vascular regeneration, facilitating the identification of mechanisms that could be targeted to trigger cardiac regeneration, reduce fibrosis, and improve functional recovery post-MI. Reprogramming adult cardiomyocytes to regain their proliferative potential, along with the modulation of target cells responsible for neovascularisation, represents promising therapeutic strategies. An updated overview of endogenous mechanisms that regulate both myocardial and coronary vasculature regeneration-including stem and progenitor cells, growth factors, cell cycle regulators, and key signalling pathways-could help identify new critical intervention points for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (E.F.); (D.R.); (R.V.); (M.A.); (C.G.)
| |
Collapse
|
9
|
Yan W, Li Y, Wang G, Huang Y, Xie P. Clinical application and immune infiltration landscape of stemness-related genes in heart failure. ESC Heart Fail 2024. [PMID: 39275894 DOI: 10.1002/ehf2.15055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Stemness refers to the self-renewal and differentiation ability of cells. However, little is known about the heart's stemness properties. Thus, the current study aims to identify putative stemness-related biomarkers to construct a viable prediction model of HF and characterize the immune infiltration features of HF. METHODS HF datasets from the Gene Expression Omnibus (GEO) database were adopted as the training and validation cohorts while stemness-related genes were obtained from GeneCards and previously published papers. Feature selection was performed using two machine learning algorithms. Nomogram models were then constructed to predict HF risk based on the selected key genes. Moreover, the biological functions of the key genes were evaluated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes Genomes (KEGG) pathway analyses, and gene set variation analysis (GSVA) and enrichment analysis (GSEA) were performed between the high- and low-risk groups. The immune infiltration landscape in HF was investigated, and the interaction network of key genes was analysed to predict potential targets and molecular mechanisms. RESULTS Seven key genes, namely SMOC2, LUM, FNDC1, SCUBE2, CD163, BLM and S1PR3, were included in the proposed nomogram. This nomogram showed good predictive performance for HF diagnosis in the training and validation sets. GO and KEGG analyses revealed that the key genes were primarily associated with ageing, inflammatory processes and DNA oxidation. GSEA and GSVA identified various inflammatory and immune signalling pathways that were enriched between the high- and low-risk groups. The infiltration of 15 immune cell subsets suggests that adaptive immunity has an important role in HF. CONCLUSIONS Our study identified a clinically significant stemness-related signature for predicting HF risk, with the potential to improve early disease diagnosis, optimize risk stratification and provide new strategies for treating patients with HF.
Collapse
Affiliation(s)
- Wenting Yan
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yanling Li
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Gang Wang
- First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yuan Huang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Ping Xie
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
10
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Albericio G, Higuera M, Araque P, Sánchez C, Herrero D, García-Brenes MA, Formentini L, Torán JL, Mora C, Bernad A. Development of a Bmi1+ Cardiac Mouse Progenitor Immortalized Model to Unravel the Relationship with Its Protective Vascular Endothelial Niche. Int J Mol Sci 2024; 25:8815. [PMID: 39201501 PMCID: PMC11354400 DOI: 10.3390/ijms25168815] [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: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
The adult mammalian heart has been demonstrated to be endowed with low but real turnover capacity, especially for cardiomyocytes, the key functional cell type. The source, however, of that turnover capacity remains controversial. In this regard, we have defined and characterized a resident multipotent cardiac mouse progenitor population, Bmi1+DR (for Bmi1+ Damage-Responsive cells). Bmi1+DR is one of the cell types with the lowest ROS (Reactive Oxygen Species) levels in the adult heart, being particularly characterized by their close relationship with cardiac vessels, most probably involved in the regulation of proliferation/maintenance of Bmi1+DR. This was proposed to work as their endothelial niche. Due to the scarcity of Bmi1+DR cells in the adult mouse heart, we have generated an immortalization/dis-immortalization model using Simian Vacuolating Virus 40-Large Antigen T (SV40-T) to facilitate their in vitro characterization. We have obtained a heterogeneous population of immortalized Bmi1+DR cells (Bmi1+DRIMM) that was validated attending to different criteria, also showing a comparable sensitivity to strong oxidative damage. Then, we concluded that the Bmi1-DRIMM population is an appropriate model for primary Bmi1+DR in vitro studies. The co-culture of Bmi1+DRIMM cells with endothelial cells protects them against oxidative damage, showing a moderate depletion in non-canonical autophagy and also contributing with a modest metabolic regulation.
Collapse
Affiliation(s)
- Guillermo Albericio
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
- Molecular Biology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Marina Higuera
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Paula Araque
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Cristina Sánchez
- Molecular Biology Department, Molecular Biology Center Severo Ochoa (CBMSO), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Diego Herrero
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Miguel A. García-Brenes
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Laura Formentini
- Molecular Biology Department, Molecular Biology Center Severo Ochoa (CBMSO), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - José Luis Torán
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Carmen Mora
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Antonio Bernad
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| |
Collapse
|
12
|
Jin A, Shao Y, Wang F, Feng J, Lei L, Dai M. Designing polysaccharide materials for tissue repair and regeneration. APL MATERIALS 2024; 12. [DOI: 10.1063/5.0223937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Tissue repair and regeneration are critical processes for maintaining the integrity and function of various organs and tissues. Recently, polysaccharide materials and protein materials have garnered interest for use in tissue repair strategies. However, polysaccharides are more stable and unaffected by temperature and pH changes compared to proteins, and some polysaccharides can provide stronger mechanical support, which is particularly important for constructing tissue-engineered scaffolds and wound dressings. This Review provides an in-depth overview of the origins of polysaccharides, the advantages of polysaccharide materials, and processing and design strategies. In addition, the potential of polysaccharide materials for the restoration of tissues such as skin, heart, and nerves is highlighted. Finally, we discuss in depth the challenges that polysaccharide materials still face in tissue repair, such as the stability of the material, regulating mechanical characteristics and deterioration rates under different conditions. To achieve more effective tissue repair and regeneration, future research must focus on further improving the characteristics and functionalities of polysaccharide materials.
Collapse
Affiliation(s)
- Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 1 , Hangzhou 310015, China
| | - Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 1 , Hangzhou 310015, China
| | - Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 1 , Hangzhou 310015, China
| | - Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 1 , Hangzhou 310015, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University 1 , Hangzhou 310015, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University 2 , Wenzhou 325200, China
| |
Collapse
|
13
|
Foglio E, D'Avorio E, Nieri R, Russo MA, Limana F. Epicardial EMT and cardiac repair: an update. Stem Cell Res Ther 2024; 15:219. [PMID: 39026298 PMCID: PMC11264588 DOI: 10.1186/s13287-024-03823-z] [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: 05/16/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024] Open
Abstract
Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, Latina, Italy
| | - Erica D'Avorio
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy
| | - Riccardo Nieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Limana
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy.
- Laboratorio di Patologia Cellulare e Molecolare, IRCCS San Raffaele Roma, Rome, Italy.
| |
Collapse
|
14
|
Zheng K, Hao Y, Xia C, Cheng S, Yu J, Chen Z, Li Y, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Zhao J, Li R, Zong J, Zhang H, Lai L, Huang P, Zhou C, Xia J, Zhang X, Wu J. Effects and mechanisms of the myocardial microenvironment on cardiomyocyte proliferation and regeneration. Front Cell Dev Biol 2024; 12:1429020. [PMID: 39050889 PMCID: PMC11266095 DOI: 10.3389/fcell.2024.1429020] [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: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The adult mammalian cardiomyocyte has a limited capacity for self-renewal, which leads to the irreversible heart dysfunction and poses a significant threat to myocardial infarction patients. In the past decades, research efforts have been predominantly concentrated on the cardiomyocyte proliferation and heart regeneration. However, the heart is a complex organ that comprises not only cardiomyocytes but also numerous noncardiomyocyte cells, all playing integral roles in maintaining cardiac function. In addition, cardiomyocytes are exposed to a dynamically changing physical environment that includes oxygen saturation and mechanical forces. Recently, a growing number of studies on myocardial microenvironment in cardiomyocyte proliferation and heart regeneration is ongoing. In this review, we provide an overview of recent advances in myocardial microenvironment, which plays an important role in cardiomyocyte proliferation and heart regeneration.
Collapse
Affiliation(s)
- Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoxian Cheng
- Jingshan Union Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
von Bibra C, Hinkel R. Non-human primate studies for cardiomyocyte transplantation-ready for translation? Front Pharmacol 2024; 15:1408679. [PMID: 38962314 PMCID: PMC11221829 DOI: 10.3389/fphar.2024.1408679] [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: 03/28/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Non-human primates (NHP) are valuable models for late translational pre-clinical studies, often seen as a last step before clinical application. The unique similarity between NHPs and humans is often the subject of ethical concerns. However, it is precisely this analogy in anatomy, physiology, and the immune system that narrows the translational gap to other animal models in the cardiovascular field. Cell and gene therapy approaches are two dominant strategies investigated in the research field of cardiac regeneration. Focusing on the cell therapy approach, several xeno- and allogeneic cell transplantation studies with a translational motivation have been realized in macaque species. This is based on the pressing need for novel therapeutic options for heart failure patients. Stem cell-based remuscularization of the injured heart can be achieved via direct injection of cardiomyocytes (CMs) or patch application. Both CM delivery approaches are in the late preclinical stage, and the first clinical trials have started. However, are we already ready for the clinical area? The present review concentrates on CM transplantation studies conducted in NHPs, discusses the main sources and discoveries, and provides a perspective about human translation.
Collapse
Affiliation(s)
- Constantin von Bibra
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| | - Rabea Hinkel
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| |
Collapse
|
16
|
Ivanova A, Kohl F, González-King Garibotti H, Chalupska R, Cvjetkovic A, Firth M, Jennbacken K, Martinsson S, Silva AM, Viken I, Wang QD, Wiseman J, Dekker N. In vivo phage display identifies novel peptides for cardiac targeting. Sci Rep 2024; 14:12177. [PMID: 38806609 PMCID: PMC11133476 DOI: 10.1038/s41598-024-62953-9] [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: 01/24/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
Heart failure remains a leading cause of mortality. Therapeutic intervention for heart failure would benefit from targeted delivery to the damaged heart tissue. Here, we applied in vivo peptide phage display coupled with high-throughput Next-Generation Sequencing (NGS) and identified peptides specifically targeting damaged cardiac tissue. We established a bioinformatics pipeline for the identification of cardiac targeting peptides. Hit peptides demonstrated preferential uptake by human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and immortalized mouse HL1 cardiomyocytes, without substantial uptake in human liver HepG2 cells. These novel peptides hold promise for use in targeted drug delivery and regenerative strategies and open new avenues in cardiovascular research and clinical practice.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden.
| | - Franziska Kohl
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 1, Solna, 171 77, Stockholm, Sweden
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Renata Chalupska
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Aleksander Cvjetkovic
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Sofia Martinsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Ida Viken
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - John Wiseman
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden.
| |
Collapse
|
17
|
Bongiovanni C, Bueno-Levy H, Posadas Pena D, Del Bono I, Miano C, Boriati S, Da Pra S, Sacchi F, Redaelli S, Bergen M, Romaniello D, Pontis F, Tassinari R, Kellerer L, Petraroia I, Mazzeschi M, Lauriola M, Ventura C, Heermann S, Weidinger G, Tzahor E, D'Uva G. BMP7 promotes cardiomyocyte regeneration in zebrafish and adult mice. Cell Rep 2024; 43:114162. [PMID: 38678558 DOI: 10.1016/j.celrep.2024.114162] [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: 11/08/2023] [Revised: 03/06/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.
Collapse
Affiliation(s)
- Chiara Bongiovanni
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy; National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), via di Corticella 183, 40128 Bologna, Italy
| | - Hanna Bueno-Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Herzl St. 234, Rehovot 76100, Israel
| | - Denise Posadas Pena
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Irene Del Bono
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Carmen Miano
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy; National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), via di Corticella 183, 40128 Bologna, Italy
| | - Stefano Boriati
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Silvia Da Pra
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Francesca Sacchi
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy; National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), via di Corticella 183, 40128 Bologna, Italy
| | - Simone Redaelli
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Max Bergen
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, 79104 Freiburg, Germany
| | - Donatella Romaniello
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Francesca Pontis
- Scientific and Technological Pole, IRCCS MultiMedica, via Fantoli 16/15, 20138 Milan, Italy
| | | | - Laura Kellerer
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Ilaria Petraroia
- Scientific and Technological Pole, IRCCS MultiMedica, via Fantoli 16/15, 20138 Milan, Italy
| | - Martina Mazzeschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Mattia Lauriola
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; Centre for Applied Biomedical Research (CRBA), University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Carlo Ventura
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), via di Corticella 183, 40128 Bologna, Italy
| | - Stephan Heermann
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, 79104 Freiburg, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Herzl St. 234, Rehovot 76100, Israel
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
18
|
Apolínová K, Pérez FA, Dyballa S, Coppe B, Mercader Huber N, Terriente J, Di Donato V. ZebraReg-a novel platform for discovering regulators of cardiac regeneration using zebrafish. Front Cell Dev Biol 2024; 12:1384423. [PMID: 38799508 PMCID: PMC11116629 DOI: 10.3389/fcell.2024.1384423] [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: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide with myocardial infarction being the most prevalent. Currently, no cure is available to either prevent or revert the massive death of cardiomyocytes that occurs after a myocardial infarction. Adult mammalian hearts display a limited regeneration capacity, but it is insufficient to allow complete myocardial recovery. In contrast, the injured zebrafish heart muscle regenerates efficiently through robust proliferation of pre-existing myocardial cells. Thus, zebrafish allows its exploitation for studying the genetic programs behind cardiac regeneration, which may be present, albeit dormant, in the adult human heart. To this end, we have established ZebraReg, a novel and versatile automated platform for studying heart regeneration kinetics after the specific ablation of cardiomyocytes in zebrafish larvae. In combination with automated heart imaging, the platform can be integrated with genetic or pharmacological approaches and used for medium-throughput screening of presumed modulators of heart regeneration. We demonstrate the versatility of the platform by identifying both anti- and pro-regenerative effects of genes and drugs. In conclusion, we present a tool which may be utilised to streamline the process of target validation of novel gene regulators of regeneration, and the discovery of new drug therapies to regenerate the heart after myocardial infarction.
Collapse
Affiliation(s)
- Kateřina Apolínová
- ZeClinics SL, Barcelona, Spain
- Biomedicine, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | | | | | - Benedetta Coppe
- Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
| | - Nadia Mercader Huber
- Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
- Centro Nacional de Investigaciones Cardiovasculares CNIC, Madrid, Spain
| | | | | |
Collapse
|
19
|
Abouleisa RRE, Tang XL, Ou Q, Salama ABM, Woolard A, Hammouri D, Abdelhafez H, Cayton S, Abdulwali SK, Arai M, Sithu ID, Conklin DJ, Bolli R, Mohamed TMA. Gene therapy encoding cell cycle factors to treat chronic ischemic heart failure in rats. Cardiovasc Res 2024; 120:152-163. [PMID: 38175760 PMCID: PMC10936750 DOI: 10.1093/cvr/cvae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/25/2023] [Accepted: 10/07/2023] [Indexed: 01/06/2024] Open
Abstract
AIMS Gene therapies to induce cardiomyocyte (CM) cell cycle re-entry have shown a potential to treat subacute ischaemic heart failure (IHF) but have not been tested in the more relevant setting of chronic IHF. Our group recently showed that polycistronic non-integrating lentivirus encoding Cdk1/CyclinB1 and Cdk4/CyclinD1 (TNNT2-4Fpolycistronic-NIL) is effective in inducing CM cell cycle re-entry and ameliorating subacute IHF models and preventing the subsequent IHF-induced congestions in the liver, kidneys, and lungs in rats and pigs. Here, we aim to test the long-term efficacy of TNNT2-4Fpolycistronic-NIL in a rat model of chronic IHF, a setting that differs pathophysiologically from subacute IHF and has greater clinical relevance. METHODS AND RESULTS Rats were subjected to a 2-h coronary occlusion followed by reperfusion; 4 weeks later, rats were injected intramyocardially with either TNNT2-4Fpolycistronic-NIL or LacZ-NIL. Four months post-viral injection, TNNT2-4Fpolycistronic-NIL-treated rats showed a significant reduction in scar size and a significant improvement in left ventricular (LV) systolic cardiac function but not in the LV dilatation associated with chronic IHF. A mitosis reporter system developed in our lab showed significant induction of CM mitotic activity in TNNT2-4Fpolycistronic-NIL-treated rats. CONCLUSION This study demonstrates, for the first time, that TNNT2-4Fpolycistronic-NIL gene therapy induces CM cell cycle re-entry in chronic IHF and improves LV function, and that this salubrious effect is sustained for at least 4 months. Given the high prevalence of chronic IHF, these results have significant clinical implications for developing a novel treatment for this deadly disease.
Collapse
Affiliation(s)
- Riham R E Abouleisa
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Xian-Liang Tang
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Qinghui Ou
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Abou-Bakr M Salama
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Cardiovascular Medicine, Faculty of Medicine, Zagazig University, 872 Shaibet an Nakareyah, Zagazig, Al-Sharqia Governorate 7120001, Egypt
| | - Amie Woolard
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Dana Hammouri
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Hania Abdelhafez
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Bioengineering, Speed School of Engineering, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Sarah Cayton
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Sameeha K Abdulwali
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- College of Medicine, Alfaisal University, Interconnection of Al Takhassousi،Al Zahrawi Street, Riyadh 11533, Saudi Arabia
| | - Momo Arai
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- College of Medicine, Alfaisal University, Interconnection of Al Takhassousi،Al Zahrawi Street, Riyadh 11533, Saudi Arabia
| | - Israel D Sithu
- Department of Medicine, Center for Cardiometabolic Science, Envirome Institute, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Physiology, School of Medicine, University of Louisville, Louisville, 580 South Preston Street, KY 40202, USA
| | - Daniel J Conklin
- Department of Medicine, Center for Cardiometabolic Science, Envirome Institute, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Roberto Bolli
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
| | - Tamer M A Mohamed
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Bioengineering, Speed School of Engineering, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Medicine, Center for Cardiometabolic Science, Envirome Institute, University of Louisville, 580 South Preston Street, Louisville, KY 40202, USA
- Department of Biochemistry Faculty of Pharmacy, Zagazig University, 872 Shaibet an Nakareyah, Zagazig, Zagazig, Al-Sharqia Governorate 7120001, Egypt
- Institute of Cardiovascular Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Surgery Department, Baylor College of Medicine, 6519 Fannin Street, Houston, TX, 77030, USA
| |
Collapse
|
20
|
Stougiannou TM, Christodoulou KC, Dimarakis I, Mikroulis D, Karangelis D. To Repair a Broken Heart: Stem Cells in Ischemic Heart Disease. Curr Issues Mol Biol 2024; 46:2181-2208. [PMID: 38534757 PMCID: PMC10969169 DOI: 10.3390/cimb46030141] [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: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Despite improvements in contemporary medical and surgical therapies, cardiovascular disease (CVD) remains a significant cause of worldwide morbidity and mortality; more specifically, ischemic heart disease (IHD) may affect individuals as young as 20 years old. Typically managed with guideline-directed medical therapy, interventional or surgical methods, the incurred cardiomyocyte loss is not always completely reversible; however, recent research into various stem cell (SC) populations has highlighted their potential for the treatment and perhaps regeneration of injured cardiac tissue, either directly through cellular replacement or indirectly through local paracrine effects. Different stem cell (SC) types have been employed in studies of infarcted myocardium, both in animal models of myocardial infarction (MI) as well as in clinical studies of MI patients, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), Muse cells, multipotent stem cells such as bone marrow-derived cells, mesenchymal stem cells (MSCs) and cardiac stem and progenitor cells (CSC/CPCs). These have been delivered as is, in the form of cell therapies, or have been used to generate tissue-engineered (TE) constructs with variable results. In this text, we sought to perform a narrative review of experimental and clinical studies employing various stem cells (SC) for the treatment of infarcted myocardium within the last two decades, with an emphasis on therapies administered through thoracic incision or through percutaneous coronary interventions (PCI), to elucidate possible mechanisms of action and therapeutic effects of such cell therapies when employed in a surgical or interventional manner.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Konstantinos C. Christodoulou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Ioannis Dimarakis
- Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, WA 98195, USA;
| | - Dimitrios Mikroulis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Dimos Karangelis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| |
Collapse
|
21
|
Yuan X, Braun T. Toward drug-induced heart regeneration. NATURE CARDIOVASCULAR RESEARCH 2024; 3:260-261. [PMID: 39196122 DOI: 10.1038/s44161-024-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
22
|
Della Rocca Y, Diomede F, Konstantinidou F, Trubiani O, Soundara Rajan T, Pierdomenico SD, Gatta V, Stuppia L, Marconi GD, Pizzicannella J. Protective effect of oral stem cells extracellular vesicles on cardiomyocytes in hypoxia-reperfusion. Front Cell Dev Biol 2024; 11:1260019. [PMID: 38288344 PMCID: PMC10823008 DOI: 10.3389/fcell.2023.1260019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/21/2023] [Indexed: 01/31/2024] Open
Abstract
Hypoxia signaling plays an important role in physiological and pathological conditions. Hypoxia in the heart tissue can produce different consequences depending on the duration of exposure to the hypoxic state. While acute hypoxic exposure leads to a reversible acclimatization in heart tissue with normal systemic oxygen supply, chronic hypoxia exacerbates cardiac dysfunction, leads to a destruction of the tissue. Extracellular vesicles (EVs) are small membrane vesicles that act as mediators of intercellular communication. EVs are secreted by different cell types and those produced by oral cavity-derived mesenchymal stem cells (MSCs), including human gingival MSCs (hGMSCs), have pro-angiogenic and anti-inflammatory effects and showed therapeutic role in tissue regeneration. The aim of the present work was to evaluate the potential protective and regenerative role of EVs produced by hGMSCs, in an in vitro model of hypoxia-conditioned HL-1 cardiomyocytes through the expression analysis of following inflammatory, oxidative stress, angiogenesis, cell survival and apoptotic markers: HIF-1α, P300, NFkB, CCL2, IL1B, IL6, NRF2, CASP-3, BAX and VEGF. Results showed that hGMSCs-derived EVs exerted protection HL-1 cardiomyocytes exposed to both pre and post hypoxic conditions. Moreover, modulation of CASP3 and BAX expression demonstrated that EVs reduced the apoptosis. The analysis of microRNAs in EVs derived from hGMSCs was performed to assess the epigenetic regulation of the presented markers. The following microRNAs: hsa-miR-138-5p, hsa-miR-17-5p, hsa-miR-18a-5p, hsa-miR-21-5p, hsa-miR-324-5p, hsa-miR-133a-3p, hsa-miR-150-5p, hsa-miR-199a-5p, hsa-miR-128-3p and hsa-miR-221-3p can directly or indirectly target the studied genes by determining their modulation obtained in our study. The data from this study suggested that EVs obtained from hGMSCs may be considered for the cell free treatment option in hypoxia-driven cardiac tissue dysfunction.
Collapse
Affiliation(s)
- Ylenia Della Rocca
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Fanì Konstantinidou
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | - Sante D. Pierdomenico
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. D'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Guya Diletta Marconi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Jacopo Pizzicannella
- Department of Engineering and Geology, University “G. D’ Annunzio” Chieti-Pescara, Pescara, Italy
| |
Collapse
|
23
|
Li J, Tang Y, Yin L, Lin X, Luo Z, Wang S, Yuan L, Liang P, Jiang B. Mesenchymal stem cell-derived exosomes in myocardial infarction: Therapeutic potential and application. J Gene Med 2024; 26:e3596. [PMID: 37726968 DOI: 10.1002/jgm.3596] [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: 06/15/2023] [Revised: 08/15/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023] Open
Abstract
Myocardial infarction refers to the irreversible impairment of cardiac function resulting from the permanent loss of numerous cardiomyocytes and the formation of scar tissue. This condition is caused by acute and persistent inadequate blood supply to the heart's arteries. In the treatment of myocardial infarction, Mesenchymal stem cells (MSCs) play a crucial role because of their powerful therapeutic effects. These effects primarily stem from the paracrine secretion of multiple factors by MSCs, with exosome-carried microRNAs being the most effective component in promoting cardiac function recovery after infarction. Exosome therapy has emerged as a promising cell-free treatment for myocardial infarction as a result of its relatively simple composition, low immunogenicity and controlled transplantation dose. Despite these advantages, maintaining the stability of exosomes after transplantation and enhancing their targeting effect remain significant challenges in clinical applications. In recent developments, several approaches have been designed to optimize exosome therapy. These include enhancing exosome retention, improving their ability to target specific effects, pretreating MSC-derived exosomes and employing transgenic MSC-derived exosomes. This review primarily focuses on describing the biological characteristics of exosomes, their therapeutic potential and their application in treating myocardial infarction.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Yuting Tang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Leijing Yin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Xiaofang Lin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Zhengyang Luo
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Shuxin Wang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Ludong Yuan
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bimei Jiang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Shin K, Begeman IJ, Cao J, Kang J. leptin b and its regeneration enhancer illustrate the regenerative features of zebrafish hearts. Dev Dyn 2024; 253:91-106. [PMID: 36495292 PMCID: PMC10256838 DOI: 10.1002/dvdy.556] [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: 07/14/2022] [Revised: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Zebrafish possess a remarkable regenerative capacity, which is mediated by the induction of various genes upon injury. Injury-dependent transcription is governed by the tissue regeneration enhancer elements (TREEs). Here, we utilized leptin b (lepb), an injury-specific factor, and its TREE to dissect heterogeneity of noncardiomyocytes (CMs) in regenerating hearts. RESULTS Our single-cell RNA sequencing (scRNA-seq) analysis demonstrated that the endothelium/endocardium(EC) is activated to induce distinct subpopulations upon injury. We demonstrated that lepb can be utilized as a regeneration-specific marker to subset injury-activated ECs. lepb+ ECs robustly induce pro-regenerative factors, implicating lepb+ ECs as a signaling center to interact with other cardiac cells. Our scRNA-seq analysis identified that lepb is also produced by subpopulation of epicardium (Epi) and epicardium-derived cells (EPDCs). To determine whether lepb labels injury-emerging non-CM cells, we tested the activity of lepb-linked regeneration enhancer (LEN) with chromatin accessibility profiles and transgenic lines. While nondetectable in uninjured hearts, LEN directs EC and Epi/EPDC expression upon injury. The endogenous LEN activity was assessed using LEN deletion lines, demonstrating that LEN deletion abolished injury-dependent expression of lepb, but not other nearby genes. CONCLUSIONS Our integrative analyses identify regeneration-emerging cell-types and factors, leading to the discovery of regenerative features of hearts.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Ian J. Begeman
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10021, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| |
Collapse
|
25
|
Li M, Zhang C, Tan L, Liu T, Zhu T, Wei X, Liu J, Si X, Li B. MiR-431 promotes cardiomyocyte proliferation by targeting FBXO32 expression. J Gene Med 2024; 26:e3656. [PMID: 38282147 DOI: 10.1002/jgm.3656] [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: 07/23/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND The induction of cardiomyocyte (CM) proliferation is a promising approach for cardiac regeneration following myocardial injury. MicroRNAs (miRNAs) have been reported to regulate CM proliferation. In particular, miR-431 expression decreases during cardiac development, according to Gene Expression Omnibus (GEO) microarray data. However, whether miR-431 regulates CM proliferation has not been thoroughly investigated. METHODS We used integrated bioinformatics analysis of GEO datasets to identify the most significantly differentially expressed miRNAs. Real-time quantitative PCR and fluorescence in situ hybridization were performed to determine the miRNA expression patterns in hearts. Gain- and loss-of-function assays were conducted to detect the role of miRNA in CM proliferation. Additionally, we detected whether miR-431 affected CM proliferation in a myocardial infarction model. The TargetScan, miRDB and miRWalk online databases were used to predict the potential target genes of miRNAs. Luciferase reporter assays were used to study miRNA interactions with the targeting mRNA. RESULTS First, we found a significant reduction in miR-431 levels during cardiac development. Then, by overexpression and inhibition of miR-431, we demonstrated that miR-431 promotes CM proliferation in vitro and in vivo, as determined by immunofluorescence assays of 5-ethynyl-2'-deoxyuridine (EdU), pH3, Aurora B and CM count, whereas miR-431 inhibition suppresses CM proliferation. Then, we found that miR-431 improved cardiac function post-myocardial infarction. In addition, we identified FBXO32 as a direct target gene of miR-431, with FBXO32 mRNA and protein expression being suppressed by miR-431. FBXO32 inhibited CM proliferation. Overexpression of FBXO32 blocks the enhanced effect of miR-431 on CM proliferation, suggesting that FBXO32 is a functional target of miR-431 during CM proliferation. CONCLUSION In summary, miR-431 promotes CM proliferation by targeting FBXO32, providing a potential molecular target for preventing myocardial injury.
Collapse
Affiliation(s)
- Mengsha Li
- Panzhou Renze Hospital, Panzhou, Guizhou, China
| | - Chenrui Zhang
- Guizhou University Medical College, Guiyang, Guizhou, China
| | - Lirong Tan
- Guizhou University Medical College, Guiyang, Guizhou, China
| | - Tingyan Liu
- Panzhou Renze Hospital, Panzhou, Guizhou, China
| | - Tingting Zhu
- Guizhou University Medical College, Guiyang, Guizhou, China
| | - Xuejiao Wei
- Guizhou University Medical College, Guiyang, Guizhou, China
| | - Jiacai Liu
- Panzhou People's Hospital, Panzhou, Guizhou, China
| | - Xiaoyun Si
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Bing Li
- Guizhou University Medical College, Guiyang, Guizhou, China
| |
Collapse
|
26
|
Beisaw A, Wu CC. Cardiomyocyte maturation and its reversal during cardiac regeneration. Dev Dyn 2024; 253:8-27. [PMID: 36502296 DOI: 10.1002/dvdy.557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/03/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Due to the limited proliferative and regenerative capacity of adult cardiomyocytes, the lost myocardium is not replenished efficiently and is replaced by a fibrotic scar, which eventually leads to heart failure. Current therapies to cure or delay the progression of heart failure are limited; hence, there is a pressing need for regenerative approaches to support the failing heart. Cardiomyocytes undergo a series of transcriptional, structural, and metabolic changes after birth (collectively termed maturation), which is critical for their contractile function but limits the regenerative capacity of the heart. In regenerative organisms, cardiomyocytes revert from their terminally differentiated state into a less mature state (ie, dedifferentiation) to allow for proliferation and regeneration to occur. Importantly, stimulating adult cardiomyocyte dedifferentiation has been shown to promote morphological and functional improvement after myocardial infarction, further highlighting the importance of cardiomyocyte dedifferentiation in heart regeneration. Here, we review several hallmarks of cardiomyocyte maturation, and summarize how their reversal facilitates cardiomyocyte proliferation and heart regeneration. A detailed understanding of how cardiomyocyte dedifferentiation is regulated will provide insights into therapeutic options to promote cardiomyocyte de-maturation and proliferation, and ultimately heart regeneration in mammals.
Collapse
Affiliation(s)
- Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
| | - Chi-Chung Wu
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
27
|
Navab R, Haward R, Chacko J, Haward R. Platelet-Rich Plasma for Heart Cell Regeneration Post-myocardial Infarction: A Propitious Therapeutic Approach. Cureus 2024; 16:e51951. [PMID: 38333505 PMCID: PMC10852202 DOI: 10.7759/cureus.51951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 02/10/2024] Open
Abstract
Globally, one of the primary factors leading to death is cardiovascular disorders, specifically coronary artery disease, which leads to myocardial infarction (MI). This article investigates the potential of platelet-rich plasma (PRP) therapy for regenerating cardiac cells following MI. We look into the pathophysiology of MI, current treatment methods, and the heart's limited ability to heal itself. This is done to see if PRP could help the heart heal faster, reduce the size of the infarct, and stop scar tissue from forming. We analyze the production procedure of PRP, its composition of growth factors, and its utilization in many medical domains. The ways that PRP helps the heart heal are also being looked into. This includes how it affects inflammation, oxidative stress, angiogenesis, and cell proliferation. Although we recognize the existing constraints, we meticulously take into account issues such as standardization, therapeutic variance, and potential harmful effects. This study highlights the importance of comprehensive guidelines, continuous research, and enhanced clinical applications to fully harness the potential of platelet-rich plasma in the regeneration of cardiac cells after a heart attack.
Collapse
Affiliation(s)
- Rahul Navab
- Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, IND
| | - Raymond Haward
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Joshua Chacko
- Internal Medicine, Father Muller Medical College, Mangalore, IND
| | - Rachel Haward
- Internal Medicine, KVG Medical College and Hospital, Sullia, IND
| |
Collapse
|
28
|
Houser SR. Will Induction of Transient Myocyte Proliferation Be a Regenerative Therapy? Circ Res 2023; 133:505-507. [PMID: 37651544 DOI: 10.1161/circresaha.123.323399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Affiliation(s)
- Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA
| |
Collapse
|
29
|
Romitti M, Costagliola S. Progress Toward and Challenges Remaining for Thyroid Tissue Regeneration. Endocrinology 2023; 164:bqad136. [PMID: 37690118 PMCID: PMC10516459 DOI: 10.1210/endocr/bqad136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
Thyroid hormones play a pivotal role in diverse physiological processes, and insufficient synthesis of these hormones results in hypothyroidism, a prevalent disorder with a significant global impact. Research has shown that the residual thyroid tissue following surgery fails to fully regenerate the gland and restore normal function. The slow turnover rate of the thyroid gland and the presence of resident stem cells, which may contribute to regeneration within adult thyroid tissue, are topics of ongoing debate. This comprehensive review summarizes current research findings concerning the regeneration of the adult thyroid. Investigations have identified potential cellular mechanisms implicated in thyroid regeneration following partial tissue damage, including cells within microfollicles and a cluster of potential thyroid progenitors cells. Nevertheless, the exact mechanisms remain elusive. In cases of complete removal of the thyroid gland, regeneration does not occur, underscoring the necessity for an external source of thyroid tissue. The transplantation of thyroid organoids has emerged as a promising approach to restore thyroid function. Researchers have successfully derived thyroid organoids from various sources and demonstrated their functionality in both in vitro and in vivo animal models. Despite the challenges that still need to be addressed in achieving full maturation and functionality of human thyroid organoids, significant strides have been made in this regard. This review explores the potential of thyroid organoid transplantation and its implications for the field of regenerative medicine.
Collapse
Affiliation(s)
- Mírian Romitti
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sabine Costagliola
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
30
|
Alipour Symakani RS, van Genuchten WJ, Zandbergen LM, Henry S, Taverne YJHJ, Merkus D, Helbing WA, Bartelds B. The right ventricle in tetralogy of Fallot: adaptation to sequential loading. Front Pediatr 2023; 11:1098248. [PMID: 37009270 PMCID: PMC10061113 DOI: 10.3389/fped.2023.1098248] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/27/2023] [Indexed: 04/04/2023] Open
Abstract
Right ventricular dysfunction is a major determinant of outcome in patients with complex congenital heart disease, as in tetralogy of Fallot. In these patients, right ventricular dysfunction emerges after initial pressure overload and hypoxemia, which is followed by chronic volume overload due to pulmonary regurgitation after corrective surgery. Myocardial adaptation and the transition to right ventricular failure remain poorly understood. Combining insights from clinical and experimental physiology and myocardial (tissue) data has identified a disease phenotype with important distinctions from other types of heart failure. This phenotype of the right ventricle in tetralogy of Fallot can be described as a syndrome of dysfunctional characteristics affecting both contraction and filling. These characteristics are the end result of several adaptation pathways of the cardiomyocytes, myocardial vasculature and extracellular matrix. As long as the long-term outcome of surgical correction of tetralogy of Fallot remains suboptimal, other treatment strategies need to be explored. Novel insights in failure of adaptation and the role of cardiomyocyte proliferation might provide targets for treatment of the (dysfunctional) right ventricle under stress.
Collapse
Affiliation(s)
- Rahi S. Alipour Symakani
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
- Department of Cardiology, Division of Experimental Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Wouter J. van Genuchten
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
| | - Lotte M. Zandbergen
- Department of Cardiology, Division of Experimental Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, Munich, Germany
| | - Surya Henry
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Daphne Merkus
- Department of Cardiology, Division of Experimental Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Willem A. Helbing
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
| | - Beatrijs Bartelds
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
| |
Collapse
|
31
|
Singh BN, Yucel D, Garay BI, Tolkacheva EG, Kyba M, Perlingeiro RCR, van Berlo JH, Ogle BM. Proliferation and Maturation: Janus and the Art of Cardiac Tissue Engineering. Circ Res 2023; 132:519-540. [PMID: 36795845 PMCID: PMC9943541 DOI: 10.1161/circresaha.122.321770] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.
Collapse
Affiliation(s)
- Bhairab N. Singh
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
| | - Dogacan Yucel
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Bayardo I. Garay
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Medical Scientist Training Program, University of Minnesota Medical School, MN, USA
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Rita C. R. Perlingeiro
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Jop H. van Berlo
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Brenda M. Ogle
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
- Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
32
|
Løgstrup BB. Heart Failure in Rheumatic Disease: Secular Trends and Novel Insights. Rheum Dis Clin North Am 2023; 49:67-79. [PMID: 36424027 DOI: 10.1016/j.rdc.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is a significant increase in risk of heart failure in several rheumatic diseases. Common cardiovascular risk factors and inflammatory processes, present in both rheumatic diseases and heart failure, are contributing to this increase. The opportunities for using immune-based strategies to fight development of heart failure in rheumatic diseases are evolving. The diversity of inflammation calls for a tailored characterization of inflammation, enabling differentiation of inflammation and subsequent introduction of precision medicine using target-specific strategies and immunomodulatory therapy. As the field of rheuma-cardiology is still evolving, clear recommendations cannot be given yet.
Collapse
Affiliation(s)
- Brian Bridal Løgstrup
- Department of Cardiology, Institute of Clinical Medicine, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, Aarhus N 8200, Denmark.
| |
Collapse
|
33
|
Kanda M, Nagai T, Kondo N, Matsuura K, Akazawa H, Komuro I, Kobayashi Y. Pericardial Grafting of Cardiac Progenitor Cells in Self-Assembling Peptide Scaffold Improves Cardiac Function After Myocardial Infarction. Cell Transplant 2023; 32:9636897231174078. [PMID: 37191272 PMCID: PMC10192947 DOI: 10.1177/09636897231174078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Many studies have explored cardiac progenitor cell (CPC) therapy for heart disease. However, optimal scaffolds are needed to ensure the engraftment of transplanted cells. We produced a three-dimensional hydrogel scaffold (CPC-PRGmx) in which high-viability CPCs were cultured for up to 8 weeks. CPC-PRGmx contained an RGD peptide-conjugated self-assembling peptide with insulin-like growth factor-1 (IGF-1). Immediately after creating myocardial infarction (MI), we transplanted CPC-PRGmx into the pericardial space on to the surface of the MI area. Four weeks after transplantation, red fluorescent protein-expressing CPCs and in situ hybridization analysis in sex-mismatched transplantations revealed the engraftment of CPCs in the transplanted scaffold (which was cellularized with host cells). The average scar area of the CPC-PRGmx-treated group was significantly smaller than that of the non-treated group (CPC-PRGmx-treated group = 46 ± 5.1%, non-treated MI group = 59 ± 4.5%; p < 0.05). Echocardiography showed that the transplantation of CPC-PRGmx improved cardiac function and attenuated cardiac remodeling after MI. The transplantation of CPCs-PRGmx promoted angiogenesis and inhibited apoptosis, compared to the untreated MI group. CPCs-PRGmx secreted more vascular endothelial growth factor than CPCs cultured on two-dimensional dishes. Genetic fate mapping revealed that CPC-PRGmx-treated mice had more regenerated cardiomyocytes than non-treated mice in the MI area (CPC-PRGmx-treated group = 0.98 ± 0.25%, non-treated MI group = 0.25 ± 0.04%; p < 0.05). Our findings reveal the therapeutic potential of epicardial-transplanted CPC-PRGmx. Its beneficial effects may be mediated by sustainable cell viability, paracrine function, and the enhancement of de novo cardiomyogenesis.
Collapse
Affiliation(s)
- Masato Kanda
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Nagai
- Department of Cardiology, Chemotherapy
Research Institute, KAKEN Hospital, International University of Health and Welfare,
Ichikawa-shi, Japan
| | - Naomichi Kondo
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical
Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women’s
Medical University, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
34
|
Vrachatis DA, Papathanasiou KA, Giotaki SG, Raisakis K, Kaoukis A, Kossyvakis C, Theodorakis A, Pediotidis S, Avramides D, Siasos G, Deftereos S. Advances in the Management of Heart Failure with Reduced Ejection Fraction; The Role of SGLT2is, ARNI, Myotropes, Vericiguat, and Anti-inflammatory Agents: A Mini-review. Curr Pharm Des 2023; 29:509-518. [PMID: 36927423 DOI: 10.2174/1381612829666230316142450] [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: 08/25/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 03/18/2023]
Abstract
Heart failure with reduced ejection fraction (HFrEF) has been associated with poor prognosis, reduced quality of life, and increased healthcare expenditure. Despite tremendous advances in HFrEF management, reduced survival and a high rate of hospitalization remain unsolved issues. Furthermore, HFrEF morbidity and economic burden are estimated to increase in the following years; hence, new therapies are constantly emerging. In the last few years, a series of landmark clinical trials have expanded our therapeutic armamentarium with a ground-breaking change in HFrEF-related outcomes. Sodium-glucose co-transporter 2 inhibitors (mainly dapagliflozin and empagliflozin) have already revolutionized the management of HFrEF patients via a significant reduction in cardiovascular mortality and heart failure hospitalizations. Furthermore, vericiguat and omecamtiv mecarbil have emerged as promising and novel disease-modifying therapies. The former restores the impaired cyclic guanosine monophosphate pathway, and the latter stimulates cardiac myosin without marked arrhythmogenesis. Both vericiguat and omecamtiv mecarbil have been shown to reduce heart failure admissions. Sacubitril/valsartan is an established and effective therapy in HFrEF patients and should be considered as a replacement for angiotensin-converting enzyme inhibitors (ACEi) or angiotensin II receptor blockers (ARBs). Lastly, inflammasome activity is implicated in HFrEF pathophysiology, and the role of anti-inflammatory agents in HFrEF trajectories is readily scrutinized, yet available therapies are ineffective. This mini-review summarizes the major and most recent studies in this field, thus covering the current advances in HFrEF therapeutics.
Collapse
Affiliation(s)
| | - Konstantinos A Papathanasiou
- Second Department of Cardiology, National & Kapodistrian University of Athens, School of Medicine, University General Hospital ATTIKON, Athens, Greece
| | - Sotiria G Giotaki
- Second Department of Cardiology, National & Kapodistrian University of Athens, School of Medicine, University General Hospital ATTIKON, Athens, Greece
| | | | - Andreas Kaoukis
- Deparment of Cardiology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | | | - Andreas Theodorakis
- Deparment of Cardiology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Stauros Pediotidis
- Deparment of Cardiology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Dimitrios Avramides
- Deparment of Cardiology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Gerasimos Siasos
- Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Spyridon Deftereos
- Department of Cardiology, National & Kapodistrian University of Athens, School of Medicine, University General Hospital ATTIKON, Athens, Greece
| |
Collapse
|
35
|
Smits AM, Goumans MJ. Chasing youth on the outside: unique features of the human fetal epicardium. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1130-1131. [PMID: 39196167 DOI: 10.1038/s44161-022-00191-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Anke M Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.
| | - Marie Jose Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
36
|
Development of an injectable alginate-collagen hydrogel for cardiac delivery of extracellular vesicles. Int J Pharm 2022; 629:122356. [DOI: 10.1016/j.ijpharm.2022.122356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
|
37
|
Powers N, Huang GN. Mending a broken heart with novel cardiogenic small molecules. CELL REGENERATION 2022; 11:20. [PMID: 35511305 PMCID: PMC9072610 DOI: 10.1186/s13619-022-00120-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adult mammalian cardiomyocytes are unable to proliferate to regenerate lost tissue after heart injury. Du et al., reporting in Cell Stem Cell, employ a FUCCI- and MADM-based system to screen for small molecules combinations that produced a collaborative effect on cardiomyocyte cycling and cytokinesis. The authors generate a cocktail of five small molecules that increase cardiomyocyte proliferation and regeneration in vitro and in vivo with high efficiency, and explore its potential in cardiac regenerative repair after myocardial infarction through a new potential pathway for cardiomyocyte cell-cycle re-entry.
Collapse
|
38
|
Kalyuzhin VV, Teplyakov AT, Bespalova ID, Kalyuzhina EV, Terentyeva NN, Grakova EV, Kopeva KV, Usov VY, Garganeeva NP, Pavlenko OA, Gorelova YV, Teteneva AV. Promising directions in the treatment of chronic heart failure: improving old or developing new ones? BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-181-197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Unprecedented advances of recent decades in clinical pharmacology, cardiac surgery, arrhythmology, and cardiac pacing have significantly improved the prognosis in patients with chronic heart failure (CHF). However, unfortunately, heart failure continues to be associated with high mortality. The solution to this problem consists in simultaneous comprehensive use in clinical practice of all relevant capabilities of continuously improving methods of heart failure treatment proven to be effective in randomized controlled trials (especially when confirmed by the results of studies in real clinical practice), on the one hand, and in development and implementation of innovative approaches to CHF treatment, on the other hand. This is especially relevant for CHF patients with mildly reduced and preserved left ventricular ejection fraction, as poor evidence base for the possibility of improving the prognosis in such patients cannot justify inaction and leaving them without hope of a clinical improvement in their condition. The lecture consistently covers the general principles of CHF treatment and a set of measures aimed at inotropic stimulation and unloading (neurohormonal, volumetric, hemodynamic, and immune) of the heart and outlines some promising areas of disease-modifying therapy.
Collapse
Affiliation(s)
| | - A. T. Teplyakov
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | | | | | | | - E. V. Grakova
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - K. V. Kopeva
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - V. Yu. Usov
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | | | | | | | | |
Collapse
|
39
|
Liao R, Li Z, Wang Q, Lin H, Sun H. Revascularization of chronic total occlusion coronary artery and cardiac regeneration. Front Cardiovasc Med 2022; 9:940808. [PMID: 36093131 PMCID: PMC9455703 DOI: 10.3389/fcvm.2022.940808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary chronic total occlusion (CTO) contributes to the progression of heart failure in patients with ischemic cardiomyopathy. Randomized controlled trials demonstrated that percutaneous coronary intervention (PCI) for CTO significantly improves angina symptoms and quality of life but fails to reduce clinical events compared with optimal medical therapy. Even so, intervening physicians strongly support CTO-PCI. Cardiac regeneration therapy after CTO-PCI should be a promising approach to improving the prognosis of ischemic cardiomyopathy. However, the relationship between CTO revascularization and cardiac regeneration has rarely been studied, and experimental studies on cardiac regeneration usually employ rodent models with permanent ligation of the coronary artery rather than reopening of the occlusive artery. Limited early-stage clinical trials demonstrated that cell therapy for cardiac regeneration in ischemic cardiomyopathy reduces scar size, reverses cardiac remodeling, and promotes angiogenesis. This review focuses on the status quo of CTO-PCI in ischemic cardiomyopathy and the clinical prospect of cardiac regeneration in this setting.
Collapse
Affiliation(s)
- Ruoxi Liao
- Department of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Zhihong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiancheng Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hairuo Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Hairuo Lin, ,
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Huijun Sun,
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Cardiovascular diseases are the leading cause of death worldwide, largely due to the limited regenerative capacity of the adult human heart. In contrast, teleost zebrafish hearts possess natural regeneration capacity by proliferation of pre-existing cardiomyocytes after injury. Hearts of mice can regenerate if injured in a few days after birth, which coincides with the transient capacity for cardiomyocyte proliferation. This review tends to elaborate the roles and mechanisms of Wnt/β-catenin signaling in heart development and regeneration in mammals and non-mammalian vertebrates. RECENT FINDINGS Studies in zebrafish, mice, and human embryonic stem cells demonstrate the binary effect for Wnt/β-catenin signaling during heart development. Both Wnts and Wnt antagonists are induced in multiple cell types during cardiac development and injury repair. In this review, we summarize composites of the Wnt signaling pathway and their different action routes, followed by the discussion of their involvements in cardiac specification, proliferation, and patterning. We provide overviews about canonical and non-canonical Wnt activity during heart homeostasis, remodeling, and regeneration. Wnt/β-catenin signaling exhibits biphasic and antagonistic effects on cardiac specification and differentiation depending on the stage of embryogenesis. Inhibition of Wnt signaling is beneficial for cardiac wound healing and functional recovery after injury. Understanding of the roles and mechanisms of Wnt signaling pathway in injured animal hearts will contribute to the development of potential therapeutics for human diseased hearts.
Collapse
Affiliation(s)
- Dongliang Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jianjian Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China
| | - Tao P Zhong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
41
|
Mayerich D, Wythe JD. Computational insights on coronary artery function. NATURE CARDIOVASCULAR RESEARCH 2022; 1:691-693. [PMID: 37564924 PMCID: PMC10414787 DOI: 10.1038/s44161-022-00115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Collateral arteries may act as natural bypasses that reduce hypoperfusion after a coronary blockage. 3D imaging of neonatal and adult mouse hearts, plus human fetal and diseased adult hearts, is now used to computationally predict flow within the heart, and understand the cardioprotective role of collateral arteries in vivo.
Collapse
Affiliation(s)
- David Mayerich
- Department of Electrical and Computer Engineering, University of Houston, Houston, TX, USA
| | - Joshua D Wythe
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Renewal and Repair, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
42
|
Visualization of regenerating and repairing hearts. Clin Sci (Lond) 2022; 136:787-798. [PMID: 35621122 PMCID: PMC9886236 DOI: 10.1042/cs20211116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023]
Abstract
With heart failure continuing to become more prevalent, investigating the mechanisms of heart injury and repair holds much incentive. In contrast with adult mammals, other organisms such as teleost fish, urodele amphibians, and even neonatal mammals are capable of robust cardiac regeneration to replenish lost or damaged myocardial tissue. Long-term high-resolution intravital imaging of the behaviors and interactions of different cardiac cell types in their native environment could yield unprecedented insights into heart regeneration and repair. However, this task remains challenging for the heart due to its rhythmic contraction and anatomical location. Here, we summarize recent advances in live imaging of heart regeneration and repair, discuss the advantages and limitations of current systems, and suggest future directions for novel imaging technology development.
Collapse
|
43
|
|