51
|
Okamoto K, Matsunari H, Nakano K, Umeyama K, Hasegawa K, Uchikura A, Takayanagi S, Watanabe M, Ohgane J, Stirm M, Kurome M, Klymiuk N, Nagaya M, Wolf E, Nagashima H. Phenotypic features of genetically modified DMD-X KOX WT pigs. Regen Ther 2023; 24:451-458. [PMID: 37772130 PMCID: PMC10523442 DOI: 10.1016/j.reth.2023.09.010] [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: 06/26/2023] [Revised: 08/22/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disorder caused by mutation in the dystrophin gene (DMD) on the X chromosome. Female DMD carriers occasionally exhibit symptoms such as muscle weakness and heart failure. Here, we investigated the characteristics and representativeness of female DMD carrier (DMD-XKOXWT) pigs as a suitable disease model. Methods In vitro fertilization using sperm from a DMD-XKOY↔XWTXWT chimeric boar yielded DMD-XKOXWT females, which were used to generate F2 and F3 progeny, including DMD-XKOXWT females. F1-F3 piglets were genotyped and subjected to biochemical analysis for blood creatine kinase (CK), aspartate aminotransferase, and lactate dehydrogenase. Skeletal muscle and myocardial tissue were analyzed for the expression of dystrophin and utrophin, as well as for lymphocyte and macrophage infiltration. Results DMD-XKOXWT pigs exhibited various characteristics common to human DMD carrier patients, namely, asymptomatic hyperCKemia, dystrophin expression patterns in the skeletal and cardiac muscles, histopathological features of skeletal muscle degeneration, myocardial lesions in adulthood, and sporadic death. Pathological abnormalities observed in the skeletal muscles in DMD-XKOXWT pigs point to a frequent incidence of pathological abnormalities in the musculoskeletal tissues of latent DMD carriers. Our findings suggest a higher risk of myocardial abnormalities in DMD carrier women than previously believed. Conclusions We demonstrated that DMD-XKOXWT pigs could serve as a suitable large animal model for understanding the pathogenic mechanism in DMD carriers and developing therapies for female DMD carriers.
Collapse
Affiliation(s)
- Kazutoshi Okamoto
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Kazuaki Nakano
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Koki Hasegawa
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Ayuko Uchikura
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Shuko Takayanagi
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Jun Ohgane
- Laboratory of Genomic Function Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Mayuko Kurome
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Eckhard Wolf
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Hiroshi Nagashima
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| |
Collapse
|
52
|
Sethi Y, Padda I, Sebastian SA, Malhi A, Malhi G, Fulton M, Khehra N, Mahtani A, Parmar M, Johal G. Glucocorticoid Receptor Antagonism and Cardiomyocyte Regeneration Following Myocardial Infarction: A Systematic Review. Curr Probl Cardiol 2023; 48:101986. [PMID: 37481215 DOI: 10.1016/j.cpcardiol.2023.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Myocardial regeneration has been a topic of interest in literature and research in recent years. An evolving approach reported is glucocorticoid (GC) receptor antagonism and its role in the regeneration of cardiomyocytes. The authors of this study aim to explore the reported literature on GC receptor antagonism and its effects on cardiomyocyte remodeling, hypertrophy, scar formation, and ongoing cardiomyocyte death following cardiac injury. This article overviews cellular biology, mechanisms of action, clinical implications, challenges, and future considerations. The authors of this study conducted a systematic review utilizing the Cochrane methodology and PRISMA guidelines. This study includes data collected and interpreted from 30 peer-reviewed articles from 3 databases with the topic of interest. The mammalian heart has regenerative potential during its embryonic and fetal phases which is lost during its developmental processes. The microenvironment, intrinsic molecular mechanisms, and systemic and external factors impact cardiac regeneration. GCs influence these aspects in some cases. Consequently, GC receptor antagonism is emerging as a promising potential target for stimulating endogenous cardiomyocyte proliferation, aiding in cardiomyocyte regeneration following a cardiac injury such as a myocardial infarction (MI). Experimental studies on neonatal mice and zebrafish have shown promising results with GC receptor ablation (or brief pharmacological antagonism) promoting the survival of myocardial cells, re-entry into the cell cycle, and cellular division, resulting in cardiac muscle regeneration and diminished scar formation. Transient GC receptor antagonism has the potential to stimulate cardiomyocyte regeneration and help prevent the dreaded complications of MI. More trials based on human populations are encouraged to justify their applications and weigh the risk-benefit ratio.
Collapse
Affiliation(s)
- Yashendra Sethi
- Department of Medicine, Government Doon Medical College, Dehradun, Uttrakhand, India; PearResearch, Dehradun, Uttarakhand, India.
| | - Inderbir Padda
- Department of Medicine, Richmond University Medical Center, Staten Island, NY
| | | | - Amarveer Malhi
- Department of Medicine, Caribbean Medical University SOM, Willemstad, Curacao, The Netherlands
| | - Gurnaaz Malhi
- Department of Medicine, Caribbean Medical University SOM, Willemstad, Curacao, The Netherlands
| | - Matthew Fulton
- Department of Medicine, Richmond University Medical Center, Staten Island, NY
| | - Nimrat Khehra
- Department of Medicine, Saint James School of Medicine, Arnos Vale, Saint Vincent and the Grenadines
| | - Arun Mahtani
- Department of Medicine, Richmond University Medical Center, Staten Island, NY
| | - Mayur Parmar
- Department of Foundational Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL
| | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, WA
| |
Collapse
|
53
|
Allemann MS, Lee P, Beer JH, Saeedi Saravi SS. Targeting the redox system for cardiovascular regeneration in aging. Aging Cell 2023; 22:e14020. [PMID: 37957823 PMCID: PMC10726899 DOI: 10.1111/acel.14020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.
Collapse
Affiliation(s)
- Meret Sarah Allemann
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Pratintip Lee
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of CardiologyUniversity Hospital Zurich, University of ZurichSchlierenSwitzerland
| |
Collapse
|
54
|
Kmiotek-Wasylewska K, Bobis-Wozowicz S, Karnas E, Orpel M, Woźnicka O, Madeja Z, Dawn B, Zuba-Surma EK. Anti-inflammatory, Anti-fibrotic and Pro-cardiomyogenic Effects of Genetically Engineered Extracellular Vesicles Enriched in miR-1 and miR-199a on Human Cardiac Fibroblasts. Stem Cell Rev Rep 2023; 19:2756-2773. [PMID: 37700183 PMCID: PMC10661813 DOI: 10.1007/s12015-023-10621-2] [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] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
RATIONALE Emerging evidence indicates that stem cell (SC)- derived extracellular vesicles (EVs) carrying bioactive miRNAs are able to repair damaged or infarcted myocardium and ameliorate adverse remodeling. Fibroblasts represent a major cell population responsible for scar formation in the damaged heart. However, the effects of EVs on cardiac fibroblast (CFs) biology and function has not been investigated. OBJECTIVE To analyze the biological impact of stem cell-derived EVs (SC-EVs) enriched in miR-1 and miR-199a on CFs and to elucidate the underlying molecular mechanisms. METHODS AND RESULTS Genetically engineered human induced pluripotent stem cells (hiPS) and umbilical cord-derived mesenchymal stem cells (UC-MSCs) expressing miR-1 or miR-199a were used to produce miR-EVs. Cells and EVs were thoughtfully analyzed for miRNA expression using RT-qPCR method. Both hiPS-miRs-EVs and UC-MSC-miRs-EVs effectively transferred miRNAs to recipient CFs, however, hiPS-miRs-EVs triggered cardiomyogenic gene expression in CFs more efficiently than UC-MSC-miRs-EVs. Importantly, hiPS-miR-1-EVs exhibited cytoprotective effects on CFs by reducing apoptosis, decreasing levels of pro-inflammatory cytokines (CCL2, IL-1β, IL-8) and downregulating the expression of a pro-fibrotic gene - α-smooth muscle actin (α-SMA). Notably, we identified a novel role of miR-199a-3p delivered by hiPS-EVs to CFs, in triggering the expression of cardiomyogenic genes (NKX2.5, TNTC, MEF2C) and ion channels involved in cardiomyocyte contractility (HCN2, SCN5A, KCNJ2, KCND3). By targeting SERPINE2, miR-199a-3p may reduce pro-fibrotic properties of CFs, whereas miR-199a-5p targeted BCAM and TSPAN6, which may be implicated in downregulation of inflammation. CONCLUSIONS hiPS-EVs carrying miR-1 and miR-199a attenuate apoptosis and pro-fibrotic and pro-inflammatory activities of CFs, and increase cardiomyogenic gene expression. These finding serve as rationale for targeting fibroblasts with novel EV-based miRNA therapies to improve heart repair after myocardial injury.
Collapse
Affiliation(s)
- Katarzyna Kmiotek-Wasylewska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Sylwia Bobis-Wozowicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Monika Orpel
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Olga Woźnicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, 1701 W Charleston Blvd., Las Vegas, NV, 89102, USA
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| |
Collapse
|
55
|
Yuan X, Liu K, Dong P, Han H. Protective effect and mechanism of different proportions of " Danggui-Kushen" herb pair on ischemic heart disease. Heliyon 2023; 9:e22150. [PMID: 38034717 PMCID: PMC10685368 DOI: 10.1016/j.heliyon.2023.e22150] [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: 06/05/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
This study aims to investigate the protective effect and mechanism of "Danggui-Kushen" herb pair (DKHP) on ischemic heart disease (IHD). The rat model of myocardial reperfusion injury (MIRI) was established by ligation of the left anterior descending coronary artery. Rats were randomly divided into seven groups and administered orally for 7 days: control group, IHD group, DKHP1:1 group, DKHP1:2 group, DKHP2:1 group, DKHP1:3 group, DKHP3:1 group, the dosage was 2.7 g/kg. Measure electrocardiogram (ECG), myocardial infarction and injury assessment, Hematoxylin and eosin (HE) staining to evaluate myocardial injury and the protective effect of DKHP. Lactate dehydrogenase (LDH), Reactive oxygen species (ROS), IL-1β and IL-6 kit detection, immunohistochemical analysis, establishment of H9c2 cardiomyocyte hypoxia (Hypoxia) model, DKHP pretreatment for 3 h, MTT method to detect cell survival rate, cell immunofluorescence to observe NF- The expression of TLR-4, NF-κB, p-NF-κB, IKβα, p-IKβα, HIF-1α, VEGF and other genes and proteins were detected by κB nuclear translocation, mitochondrial membrane potential measurement, Western blot and Polymerase Chain Reaction (PCR). Compared with the model group, DKHP can reduce the size of myocardial infarction, reduce the levels of factors such as LDH, ROS, IL-1β and IL-6, and improve the cell survival rate; Compared with the model group, DKHP can inhibit the nuclear transfer of NF-κB and reduce mitochondrial damage; the results of immunohistochemical analysis, PCR and Western blot showed that compared with the model group, DKHP can reduce TLR-4, p-NF-κB, Expression levels of p-IKβα, HIF-1α, VEGF and other proteins. Reveal that DKHP may play a protective role in ischemic heart disease by reducing inflammation and oxidative stress damage. DKHP may have protective effect on ischemic heart disease, and its mechanism may be through reducing inflammatory response and oxidative stress damage to achieve this protective effect.
Collapse
Affiliation(s)
- Xu Yuan
- College of Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Kemeng Liu
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Peiliang Dong
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hua Han
- College of Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| |
Collapse
|
56
|
Thamrin AMH, Soetisna TW, Ramadhani ANE, Hendarto H. Cell sheet transplantation for ischemic heart disease: a systematic review. Indian J Thorac Cardiovasc Surg 2023; 39:577-587. [PMID: 37885940 PMCID: PMC10597942 DOI: 10.1007/s12055-023-01554-z] [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: 03/15/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 10/28/2023] Open
Abstract
Objective Cell sheet transplantation is emerging as an appealing alternative for ischemic heart disease patients as it potentially can increase stem cell viability and retention. But the outcomes and safety of this treatment are still limited in literature and the result varies widely. We conduct a systematic review to look at the efficacy and safety of this promising transplantation method. Methods A systematic review was performed according to PRISMA guidelines. A comprehensive literature search was undertaken using the PubMed, Scopus, and Embase databases. Articles were thoroughly evaluated and analyzed. Results Seven publications about cell sheet transplantation for ischemic heart disease patients were included. The primary outcomes measured were left ventricular ejection fraction (LVEF) and New York Heart Association (NYHA) class. Safety measurement was depicted by cardiac-related readmission and deaths. The follow-up time ranged from 3 to 36 months for clinical outcomes and 8.5 years for safety outcomes. Cell sheet transplantation showed improvement in LVEF and NYHA class in most studies. Cardiac-related readmission and adverse events of cell sheet transplantation range from 0 to 30.4%, all were nonfatal as no cardiac-related death was reported. Patient preoperative status seems can affect the patient's response to cell sheet therapy. Conclusion Cell sheet transplantation can safely improve LVEF and NYHA class in ischemic heart disease patients, even in very low ejection fraction patients with unsuccessful standard therapy before. Further studies with better patient inclusion, larger population, and long-term follow-up required to confirm these results.
Collapse
Affiliation(s)
- Ahmad Muslim Hidayat Thamrin
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, S. Parman Street Cavling 87, Jakarta, Indonesia
- Faculty of Medicine Syarif Hidayatullah State Islamic University – Haji Hospital, Jakarta, Indonesia
| | - Tri Wisesa Soetisna
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, S. Parman Street Cavling 87, Jakarta, Indonesia
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Andi Nurul Erisya Ramadhani
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, S. Parman Street Cavling 87, Jakarta, Indonesia
| | - Hari Hendarto
- Faculty of Medicine Syarif Hidayatullah State Islamic University – Haji Hospital, Jakarta, Indonesia
| |
Collapse
|
57
|
Li W, Zhu Y, Wang W, He D, Feng L, Li Z. Src tyrosine kinase promotes cardiac remodeling induced by chronic sympathetic activation. Biosci Rep 2023; 43:BSR20231097. [PMID: 37650260 PMCID: PMC10611920 DOI: 10.1042/bsr20231097] [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: 06/14/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023] Open
Abstract
Cardiac remodeling serves as the underlying pathological basis for numerous cardiovascular diseases and represents a pivotal stage for intervention. The excessive activation of β-adrenergic receptors (β-ARs) assumes a crucial role in cardiac remodeling. Nonetheless, the underlying molecular mechanisms governing β-AR-induced cardiac remodeling remain largely unresolved. In the present study, we identified Src tyrosine kinase as a key player in the cardiac remodeling triggered by excessive β-AR activation. Our findings demonstrated that Src mediates isoproterenol (ISO)-induced cardiac hypertrophy, fibrosis, and inflammation in vivo. Furthermore, Src facilitates β-AR-mediated proliferation and transdifferentiation of cardiac fibroblasts, and hypertrophy and cardiomyocytes in vitro. Subsequent investigations have substantiated that Src mediates β-AR induced the extracellular signal-regulated protein kinase (ERK1/2) signaling pathway activated by β-AR. Our research presents compelling evidence that Src promotes β-AR-induced cardiac remodeling in both in vivo and in vitro settings. It establishes the promoting effect of the β-AR/Src/ERK signaling pathway on overall cardiac remodeling in cardiac fibroblasts and underscores the potential of Src as a therapeutic target for cardiac remodeling.
Collapse
Affiliation(s)
- Wenqi Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuzhong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Wenjing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences. Beijing 100191, China
| | - Dan He
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences. Beijing 100191, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences. Beijing 100191, China
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| |
Collapse
|
58
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
59
|
Wang C, Hou X, Guan Q, Zhou H, Zhou L, Liu L, Liu J, Li F, Li W, Liu H. RNA modification in cardiovascular disease: implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:412. [PMID: 37884527 PMCID: PMC10603151 DOI: 10.1038/s41392-023-01638-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/15/2023] [Accepted: 09/03/2023] [Indexed: 10/28/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world, with a high incidence and a youth-oriented tendency. RNA modification is ubiquitous and indispensable in cell, maintaining cell homeostasis and function by dynamically regulating gene expression. Accumulating evidence has revealed the role of aberrant gene expression in CVD caused by dysregulated RNA modification. In this review, we focus on nine common RNA modifications: N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), N4-acetylcytosine (ac4C), pseudouridine (Ψ), uridylation, adenosine-to-inosine (A-to-I) RNA editing, and modifications of U34 on tRNA wobble. We summarize the key regulators of RNA modification and their effects on gene expression, such as RNA splicing, maturation, transport, stability, and translation. Then, based on the classification of CVD, the mechanisms by which the disease occurs and progresses through RNA modifications are discussed. Potential therapeutic strategies, such as gene therapy, are reviewed based on these mechanisms. Herein, some of the CVD (such as stroke and peripheral vascular disease) are not included due to the limited availability of literature. Finally, the prospective applications and challenges of RNA modification in CVD are discussed for the purpose of facilitating clinical translation. Moreover, we look forward to more studies exploring the mechanisms and roles of RNA modification in CVD in the future, as there are substantial uncultivated areas to be explored.
Collapse
Affiliation(s)
- Cong Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xuyang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qing Guan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huiling Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, The Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lijun Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jijia Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Feng Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Haidan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
60
|
Cui H, Yu ZX, Huang Y, Hann SY, Esworthy T, Shen YL, Zhang LG. 3D printing of thick myocardial tissue constructs with anisotropic myofibers and perfusable vascular channels. BIOMATERIALS ADVANCES 2023; 153:213579. [PMID: 37566935 DOI: 10.1016/j.bioadv.2023.213579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Engineering of myocardial tissues has become a promising therapeutic strategy for treating myocardial infarction (MI). However, a significant challenge remains in generating clinically relevant myocardial tissues that possess native microstructural characteristics and fulfill the requirements for implantation within the human body. In this study, a thick 3D myocardial construct with anisotropic myofibers and perfusable branched vascular channels is created with clinically relevant dimensions using a customized beam-scanning stereolithography printing technique. To obtain tissue-specific matrix niches, a decellularized extracellular matrix microfiber-reinforced gelatin-based bioink is developed. The bioink plays a crucial role in facilitating the precise manufacturing of a hierarchical microstructure, enabling us to better replicate the physiological characteristics of the native myocardial tissue matrix in terms of structure, biomechanics, and bioactivity. Through the integration of the tailored bioink with our printing method, we demonstrate a biomimetic architecture, appropriate biomechanical properties, vascularization, and improved functionality of induced pluripotent stem cell-derived cardiomyocytes in the thick tissue construct in vitro. This work not only offers a novel and effective means to generate biomimetic heart tissue in vitro for the treatment of MI, but also introduces a potential methodology for creating clinically relevant tissue products to aid in other complex tissue/organ regeneration and disease model applications.
Collapse
Affiliation(s)
- Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Yimin Huang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Yin-Lin Shen
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, United States of America; Departments of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, United States of America; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, United States of America; Department of Medicine, The George Washington University, Washington, DC 20052, United States of America.
| |
Collapse
|
61
|
Guo C, Ji W, Yang W, Deng Q, Zheng T, Wang Z, Sui W, Zhai C, Yu F, Xi B, Yu X, Xu F, Zhang Q, Zhang W, Kong J, Zhang M, Zhang C. NKRF in Cardiac Fibroblasts Protects against Cardiac Remodeling Post-Myocardial Infarction via Human Antigen R. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303283. [PMID: 37667861 PMCID: PMC10602562 DOI: 10.1002/advs.202303283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/09/2023] [Indexed: 09/06/2023]
Abstract
Myocardial infarction (MI) remains the leading cause of death worldwide. Cardiac fibroblasts (CFs) are abundant in the heart and are responsible for cardiac repair post-MI. NF-κB-repressing factor (NKRF) plays a significant role in the transcriptional inhibition of various specific genes. However, the NKRF action mechanism in CFs remains unclear in cardiac repair post-MI. This study investigates the NKRF mechanism in cardiac remodeling and dysfunction post-MI by establishing a CF-specific NKRF-knockout (NKRF-CKO) mouse model. NKRF expression is downregulated in CFs in response to pathological cardiac remodeling in vivo and TNF-α in vitro. NKRF-CKO mice demonstrate worse cardiac function and survival and increased infarct size, heart weight, and MMP2 and MMP9 expression post-MI compared with littermates. NKRF inhibits CF migration and invasion in vitro by downregulating MMP2 and MMP9 expression. Mechanistically, NKRF inhibits human antigen R (HuR) transcription by binding to the classical negative regulatory element within the HuR promoter via an NF-κB-dependent mechanism. This decreases HuR-targeted Mmp2 and Mmp9 mRNA stability. This study suggests that NKRF is a therapeutic target for pathological cardiac remodeling.
Collapse
Affiliation(s)
- Chenghu Guo
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Wei Ji
- Department of UltrasonographyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinan250014China
| | - Wei Yang
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Qiming Deng
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Zunzhe Wang
- Department of Geriatric CardiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan250021China
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Chungang Zhai
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Fangpu Yu
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Bo Xi
- Department of EpidemiologySchool of Public HealthCheeloo College of MedicineShandong UniversityJinan250012China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of EducationDepartment of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinan250012China
| | - Feng Xu
- Department of Emergency MedicineChest Pain CenterShandong Provincial Clinical Research Center for Emergency and Critical Care MedicineQilu HospitalShandong UniversityJinan250012China
| | - Qunye Zhang
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Wencheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Jing Kong
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
- Cardiovascular Disease Research Center of Shandong First Medical UniversityCentral Hospital Affiliated to Shandong First Medical UniversityJinan250013China
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing TheoryThe Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of EducationChinese National Health Commission and Chinese Academy of Medical SciencesDepartment of CardiologyQilu Hospital of Shandong UniversityJinan250012China
- Cardiovascular Disease Research Center of Shandong First Medical UniversityCentral Hospital Affiliated to Shandong First Medical UniversityJinan250013China
| |
Collapse
|
62
|
Han X, Liu X, Zhao X, Wang X, Sun Y, Qu C, Liang J, Yang B. Dapagliflozin ameliorates sepsis-induced heart injury by inhibiting cardiomyocyte apoptosis and electrical remodeling through the PI3K/Akt pathway. Eur J Pharmacol 2023; 955:175930. [PMID: 37479014 DOI: 10.1016/j.ejphar.2023.175930] [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: 03/26/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Sepsis-induced heart injury is one of the leading causes of circulation disorders worldwide. Dapagliflozin, a sodium-glucose cotransporter 2 inhibitor mainly used for controlling blood glucose, has been shown to exert a protective effect on cardiomyocytes. However, the protective effect of dapagliflozin against sepsis-induced cardiac injury and the underlying mechanism needs to be studied. AIM This study aims to investigate the effect of dapagliflozin on sepsis-induced cardiomyopathy and the potential mechanisms involved. METHODS The rat model of sepsis was constructed by intraperitoneal injection of lipopolysaccharide. Echocardiography and electrophysiological studies were performed to detect changes in cardiac function and electrical activity. Cardiac pathological alternation and cardiomyocyte apoptosis were measured by H&E staining, serological analysis, immunohistochemical, immunofluorescence, and TUNEL assays. Western blot and qRT-PCR were performed to elucidate the underlying mechanism of dapagliflozin. Additionally, corresponding experiments in H9c2 cells were performed to further validate the mechanisms in vitro. RESULTS Dapagliflozin improved cardiac dysfunction and reduced the susceptibility to ventricular arrhythmias in sepsis rats by ameliorating cardiac inflammation, suppressing cardiomyocyte apoptosis, and alleviating ventricular electrical remodeling. The PI3K/Akt signaling pathway inhibitor inhibited the anti-apoptotic effect of dapagliflozin, indicating that the protective effect was related to the activation of the PI3K/Akt pathway. CONCLUSION Dapagliflozin ameliorated sepsis-induced cardiac injury by suppressing electrical remodeling and cardiomyocyte apoptosis, which could be attributed to the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Jinjun Liang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
63
|
Tufan Y, Öztatlı H, Doganay D, Buyuksungur A, Cicek MO, Döş İT, Berberoğlu Ç, Unalan HE, Garipcan B, Ercan B. Multifunctional Silk Fibroin/Carbon Nanofiber Scaffolds for In Vitro Cardiomyogenic Differentiation of Induced Pluripotent Stem Cells and Energy Harvesting from Simulated Cardiac Motion. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42271-42283. [PMID: 37643896 PMCID: PMC10510024 DOI: 10.1021/acsami.3c08601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
In this proof-of-concept study, cardiomyogenic differentiation of induced pluripotent stem cells (iPSCs) is combined with energy harvesting from simulated cardiac motion in vitro. To achieve this, silk fibroin (SF)-based porous scaffolds are designed to mimic the mechanical and physical properties of cardiac tissue and used as triboelectric nanogenerator (TENG) electrodes. The load-carrying mechanism, β-sheet content, degradation characteristics, and iPSC interactions of the scaffolds are observed to be interrelated and regulated by their pore architecture. The SF scaffolds with a pore size of 379 ± 34 μm, a porosity of 79 ± 1%, and a pore interconnectivity of 67 ± 1% upregulated the expression of cardiac-specific gene markers TNNT2 and NKX2.5 from iPSCs. Incorporating carbon nanofibers (CNFs) enhances the elastic modulus of the scaffolds to 45 ± 3 kPa and results in an electrical conductivity of 0.021 ± 0.006 S/cm. The SF and SF/CNF scaffolds are used as conjugate TENG electrodes and generate a maximum power output of 0.37 × 10-3 mW/m2, with an open-circuit voltage and a short circuit current of 0.46 V and 4.5 nA, respectively, under simulated cardiac motion. A novel approach is demonstrated for fabricating scaffold-based cardiac patches that can serve as tissue scaffolds and simultaneously allow energy harvesting.
Collapse
Affiliation(s)
- Yiğithan Tufan
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - Hayriye Öztatlı
- Institute
of Biomedical Engineering, Boğaziçi
University, 34684 İstanbul, Turkey
| | - Doga Doganay
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - Arda Buyuksungur
- Department
of Basic Medical Sciences, Faculty of Dentistry, Ankara University, 06560 Ankara, Turkey
| | - Melih Ogeday Cicek
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - İpek Tuğçe Döş
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - Çağla Berberoğlu
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - Husnu Emrah Unalan
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| | - Bora Garipcan
- Institute
of Biomedical Engineering, Boğaziçi
University, 34684 İstanbul, Turkey
| | - Batur Ercan
- Department
of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
- Biomedical
Engineering Program, Middle East Technical
University, Çankaya, 06800 Ankara, Turkey
- BIOMATEN,
Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Çankaya, 06800 Ankara, Turkey
| |
Collapse
|
64
|
Yang Z, Zhang Y, Wang J, Yin J, Wang Z, Pei R. Cardiac organoid: multiple construction approaches and potential applications. J Mater Chem B 2023; 11:7567-7581. [PMID: 37477533 DOI: 10.1039/d3tb00783a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The human cardiac organoid (hCO) is three-dimensional tissue model that is similar to an in vivo organ and has great potential on heart development biology, disease modeling, drug screening and regenerative medicine. However, the construction of hCO presents a unique challenge compared with other organoids such as the lung, small intestine, pancreas, liver. Since heart disease is the dominant cause of death and the treatment of such disease is one of the most unmet medical needs worldwide, developing technologies for the construction and application of hCO is a critical task for the scientific community. In this review, we discuss the current classification and construction methods of hCO. In addition, we describe its applications in drug screening, disease modeling, and regenerative medicine. Finally, we propose the limitations of the cardiac organoid and future research directions. A detailed understanding of hCO will provide ways to improve its construction and expand its applications.
Collapse
Affiliation(s)
- Ziyi Yang
- School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Jine Wang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Jingbo Yin
- School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Zheng Wang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China.
| |
Collapse
|
65
|
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
|
66
|
Fang Z, Li S, Yushanjiang F, Feng G, Cui S, Hu S, Jiang X, Liu C. Curcumol alleviates cardiac remodeling via the AKT/NF-κB pathway. Int Immunopharmacol 2023; 122:110527. [PMID: 37392572 DOI: 10.1016/j.intimp.2023.110527] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 07/03/2023]
Abstract
Cardiac remodeling is the final stage of almost all cardiovascular diseases, leading to heart failure and arrhythmias. However, the pathogenesis of cardiac remodeling is not fully understood, and specific treatment schemes are currently unavailable. Curcumol is a bioactive sesquiterpenoid that has anti-inflammatory, anti-apoptotic, and anti-fibrotic properties. This study aimed to investigate the protective effect of curcumol on cardiac remodeling and elucidate its relevant underlying mechanism. Curcumol significantly attenuated cardiac dysfunction, myocardial fibrosis, and hypertrophy in the animal model of isoproterenol (ISO)-induced cardiac remodeling. Curcumol also alleviated cardiac electrical remodeling, thereby reducing the risk of ventricular fibrillation (VF) after heart failure. Inflammation and apoptosis are critical pathological processes involved in cardiac remodeling. Curcumol inhibited the inflammation and apoptosis induced by ISO and TGF-β1 in mouse myocardium and neonatal rat cardiomyocytes (NRCMs). Furthermore, the protective effects of curcumol were found to be mediated through the inhibition of the protein kinase B (AKT)/nuclear factor-kappa B (NF-κB) pathway. The administration of an AKT agonist reversed the anti-fibrotic, anti-inflammatory, and anti-apoptotic effects of curcumol and restored the inhibition of NF-κB nuclear translocation in TGF-β1-induced NRCMs. Our study suggests that curcumol is a potential therapeutic agent for the treatment of cardiac remodeling.
Collapse
Affiliation(s)
- Zhao Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Shuang Li
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Feierkaiti Yushanjiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Gaoke Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Shengyu Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Chengyin Liu
- Department of Geriatrics, The Affiliated Hospital of Yangzhou University, Yangzhou 225000, China.
| |
Collapse
|
67
|
Zhang C, Hao H, Wang Y, Mu N, Jiang W, Zhang Z, Yin Y, Yu L, Chang ACY, Ma H. Intercellular mitochondrial component transfer triggers ischemic cardiac fibrosis. Sci Bull (Beijing) 2023; 68:1784-1799. [PMID: 37517989 DOI: 10.1016/j.scib.2023.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Myocardial fibrosis is the villain of sudden cardiac death. Myocardial ischemia/reperfusion (MI/R) injury induces cardiomyocyte damage or even death, which in turn stimulates fibroblast activation and fibrosis, but the intercellular communication mechanism remains unknown. Recent studies have shown that small extracellular vesicles (sEVs) significantly contribute to intercellular communication. Whether and how sEV might mediate post-MI/R cardiomyocyte/fibroblasts communication remain unknown. Here, in vivo and in vitro MI/R models were established. We demonstrate that sEVs derived from cardiomyocyte (Myo-sEVs) carry mitochondrial components, which enter fibroblasts to initiate myocardial fibrosis. Based on bioinformatics screening and experimental verification, the activating molecule in Beclin1-regulated autophagy protein 1 (autophagy/beclin-1 regulator 1, Ambra1) was found to be a critical component of these sEV and might be a new marker for Myo-sEVs. Interestingly, release of Ambra1+-Myo-sEVs was caused by secretory rather than canonical autophagy after MI/R injury and thereby escaped degradation. In ischemic and peripheral areas, Ambra1+-Myo-sEVs were internalized by fibroblasts, and the delivered mtDNA components to activate the fibroblast cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to promote fibroblast activation and proliferation. In addition, our data show that Ambra1 is expressed on the EV surface and cardiac-specific Ambra1 down regulation inhibits the Ambra1+-Myo-sEVs release and fibroblast uptake, effectively inhibiting ischemic myocardial fibrosis. This finding newly provides the evidence that myocardial secretory autophagy plays a role in intercellular communication during cardiac fibrosis. Ambra1 is a newly characterized molecule with bioactivity and might be a marker for Myo-sEVs, providing new therapeutic targets for cardiac remodeling.
Collapse
Affiliation(s)
- Chan Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Hao Hao
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wenhua Jiang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zihui Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Alex Chia Yu Chang
- Department of Cardiology and Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 211125, China.
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
68
|
Nair RS, Sobhan PK, Shenoy SJ, Prabhu MA, Kumar V, Ramachandran S, Anilkumar TV. Mitigation of Fibrosis after Myocardial Infarction in Rats by Using a Porcine Cholecyst Extracellular Matrix. Comp Med 2023; 73:312-323. [PMID: 37527924 PMCID: PMC10702285 DOI: 10.30802/aalas-cm-22-000097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/27/2022] [Accepted: 12/09/2022] [Indexed: 08/03/2023]
Abstract
Fibrosis that occurs after nonfatal myocardial infarction (MI) is an irreversible reparative cardiac tissue remodeling process characterized by progressive deposition of highly cross-linked type I collagen. No currently available therapeutic strategy prevents or reverses MI-associated fibrotic scarring of myocardium. In this study, we used an epicardial graft prepared of porcine cholecystic extracellular matrix to treat experimental nonfatal MI in rats. Graft-assisted healing was characterized by reduced fibrosis, with scanty deposition of type I collagen. Histologically, the tissue response was associated with a favorable regenerative reaction predominated by CD4-positive helper T lymphocytes, enhanced angiogenesis, and infiltration of proliferating cells. These observations indicate that porcine cholecystic extracellular matrix delayed the fibrotic reaction and support its use as a potential biomaterial for mitigating fibrosis after MI. Delaying the progression of cardiac tissue remodeling may widen the therapeutic window for management of scarring after MI.
Collapse
Affiliation(s)
- Reshma S Nair
- Division of Experimental Pathology; Department of Biochemistry and Molecular Medicine, Université de Montréal and Montreal Heart Institute, Montréal, Québec, Canada
| | | | - Sachin J Shenoy
- Division of In Vivo Models and Testing, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Mukund A Prabhu
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India; Department of Cardiology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka
| | - Vikas Kumar
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India Current affiliations; Diabetes Research Program, Department of Medicine, New York University School of Medicine, New York
| | - Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India Current affiliations
| | | |
Collapse
|
69
|
Zhuo D, Lei I, Li W, Liu L, Li L, Ni J, Liu Z, Fan G. The origin, progress, and application of cell-based cardiac regeneration therapy. J Cell Physiol 2023; 238:1732-1755. [PMID: 37334836 DOI: 10.1002/jcp.31060] [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: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/21/2023]
Abstract
Cardiovascular disease (CVD) has become a severe threat to human health, with morbidity and mortality increasing yearly and gradually becoming younger. When the disease progresses to the middle and late stages, the loss of a large number of cardiomyocytes is irreparable to the body itself, and clinical drug therapy and mechanical support therapy cannot reverse the development of the disease. To explore the source of regenerated myocardium in model animals with the ability of heart regeneration through lineage tracing and other methods, and develop a new alternative therapy for CVDs, namely cell therapy. It directly compensates for cardiomyocyte proliferation through adult stem cell differentiation or cell reprogramming, which indirectly promotes cardiomyocyte proliferation through non-cardiomyocyte paracrine, to play a role in heart repair and regeneration. This review comprehensively summarizes the origin of newly generated cardiomyocytes, the research progress of cardiac regeneration based on cell therapy, the opportunity and development of cardiac regeneration in the context of bioengineering, and the clinical application of cell therapy in ischemic diseases.
Collapse
Affiliation(s)
- Danping Zhuo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjun Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Liu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihao Liu
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
70
|
Tang J, Jia X, Li Q, Cui Z, Liang A, Ke B, Yang D, Yao C. A DNA-based hydrogel for exosome separation and biomedical applications. Proc Natl Acad Sci U S A 2023; 120:e2303822120. [PMID: 37399419 PMCID: PMC10334772 DOI: 10.1073/pnas.2303822120] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Exosomes (EXOs) have been proven as biomarkers for disease diagnosis and agents for therapeutics. Great challenge remains in the separation of EXOs with high-purity and low-damage from complex biological media, which is critical for the downstream applications. Herein, we report a DNA-based hydrogel to realize the specific and nondestructive separation of EXOs from complex biological media. The separated EXOs were directly utilized in the detection of human breast cancer in clinical samples, as well as applied in the therapeutics of myocardial infarction in rat models. The materials chemistry basis of this strategy involved the synthesis of ultralong DNA chains via an enzymatic amplification, and the formation of DNA hydrogels through complementary base-pairing. These ultralong DNA chains that contained polyvalent aptamers were able to recognize and bind with the receptors on EXOs, and the specific and efficient binding ensured the selective separation of EXOs from media into the further formed networked DNA hydrogel. Based on this DNA hydrogel, rationally designed optical modules were introduced for the detection of exosomal pathogenic microRNA, which achieved the classification of breast cancer patients versus healthy donors with 100% precision. Furthermore, the DNA hydrogel that contained mesenchymal stem cell-derived EXOs was proved with significant therapeutic efficacy in repairing infarcted myocardium of rat models. We envision that this DNA hydrogel-based bioseparation system is promising as a powerful biotechnology, which will promote the development of extracellular vesicles in nanobiomedicine.
Collapse
Affiliation(s)
- Jianpu Tang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Xuemei Jia
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Qian Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Zhen Cui
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Aiqi Liang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Bin Ke
- Tianjin Medical University Cancer Institute and Hospital, Tianjin300060, P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| |
Collapse
|
71
|
Roshanravan N, Ghaffari S, Bastani S, Pahlavan S, Asghari S, Doustvandi MA, Jalilzadeh- Razin S, Dastouri M. Human cardiac organoids: A recent revolution in disease modeling and regenerative medicine. J Cardiovasc Thorac Res 2023; 15:68-72. [PMID: 37654821 PMCID: PMC10466470 DOI: 10.34172/jcvtr.2023.31830] [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: 04/04/2023] [Accepted: 06/10/2023] [Indexed: 09/02/2023] Open
Abstract
Three-dimensional (3D) myocardial tissues for studying human heart biology, physiology and pharmacology have recently received lots of attention. Organoids as 3D mini-organs are created from multiple cell types (i.e. induced pluripotent stem cells (iPSCs) or embryonic stem cells (ESCs)) with other supporting co-cultured cells such as endothelial cells or fibroblasts. Cardiac organoid culture technologies are bringing about significant advances in organ research and allows for the establishment of tissue regeneration and disease modeling. The present review provides an overview of the recent advances in human cardiac organoid platforms in disease biology and for cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Bastani
- Department of Immunology, Leiden University Medical Science, Leiden, Netherlands
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samira Asghari
- University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | | | - Sepideh Jalilzadeh- Razin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Dastouri
- Ankara University Biotechnology Institute and SISBIYOTEK Advanced Research Unit, Gumusdere Yerleskesi, Kecioren, Ankara, Turkey
| |
Collapse
|
72
|
Zhou Y, Liang Q, Wu X, Duan S, Ge C, Ye H, Lu J, Zhu R, Chen Y, Meng F, Yin L. siRNA Delivery against Myocardial Ischemia Reperfusion Injury Mediated by Reversibly Camouflaged Biomimetic Nanocomplexes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210691. [PMID: 36913720 DOI: 10.1002/adma.202210691] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/22/2023] [Indexed: 06/09/2023]
Abstract
siRNA-mediated management of myocardial ischemia reperfusion (IR) injury is greatly hampered by the inefficient myocardial enrichment and cardiomyocyte transfection. Herein, nanocomplexes (NCs) reversibly camouflaged with a platelet-macrophage hybrid membrane (HM) are developed to efficiently deliver Sav1 siRNA (siSav1) into cardiomyocytes, suppressing the Hippo pathway and inducing cardiomyocyte regeneration. The biomimetic BSPC@HM NCs consist of a cationic nanocore assembled from a membrane-penetrating helical polypeptide (P-Ben) and siSav1, a charge-reversal intermediate layer of poly(l-lysine)-cis-aconitic acid (PC), and an outer shell of HM. Due to HM-mediated inflammation homing and microthrombus targeting, intravenously injected BSPC@HM NCs can efficiently accumulate in the IR-injured myocardium, where the acidic inflammatory microenvironment triggers charge reversal of PC to shed off both HM and PC layers and allow the penetration of the exposed P-Ben/siSav1 NCs into cardiomyocytes. In rats and pigs, BSPC@HM NCs remarkably downregulates Sav1 in IR-injured myocardium, promotes myocardium regeneration, suppresses myocardial apoptosis, and recovers cardiac functions. This study reports a bioinspired strategy to overcome the multiple systemic barriers against myocardial siRNA delivery, and holds profound potential for gene therapy against cardiac injuries.
Collapse
Affiliation(s)
- Yang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qiujun Liang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Xuejie Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Shanzhou Duan
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Chenglong Ge
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Huan Ye
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Jianhui Lu
- Department of Vasculocardiology, Haimen Traditional Chinese Medicine Hospital, Haimen, 226100, China
| | - Rongying Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fenghua Meng
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
73
|
Aries A, Zanetti C, Hénon P, Drénou B, Lahlil R. Deciphering the Cardiovascular Potential of Human CD34 + Stem Cells. Int J Mol Sci 2023; 24:ijms24119551. [PMID: 37298503 DOI: 10.3390/ijms24119551] [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: 04/26/2023] [Revised: 05/17/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Ex vivo monitored human CD34+ stem cells (SCs) injected into myocardium scar tissue have shown real benefits for the recovery of patients with myocardial infarctions. They have been used previously in clinical trials with hopeful results and are expected to be promising for cardiac regenerative medicine following severe acute myocardial infarctions. However, some debates on their potential efficacy in cardiac regenerative therapies remain to be clarified. To elucidate the levels of CD34+ SC implication and contribution in cardiac regeneration, better identification of the main regulators, pathways, and genes involved in their potential cardiovascular differentiation and paracrine secretion needs to be determined. We first developed a protocol thought to commit human CD34+ SCs purified from cord blood toward an early cardiovascular lineage. Then, by using a microarray-based approach, we followed their gene expression during differentiation. We compared the transcriptome of undifferentiated CD34+ cells to those induced at two stages of differentiation (i.e., day three and day fourteen), with human cardiomyocyte progenitor cells (CMPCs), as well as cardiomyocytes as controls. Interestingly, in the treated cells, we observed an increase in the expressions of the main regulators usually present in cardiovascular cells. We identified cell surface markers of the cardiac mesoderm, such as kinase insert domain receptor (KDR) and the cardiogenic surface receptor Frizzled 4 (FZD4), induced in the differentiated cells in comparison to undifferentiated CD34+ cells. The Wnt and TGF-β pathways appeared to be involved in this activation. This study underlined the real capacity of effectively stimulated CD34+ SCs to express cardiac markers and, once induced, allowed the identification of markers that are known to be involved in vascular and early cardiogenesis, demonstrating their potential priming towards cardiovascular cells. These findings could complement their paracrine positive effects known in cell therapy for heart disease and may help improve the efficacy and safety of using ex vivo expanded CD34+ SCs.
Collapse
Affiliation(s)
- Anne Aries
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d'Altkirch, 68100 Mulhouse, France
| | - Céline Zanetti
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d'Altkirch, 68100 Mulhouse, France
| | | | - Bernard Drénou
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d'Altkirch, 68100 Mulhouse, France
- Groupe Hospitalier de la Région de Mulhouse Sud-Alsace, Hôpital E. Muller, 20 Avenue de Dr Laennec, 68100 Mulhouse, France
| | - Rachid Lahlil
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d'Altkirch, 68100 Mulhouse, France
| |
Collapse
|
74
|
Ketabat F, Maris T, Duan X, Yazdanpanah Z, Kelly ME, Badea I, Chen X. Optimization of 3D printing and in vitro characterization of alginate/gelatin lattice and angular scaffolds for potential cardiac tissue engineering. Front Bioeng Biotechnol 2023; 11:1161804. [PMID: 37304145 PMCID: PMC10248470 DOI: 10.3389/fbioe.2023.1161804] [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: 02/08/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Background: Engineering cardiac tissue that mimics the hierarchical structure of cardiac tissue remains challenging, raising the need for developing novel methods capable of creating structures with high complexity. Three-dimensional (3D)-printing techniques are among promising methods for engineering complex tissue constructs with high precision. By means of 3D printing, this study aims to develop cardiac constructs with a novel angular structure mimicking cardiac architecture from alginate (Alg) and gelatin (Gel) composite. The 3D-printing conditions were optimized and the structures were characterized in vitro, with human umbilical vein endothelial cells (HUVECs) and cardiomyocytes (H9c2 cells), for potential cardiac tissue engineering. Methods: We synthesized the composites of Alg and Gel with varying concentrations and examined their cytotoxicity with both H9c2 cells and HUVECs, as well as their printability for creating 3D structures of varying fibre orientations (angular design). The 3D-printed structures were characterized in terms of morphology by both scanning electron microscopy (SEM) and synchrotron radiation propagation-based imaging computed tomography (SR-PBI-CT), and elastic modulus, swelling percentage, and mass loss percentage as well. The cell viability studies were conducted via measuring the metabolic activity of the live cells with MTT assay and visualizing the cells with live/dead assay kit. Results: Among the examined composite groups of Alg and Gel, two combinations with ratios of 2 to 1 and 3 to 1 (termed as Alg2Gel1 and Alg3Gel1) showed the highest cell survival; they accordingly were used to fabricate two different structures: a novel angular and a conventional lattice structure. Scaffolds made of Alg3Gel1 showed higher elastic modulus, lower swelling percentage, less mass loss, and higher cell survival compared to that of Alg2Gel1. Although the viability of H9c2 cells and HUVECs on all scaffolds composed of Alg3Gel1 was above 99%, the group of the constructs with the angular design maintained significantly more viable cells compared to other investigated groups. Conclusion: The group of angular 3D-ptinted constructs has illustrated promising properties for cardiac tissue engineering by providing high cell viability for both endothelial and cardiac cells, high mechanical strength as well as appropriate swelling, and degradation properties during 21 days of incubation. Statement of Significance: 3D-printing is an emerging method to create complex constructs with high precision in a large scale. In this study, we have demonstrated that 3D-printing can be used to create compatible constructs from the composite of Alg and Gel with endothelial cells and cardiac cells. Also, we have demonstrated that these constructs are able to enhance the viability of cardiac and endothelial cells via creating a 3D structure mimicking the alignment and orientation of the fibers in the native heart.
Collapse
Affiliation(s)
- Farinaz Ketabat
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Titouan Maris
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Institut Catholique des arts et métiers (ICAM)- Site de Toulouse, Toulouse, France
| | - Xiaoman Duan
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zahra Yazdanpanah
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michael E. Kelly
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
75
|
Bhatti JS, Khullar N, Vijayvergiya R, Navik U, Bhatti GK, Reddy PH. Mitochondrial miRNA as epigenomic signatures: Visualizing aging-associated heart diseases through a new lens. Ageing Res Rev 2023; 86:101882. [PMID: 36780957 DOI: 10.1016/j.arr.2023.101882] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Aging bears many hard knocks, but heart disorders earn a particular allusion, being the most widespread. Cardiovascular diseases (CVDs) are becoming the biggest concern to mankind due to sundry health conditions directly or indirectly related to heart-linked abnormalities. Scientists know that mitochondria play a critical role in the pathophysiology of cardiac diseases. Both environment and genetics play an essential role in modulating and controlling mitochondrial functions. Even a minor abnormality may prove detrimental to heart function. Advanced age combined with an unhealthy lifestyle can cause most cardiomyocytes to be replaced by fibrotic tissue which upsets the conducting system and leads to arrhythmias. An aging heart encounters far more heart-associated comorbidities than a young heart. Many state-of-the-art technologies and procedures are already being used to prevent and treat heart attacks worldwide. However, it remains a mystery when this heart bomb would explode because it lacks an alarm. This calls for a novel and effective strategy for timely diagnosis and a sure-fire treatment. This review article provides a comprehensive overture of prospective potentials of mitochondrial miRNAs that predict complicated and interconnected pathways concerning heart ailments and signature compilations of relevant miRNAs as biomarkers to plot the role of miRNAs in epigenomics. This article suggests that analysis of DNA methylation patterns in age-associated heart diseases may determine age-impelled biomarkers of heart disease.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India.
| | - Rajesh Vijayvergiya
- Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Departments of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
76
|
Johnson J, Yang Y, Bian Z, Schena G, Li Y, Zhang X, Eaton DM, Gross P, Angheloiu A, Shaik A, Foster M, Berretta R, Kubo H, Mohsin S, Tian Y, Houser SR. Systemic Hypoxemia Induces Cardiomyocyte Hypertrophy and Right Ventricular Specific Induction of Proliferation. Circ Res 2023; 132:723-740. [PMID: 36799218 PMCID: PMC10023496 DOI: 10.1161/circresaha.122.321604] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND A recent study suggests that systemic hypoxemia in adult male mice can induce cardiac myocytes to proliferate. The goal of the present experiments was to confirm these results, provide new insights on the mechanisms that induce adult cardiomyocyte cell cycle reentry, and to determine if hypoxemia also induces cardiomyocyte proliferation in female mice. METHODS EdU-containing mini pumps were implanted in 3-month-old, male and female C57BL/6 mice. Mice were placed in a hypoxia chamber, and the oxygen was lowered by 1% every day for 14 days to reach 7% oxygen. The animals remained in 7% oxygen for 2 weeks before terminal studies. Myocyte proliferation was also studied with a mosaic analysis with double markers mouse model. RESULTS Hypoxia induced cardiac hypertrophy in both left ventricular (LV) and right ventricular (RV) myocytes, with LV myocytes lengthening and RV myocytes widening and lengthening. Hypoxia induced an increase (0.01±0.01% in normoxia to 0.11±0.09% in hypoxia) in the number of EdU+ RV cardiomyocytes, with no effect on LV myocytes in male C57BL/6 mice. Similar results were observed in female mice. Furthermore, in mosaic analysis with double markers mice, hypoxia induced a significant increase in RV myocyte proliferation (0.03±0.03% in normoxia to 0.32±0.15% in hypoxia of RFP+ myocytes), with no significant change in LV myocyte proliferation. RNA sequencing showed upregulation of mitotic cell cycle genes and a downregulation of Cullin genes, which promote the G1 to S phase transition in hypoxic mice. There was significant proliferation of nonmyocytes and mild cardiac fibrosis in hypoxic mice that did not disrupt cardiac function. Male and female mice exhibited similar gene expression following hypoxia. CONCLUSIONS Systemic hypoxia induces a global hypertrophic stress response that was associated with increased RV proliferation, and while LV myocytes did not show increased proliferation, our results minimally confirm previous reports that hypoxia can induce cardiomyocyte cell cycle activity in vivo.
Collapse
Affiliation(s)
- Jaslyn Johnson
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Yijun Yang
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Zilin Bian
- Tandon School of Engineering, New York University, Brooklyn, NY, USA
| | | | - Yijia Li
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xiaoying Zhang
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Deborah M. Eaton
- Penn Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Polina Gross
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | | | | | | | - Remus Berretta
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hajime Kubo
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Steven R. Houser
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
77
|
Fratta Pasini AM, Stranieri C, Busti F, Di Leo EG, Girelli D, Cominacini L. New Insights into the Role of Ferroptosis in Cardiovascular Diseases. Cells 2023; 12:cells12060867. [PMID: 36980208 PMCID: PMC10047059 DOI: 10.3390/cells12060867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the principal cause of disease burden and death worldwide. Ferroptosis is a new form of regulated cell death mainly characterized by altered iron metabolism, increased polyunsaturated fatty acid peroxidation by reactive oxygen species, depletion of glutathione and inactivation of glutathione peroxidase 4. Recently, a series of studies have indicated that ferroptosis is involved in the death of cardiac and vascular cells and has a key impact on the mechanisms leading to CVDs such as ischemic heart disease, ischemia/reperfusion injury, cardiomyopathies, and heart failure. In this article, we reviewed the molecular mechanism of ferroptosis and the current understanding of the pathophysiological role of ferroptosis in ischemic heart disease and in some cardiomyopathies. Moreover, the comprehension of the machinery governing ferroptosis in vascular cells and cardiomyocytes may provide new insights into preventive and therapeutic strategies in CVDs.
Collapse
|
78
|
Hosseinkhani H, Domb AJ, Sharifzadeh G, Nahum V. Gene Therapy for Regenerative Medicine. Pharmaceutics 2023; 15:856. [PMID: 36986717 PMCID: PMC10057434 DOI: 10.3390/pharmaceutics15030856] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The development of biological methods over the past decade has stimulated great interest in the possibility to regenerate human tissues. Advances in stem cell research, gene therapy, and tissue engineering have accelerated the technology in tissue and organ regeneration. However, despite significant progress in this area, there are still several technical issues that must be addressed, especially in the clinical use of gene therapy. The aims of gene therapy include utilising cells to produce a suitable protein, silencing over-producing proteins, and genetically modifying and repairing cell functions that may affect disease conditions. While most current gene therapy clinical trials are based on cell- and viral-mediated approaches, non-viral gene transfection agents are emerging as potentially safe and effective in the treatment of a wide variety of genetic and acquired diseases. Gene therapy based on viral vectors may induce pathogenicity and immunogenicity. Therefore, significant efforts are being invested in non-viral vectors to enhance their efficiency to a level comparable to the viral vector. Non-viral technologies consist of plasmid-based expression systems containing a gene encoding, a therapeutic protein, and synthetic gene delivery systems. One possible approach to enhance non-viral vector ability or to be an alternative to viral vectors would be to use tissue engineering technology for regenerative medicine therapy. This review provides a critical view of gene therapy with a major focus on the development of regenerative medicine technologies to control the in vivo location and function of administered genes.
Collapse
Affiliation(s)
- Hossein Hosseinkhani
- Innovation Center for Advanced Technology, Matrix, Inc., New York, NY 10019, USA
| | - Abraham J. Domb
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ghorbanali Sharifzadeh
- Department of Polymer Engineering, School of Chemical Engineering, Universiti Teknologi Malaysia, Skudai 81310, Johor, Malaysia
| | - Victoria Nahum
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
79
|
Wu T, Zhang X, Liu Y, Cui C, Sun Y, Liu W. Wet adhesive hydrogel cardiac patch loaded with anti-oxidative, autophagy-regulating molecule capsules and MSCs for restoring infarcted myocardium. Bioact Mater 2023; 21:20-31. [PMID: 36017068 PMCID: PMC9386397 DOI: 10.1016/j.bioactmat.2022.07.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Hydrogel patch-based stem cell transplantation and microenvironment-regulating drug delivery strategy is promising for the treatment of myocardial infarction (MI). However, the low retention of cells and drugs limits their therapeutic efficacies. Here, we propose a prefixed sponge carpet strategy, that is, aldehyde-dextran sponge (ODS) loading anti-oxidative/autophagy-regulating molecular capsules of 2-hydroxy-β-cyclodextrin@resveratrol (HP-β-CD@Res) is first bonded to the rat's heart via capillary removal of interfacial water from the tissue surface, and the subsequent Schiff base reaction between the aldehyde groups on ODS and amino groups on myocardium tissue. Then, an aqueous biocompatible hydrazided hyaluronic acid (HHA) solution encapsulating mesenchymal stem cells (MSCs) is impregnated into the anchored carpet to form HHA@ODS@HP-β-CD@Res hydrogel in situ via click reaction, thus prolonging the in vivo retention time of therapeutic drug and cells. Importantly, the HHA added to outer surface consumes the remaining aldehydes to contribute to nonsticky top surface, avoiding adhesion to other tissues. The embedded HP-β-CD@Res molecular capsules with antioxidant and autophagy regulation bioactivities can considerably improve cardiac microenvironment, reduce cardiomyocyte apoptosis, and enhance the survival of transplanted MSCs, thereby promoting cardiac repair by facilitating angiogenesis and reducing cardiac fibrosis. Developing a wet adhesive hydrogel cardiac patch through a prefixed sponge carpet strategy. 2-Hydroxy-β-cyclodextrin@resveratrol molecular capsules improve myocardial microenvironment. Post-adding of MSCs into the prefixed sponge carpet prolongs the in vivo retention time. The hydrogel patch loading molecular capsules and MSCs restores cardiac functions.
Collapse
|
80
|
Aballo TJ, Roberts DS, Bayne EF, Zhu W, Walcott G, Mahmoud AI, Zhang J, Ge Y. Integrated proteomics reveals alterations in sarcomere composition and developmental processes during postnatal swine heart development. J Mol Cell Cardiol 2023; 176:33-40. [PMID: 36657638 PMCID: PMC10006350 DOI: 10.1016/j.yjmcc.2023.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/20/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
The neonatal swine heart possesses an endogenous ability to regenerate injured myocardium through the proliferation of pre-existing cardiomyocyte (CM) populations. However, this regenerative capacity is lost shortly after birth. Normal postnatal developmental processes and the regenerative capacity of mammalian hearts are tightly linked, but not much is known about how the swine cardiac proteome changes throughout postnatal development. Herein, we integrated robust and quantitative targeted "top-down" and global "bottom-up" proteomic workflows to comprehensively define the dynamic landscape of the swine cardiac proteome throughout postnatal maturation. Using targeted top-down proteomics, we were able to identify significant alterations in sarcomere composition, providing new insight into the proteoform landscape of sarcomeres that can disassemble, a process necessary for productive CM proliferation. Furthermore, we quantified global changes in protein abundance using bottom-up proteomics, identified over 700 differentially expressed proteins throughout postnatal development, and mapped these proteins to changes in developmental and metabolic processes. We envision these results will help guide future investigations to comprehensively understand endogenous cardiac regeneration toward the development of novel therapeutic strategies for heart failure.
Collapse
Affiliation(s)
- Timothy J Aballo
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Elizabeth F Bayne
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Gregory Walcott
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
81
|
Gene Therapy for Cardiomyocyte Renewal: Cell Cycle, a Potential Therapeutic Target. Mol Diagn Ther 2023; 27:129-140. [PMID: 36512179 PMCID: PMC10123801 DOI: 10.1007/s40291-022-00625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
Heart disease is the primary cause of death worldwide. Even though extensive research has been done, and many pharmacological and surgical treatments have been introduced to treat heart disease, the mortality rate still remains high. Gene therapy is widely used to understand molecular mechanisms of myocardial infarction and to treat cardiomyocyte loss. It was reported that adult cardiomyocytes proliferate at a very low rate; thus, targeting their proliferation has become a new regenerative therapeutic approach. Currently, re-activating cardiomyocyte proliferation appears to be one of the most promising methods to promote adult cardiomyocyte renewal. In this article, we highlight gene therapeutic targets of cell proliferation presently being pursued to re-activate the cell cycle of cardiomyocytes, including cell cycle regulators, transcription factors, microRNAs, signal transduction, and other contributing factors. We also summarize gene delivery vectors that have been used in cardiac research and major challenges to be overcome in the translation to the clinical approach and future directions.
Collapse
|
82
|
Fang Z, Liu Z, Tao B, Jiang X. Engeletin mediates antiarrhythmic effects in mice with isoproterenol-induced cardiac remodeling. Biomed Pharmacother 2023; 161:114439. [PMID: 36848751 DOI: 10.1016/j.biopha.2023.114439] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
OBJECTIVE Engeletin is a potent natural compound with antioxidant and anti-inflammatory properties. However, its role in cardiac remodeling remains unclear. Herein, the aim of the present study was to explore the effects of engeletin on cardiac structural and electrical remodeling and its underlying mechanism. METHODS and results: A cardiac remodeling mice model using isoproterenol (ISO)-induced myocardial fibrosis was constructed and divided into the following four groups: control group; engeletin group; ISO group; engeletin + ISO group. Our results demonstrated that engeletin alleviated ISO-induced myocardial fibrosis and dysfunction. Moreover, engeletin significantly prolonged the QT and corrected QT (QTc) intervals, effective refractory period (ERP), and action potential duration (APD), and enhanced connexin protein 43 (Cx43) and ion channel expressions, thereby decreasing ventricular fibrillation (VF) susceptibility. Additionally, dihydroethidium staining illustrated that engeletin decreased reactive oxygen species (ROS) production. Of note, engeletin also increased the levels of superoxide dismutase and glutathione and decreased the activity of malondialdehyde and L-Glutathione oxidized. Moreover, engeletin significantly increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Furthermore, in vitro administration of an Nrf2 inhibitor abolished the anti-oxidant properties of engeletin. CONCLUSION Engeletin ameliorated cardiac structural and electrical remodeling, ion channel remodeling, and oxidative stress induced by ISO in mice, thereby reducing VF susceptibility. These effects may be attributed to the anti-oxidant properties of engeletin associated with the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Zhao Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Zhebo Liu
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Bo Tao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China.
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China.
| |
Collapse
|
83
|
Bao S, Lu Y, Zhang J, Xue L, Zhang Y, Wang P, Zhang F, Gu N, Sun J. Rapid improvement of heart repair in rats after myocardial infarction by precise magnetic stimulation on the vagus nerve with an injectable magnetic hydrogel. NANOSCALE 2023; 15:3532-3541. [PMID: 36723151 DOI: 10.1039/d2nr05073k] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The imbalance between the sympathetic and the parasympathetic nervous system is one of the main pathogeneses of myocardial infarction (MI). Vagus nerve stimulation (VNS), which restores autonomic nervous balance by enhancing the parasympathetic drive, is shown to have benefits for patients with MI. As a clinically safe and effective remote neuromodulation method, magnetic stimulation is expected to overcome the problems of infection and nerve injury caused by electrode implantation. However, it is difficult to achieve precise stimulation on a single vagus nerve due to the poor focus of the magnetic field. Here, we described a novel magnetic vagus nerve stimulation (mVNS) system, which consisted of an injectable chitosan/β-glycerophosphate (CS/GP) hydrogel loaded with superparamagnetic iron oxide (SPIO) nanoparticles and a mild magnetic pulse sequence. The injectable hydrogel prepared from clinically safe materials ensured minimally invasive implantation, and the SPIO nanoparticles in the hydrogel mediated the precise magnetic stimulation of a single vagus nerve. Under a mild magnetic field (∼100 mT), a decrease in heart rate and a change in vagus nerve potential were found in rats under in situ injection of a magnetic CS/GP hydrogel. Magnetic stimulation on the vagus nerve for 4 weeks (20 Hz, three times daily, 5 minutes each time) significantly improved the cardiac function and reduced the infarct size of the rats subjected to myocardial infarction, accompanied by suppression of inflammatory cell infiltration and inflammation factor expression. Taken together, these results demonstrated that the mVNS exhibited promising potential for treating myocardial infarction in the clinic.
Collapse
Affiliation(s)
- Siyuan Bao
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| | - Yao Lu
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou, 221009, P. R. China
| | - Jian Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
| | - Le Xue
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, P. R. China
| | - Peng Wang
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Fengxiang Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
| | - Ning Gu
- School of Medicine, Nanjing University, Nanjing, 210009, P. R. China.
| | - Jianfei Sun
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| |
Collapse
|
84
|
Maneechote C, Kerdphoo S, Jaiwongkam T, Chattipakorn SC, Chattipakorn N. Chronic Pharmacological Modulation of Mitochondrial Dynamics Alleviates Prediabetes-Induced Myocardial Ischemia-Reperfusion Injury by Preventing Mitochondrial Dysfunction and Programmed Apoptosis. Cardiovasc Drugs Ther 2023; 37:89-105. [PMID: 34515894 DOI: 10.1007/s10557-021-07250-7] [Citation(s) in RCA: 5] [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] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
PURPOSE There is an increasing body of evidence to show that impairment in mitochondrial dynamics including excessive fission and insufficient fusion has been observed in the pre-diabetic condition. In pre-diabetic rats with cardiac ischemia-reperfusion (I/R) injury, acute treatment with a mitochondria fission inhibitor (Mdivi-1) and a fusion promoter (M1) showed cardioprotection. However, the potential preventive effects of chronic Mdivi-1 and M1 treatment in a pre-diabetic model of cardiac I/R have never been elucidated. METHODS Male Wistar rats (n = 40) were fed with a high-fat diet (HFD) for 12 weeks to induce prediabetes. Then, all pre-diabetic rats received the following treatments daily via intraperitoneal injection for 2 weeks: (1) HFDV (Vehicle, 0.1% DMSO); (2) HFMdivi1 (Mdivi-1 1.2 mg/kg); (3) HFM1 (M1 2 mg/kg); and (4) HFCom (Mdivi-1 + M1). At the end of treatment protocols, all rats underwent 30 min of coronary artery ligation followed by reperfusion for 120 min. RESULTS Chronic Mdivi-1, M1, and the combined treatment showed markedly improved cardiac mitochondrial function and dynamic control, leading to a decrease in cardiac arrhythmias, myocardial cell death, and infarct size (49%, 42%, and 51% reduction for HFMdivi1, HFM1, and HFCom, respectively vs HFDV). All of these treatments improved cardiac function following cardiac I/R injury in pre-diabetic rats. CONCLUSION Chronic inhibition of mitochondrial fission and promotion of fusion exerted cardioprevention in prediabetes with cardiac I/R injury through the relief of cardiac mitochondrial dysfunction and dynamic alterations, and reduction in myocardial infarction, thus improving cardiac function.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
85
|
An injectable conductive hydrogel restores electrical transmission at myocardial infarct site to preserve cardiac function and enhance repair. Bioact Mater 2023; 20:339-354. [PMID: 35784639 PMCID: PMC9210214 DOI: 10.1016/j.bioactmat.2022.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 11/21/2022] Open
Abstract
Myocardial infarction (MI) leads to massive cardiomyocyte death and deposition of collagen fibers. This fibrous tissue disrupts electrical signaling in the myocardium, leading to cardiac systolic and diastolic dysfunction, as well as arrhythmias. Conductive hydrogels are a promising therapeutic strategy for MI. Here, we prepared a highly water-soluble conductive material (GP) by grafting polypyrrole (PPy) onto non-conductive gelatin. This component was added to the gel system formed by the Schiff base reaction between oxidized xanthan gum (OXG) and gelatin to construct an injectable conductive hydrogel. The prepared self-healing OGGP3 (3 wt% GP) hydrogel had good biocompatibility, elastic modulus, and electrical conductivity that matched the natural heart. The prepared biomaterials were injected into the rat myocardial scar tissue 2 days after MI. We found that the cardiac function of the rats treated with OGGP3 was improved, making it more difficult to induce arrhythmias. The electrical resistivity of myocardial fibrous tissue was reduced, and the conduction velocity of myocardial tissue was increased. Histological analysis showed reduced infarct size, increased left ventricular wall thickness, increased vessel density, and decreased inflammatory response in the infarcted area. Our findings clearly demonstrate that the OGGP3 hydrogel attenuates ventricular remodeling and inhibits infarct dilation, thus showing its potential for the treatment of MI. An injectable self-healing conductive hydrogel was synthesized for the treatment of myocardial infarction (MI). The OGGP3 hydrogel had elastic modulus (20.77 kPa) and conductivity (5.52 × 10−4 S/cm) that matched the natural heart. The hydrogel could protect cardiac function, reduce arrhythmia susceptibility and the resistivity of cardiac scar tissue. The hydrogel could increase left ventricular wall thickness, reduce infarct size and cardiac fibrosis in the infarcted area. The hydrogel could promote the expression level of cardiac-specific markers, induce angiogenesis, and reduce inflammation.
Collapse
|
86
|
Castillo MG, Peralta TM, Locatelli P, Velazquez C, Herrero Y, Crottogini AJ, Olea FD, Cuniberti LA. Promoting early neovascularization by allotransplanted adipose-derived Muse cells in an ovine model of acute myocardial infarction. PLoS One 2023; 18:e0277442. [PMID: 36662847 PMCID: PMC9858827 DOI: 10.1371/journal.pone.0277442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/26/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent preclinical studies have demonstrated that bone marrow (BM)-derived Muse cells have a homing mechanism to reach damaged cardiac tissue while also being able to reduce myocardial infarct size and improve cardiac function; however, the potential of BM-Muse cells to foster new blood-vessel formation has not been fully assessed. Up to date, adipose tissue (AT)-derived Muse cells remain to be studied in acute myocardial infarction (AMI). The aim of the present study was to analyze in vitro and in vivo the neovascularization capacity of AT-Muse cells while exploring their biodistribution and differentiation potential in a translational ovine model of AMI. METHODS AND RESULTS AT-Muse cells were successfully isolated from ovine adipose tissue. In adult sheep, one or more diagonal branches of the left anterior descending coronary artery were permanently ligated for thirty minutes. Sheep were randomized in two groups and treated with intramyocardial injections: Vehicle (PBS, n = 4) and AT-Muse (2x107 AT-Muse cells labeled with PKH26 Red Fluorescent Dye, n = 4). Molecular characterization showed higher expression of angiogenic genes (VEGF, PGF and ANG) and increased number of tube formation in AT-Muse cells group compared to Adipose-derived mesenchymal stromal cells (ASCs) group. At 7 days post-IAM, the AT-Muse group showed significantly more arterioles and capillaries than the Vehicle group. Co-localization of PKH26+ cells with desmin, sarcomeric actin and troponin T implied the differentiation of Muse cells to a cardiac fate; moreover, PKH26+ cells also co-localized with a lectin marker, suggesting a possible differentiation to a vascular lineage. CONCLUSION Intramyocardially administered AT-Muse cells displayed a significant neovascularization activity and survival capacity in an ovine model of AMI.
Collapse
Affiliation(s)
- Martha G. Castillo
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Tomás M. Peralta
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Yamila Herrero
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto J. Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda D. Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Luis A. Cuniberti
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
87
|
Ren Y, Wu Y, He W, Tian Y, Zhao X. Exosomes secreted from bone marrow mesenchymal stem cells suppress cardiomyocyte hypertrophy through Hippo-YAP pathway in heart failure. Genet Mol Biol 2023; 46:e20220221. [PMID: 36929834 PMCID: PMC10021068 DOI: 10.1590/1678-4685-gmb-2022-0221] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/23/2022] [Indexed: 03/16/2023] Open
Abstract
Mesenchymal stem cells-derived exosomes (MSCs-exosomes) reportedly possess cardioprotective effects. This study investigated the therapeutic potential and mechanisms of MSCs-exosomes on heart failure (HF). H9c2 cells were used to establish a cardiomyocyte hypertrophy model by angiotensin II (Ang II) treatment. Isolated MSCs-exosomes were identified by transmission electron microscope and CD63 detection. Apoptosis rate was measured by terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) assay. Levels of inflammatory factors [interleukin (IL)-1β, IL-4, IL-6, and tumor necrosis factor (TNF)-α] and brain natriuretic peptide (BNP) were determined by ELISA. Expression of apoptosis-related proteins [Bax, B-cell lymphoma-2 (Bcl-2), and caspase 3] and Hippo-Yes-associated protein (YAP) pathway-related proteins [YAP, phosphor (p)-YAP, and tafazzin (TAZ)] was detected by western blotting. Cardiomyocyte hypertrophy of H9c2 cells induced by Ang II was ameliorated by MSCs-exosomes treatment. MSCs-exosomes downregulated Bax and caspase 3 levels and upregulated Bcl-2 level in Ang II-induced H9c2 cells. MSCs-exosomes also reduced the levels of BNP, IL-1β, IL-4, IL-6, and TNF-α in Ang II-induced H9c2 cells. Meanwhile, p-YAP was downregulated and TAZ was upregulated after MSCs-exosomes administration. In conclusion, MSCs-exosomes alleviate the apoptosis and inflammatory response of cardiomyocyte via deactivating Hippo-YAP pathway in HF.
Collapse
Affiliation(s)
- Yu Ren
- Inner Mongolia People's Hospital, Department of Scientific Research, Hohhot, China
| | - Yun Wu
- Inner Mongolia People's Hospital, Department of Cardiology, Hohhot, China.,Inner Mongolia People's Hospital, Clinical Medical Research Center in Cardiovascular Diseases, Hohhot, China
| | - Wenshuai He
- Inner Mongolia People's Hospital, Department of Cardiology, Hohhot, China.,Inner Mongolia People's Hospital, Clinical Medical Research Center in Cardiovascular Diseases, Hohhot, China
| | - Yingjie Tian
- Inner Mongolia People's Hospital, Department of Cardiology, Hohhot, China.,Inner Mongolia People's Hospital, Clinical Medical Research Center in Cardiovascular Diseases, Hohhot, China
| | - Xingsheng Zhao
- Inner Mongolia People's Hospital, Department of Cardiology, Hohhot, China.,Inner Mongolia People's Hospital, Clinical Medical Research Center in Cardiovascular Diseases, Hohhot, China
| |
Collapse
|
88
|
Wu Z, Li W, Cheng S, Liu J, Wang S. Novel fabrication of bioengineered injectable chitosan hydrogel loaded with conductive nanoparticles to improve therapeutic potential of mesenchymal stem cells in functional recovery after ischemic myocardial infarction. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102616. [PMID: 36374915 DOI: 10.1016/j.nano.2022.102616] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/19/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
In recent decades, myocardial regeneration through stem cell transplantation and tissue engineering has been viewed as a promising technique for treating myocardial infarction. As a result, the researcher attempts to see whether co-culturing modified mesenchymal stem cells with Au@Ch-SF macro-hydrogel and H9C2 may help with tissue regeneration and cardiac function recovery. The gold nanoparticles (Au) incorporated into the chitosan-silk fibroin hydrogel (Au@Ch-SF) were validated using spectral and microscopic examinations. The most essential elements of hydrogel groups were investigated in detail, including weight loss, mechanical strength, and drug release rate. Initially, the cardioblast cells (H9C2 cells) was incubated with Au@Ch-SF macro-hydrogel, followed by mesenchymal stem cells (2 × 105) were transplanted into the Au@Ch-SF macro-hydrogel+H9C2 culture at the ratio of 2:1. Further, cardiac phenotype development, cytokines expression and tissue regenerative performance of modified mesenchymal stem cells treatment were studied through various in vitro and in vivo analyses. The Au@Ch-SF macro-hydrogel gelation time was much faster than that of Ch and Ch-SF hydrogels, showing that Ch and SF exhibited greater intermolecular interactions. The obtained Au@Ch-SF macro-hydrogel has no toxicity on mesenchymal stem cells (MS) or cardiac myoblast (H9C2) cells, according to the biocompatibility investigation. MS cells co-cultured with Au@Ch-SF macro-hydrogel and H9C2 cells also stimulated cardiomyocyte fiber restoration, which has been confirmed in myocardial infarction rats using -MHC and Cx43 myocardial indicators. We developed a novel method of co-cultured therapy using MS cells, Au@Ch-SF macro-hydrogel, and H9C2 cells which could promote the regenerative activities in myocardial ischemia cells. These study findings show that co-cultured MS therapy might be effective for the treatment of myocardial injury.
Collapse
Affiliation(s)
- Zheng Wu
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, PR China; Department of 28 Division of Cardiovascular, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Wenzheng Li
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, PR China; Department of 28 Division of Cardiovascular, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Shujuan Cheng
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, PR China; Department of 28 Division of Cardiovascular, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Jinghua Liu
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, PR China; Department of 28 Division of Cardiovascular, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China.
| | - Shaoping Wang
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, PR China; Department of 28 Division of Cardiovascular, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| |
Collapse
|
89
|
Wang X, Zhou H, Liu Q, Cheng P, Zhao T, Yang T, Zhao Y, Sha W, Zhao Y, Qu H. Targeting regulatory T cells for cardiovascular diseases. Front Immunol 2023; 14:1126761. [PMID: 36911741 PMCID: PMC9995594 DOI: 10.3389/fimmu.2023.1126761] [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: 12/18/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death and disability worldwide. The CVDs are accompanied by inflammatory progression, resulting in innate and adaptive immune responses. Regulatory T cells (Tregs) have an immunosuppressive function and are one of the subsets of CD4+T cells that play a crucial role in inflammatory diseases. Whether using Tregs as a biomarker for CVDs or targeting Tregs to exert cardioprotective functions by regulating immune balance, suppressing inflammation, suppressing cardiac and vascular remodeling, mediating immune tolerance, and promoting cardiac regeneration in the treatment of CVDs has become an emerging research focus. However, Tregs have plasticity, and this plastic Tregs lose immunosuppressive function and produce toxic effects on target organs in some diseases. This review aims to provide an overview of Tregs' role and related mechanisms in CVDs, and reports on the research of plasticity Tregs in CVDs, to lay a foundation for further studies targeting Tregs in the prevention and treatment of CVDs.
Collapse
Affiliation(s)
- Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingyao Zhao
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanjing Sha
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanyan Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
90
|
Mu L, Dong R, Guo B. Biomaterials-Based Cell Therapy for Myocardial Tissue Regeneration. Adv Healthc Mater 2022; 12:e2202699. [PMID: 36572412 DOI: 10.1002/adhm.202202699] [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: 10/19/2022] [Revised: 12/11/2022] [Indexed: 12/28/2022]
Abstract
Cardiovascular diseases (CVDs) have been the leading cause of death worldwide during the past several decades. Cell loss is the main problem that results in cardiac dysfunction and further mortality. Cell therapy aiming to replenish the lost cells is proposed to treat CVDs especially ischemic heart diseases which lead to a big portion of cell loss. Due to the direct injection's low cell retention and survival ratio, cell therapy using biomaterials as cell carriers has attracted more and more attention because of their promotion of cell delivery and maintenance at the aiming sites. In this review, the three main factors involved in cell therapy for myocardial tissue regeneration: cell sources (somatic cells, stem cells, and engineered cells), chemical components of cell carriers (natural materials, synthetic materials, and electroactive materials), and categories of cell delivery materials (patches, microspheres, injectable hydrogels, nanofiber and microneedles, etc.) are systematically summarized. An introduction of the methods including magnetic resonance/radionuclide/photoacoustic and fluorescence imaging for tracking the behavior of transplanted cells in vivo is also included. Current challenges of biomaterials-based cell therapy and their future directions are provided to give both beginners and professionals a clear view of the development and future trends in this area.
Collapse
Affiliation(s)
- Lei Mu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ruonan Dong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Baolin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China.,State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
91
|
Koo KM, Kim CD, Ju FN, Kim H, Kim CH, Kim TH. Recent Advances in Electrochemical Biosensors for Monitoring Animal Cell Function and Viability. BIOSENSORS 2022; 12:bios12121162. [PMID: 36551129 PMCID: PMC9775431 DOI: 10.3390/bios12121162] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 05/28/2023]
Abstract
Redox reactions in live cells are generated by involving various redox biomolecules for maintaining cell viability and functions. These qualities have been exploited in the development of clinical monitoring, diagnostic approaches, and numerous types of biosensors. Particularly, electrochemical biosensor-based live-cell detection technologies, such as electric cell-substrate impedance (ECIS), field-effect transistors (FETs), and potentiometric-based biosensors, are used for the electrochemical-based sensing of extracellular changes, genetic alterations, and redox reactions. In addition to the electrochemical biosensors for live-cell detection, cancer and stem cells may be immobilized on an electrode surface and evaluated electrochemically. Various nanomaterials and cell-friendly ligands are used to enhance the sensitivity of electrochemical biosensors. Here, we discuss recent advances in the use of electrochemical sensors for determining cell viability and function, which are essential for the practical application of these sensors as tools for pharmaceutical analysis and toxicity testing. We believe that this review will motivate researchers to enhance their efforts devoted to accelerating the development of electrochemical biosensors for future applications in the pharmaceutical industry and stem cell therapeutics.
Collapse
|
92
|
Kędziora A, Konstanty-Kalandyk J, Litwinowicz R, Mazur P, Kapelak B, Piątek J. Hybrid techniques for myocardial regeneration: state of the art and future perspectives. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2022; 18:360-365. [PMID: 36967853 PMCID: PMC10031663 DOI: 10.5114/aic.2022.121124] [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: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Myocardium has a limited proliferative capacity, and adult hearts are considered incapable of regenerating after injury. A significant loss in the viable myocardium eventually diminishes the heart's ability to contract synchronously, leading to heart failure. Despite the development in interventional and pharmacological treatment for ischemic heart disease and heart failure, there is a significant number of highly symptomatic patients. For these individuals, treatments that stimulate myocardial regeneration can offer alleviation of dyspnea and angina and improvement in quality of life. Stem cells are known to promote neovascularization and endothelial repair. Various stem cell lines have been investigated over the years to establish those with the highest potential to differentiate into cardiomyocytes, including bone marrow-derived mononuclear cells, mesenchymal stromal cells, CD34+, CD133+, endothelial progenitor cells, and adipose-derived mesenchymal stromal cells. Stem cell studies were based on several delivery pathways: infusion into coronary vessels, direct injection into the injured region of the myocardium, and delivery within the novel bioengineered scaffolds. Acellular materials have also been investigated over the years. They demonstrate the therapeutic potential to promote angiogenesis and release of growth factors to improve the restoration of critical components of the extracellular matrix. This review summarizes hybrid cardiac regeneration treatments that combine novel bioengineering techniques with delivery approaches that cardiac surgeons can provide.
Collapse
Affiliation(s)
- Anna Kędziora
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Janusz Konstanty-Kalandyk
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Radosław Litwinowicz
- Jagiellonian University Medical College, Krakow, Poland
- Department of Cardiac Surgery, Regional Specialist Hospital, Grudziadz, Poland
| | - Piotr Mazur
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
- Department of Cardiac Surgery, Leipzig Heart Center, Leipzig, Germany
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester MIN, USA
| | - Bogusław Kapelak
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Piątek
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
93
|
Quan Z, Zhang X, Song X, Chen P, Wu Q. The use of intraoperative transit time flow measurement can reduce postoperative myocardial injury. J Card Surg 2022; 37:4246-4253. [PMID: 35998272 DOI: 10.1111/jocs.16818] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/02/2022] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This study investigates the relationship between the use of transit time flow measurement (TTFM) and postoperative myocardial injury in off-pump coronary artery bypass grafting (OPCABG). METHODS In this retrospective study, we collected basic data from patients hospitalized for OPCABG in the Department of Cardiothoracic Surgery, Changzhou Second People's Hospital Affiliated with Nanjing Medical University. According to the academic research consortium (ARC)-2 definition of significant myocardial injury, we used cardiac troponin I >2380 ng/L as a criterion for significant postoperative myocardial injury. We use logistic regression and forest plots to assess the association of TTFM use with myocardial injury outcomes. RESULTS One hundred and forty-six patients were included in this study. The overall median age of these patients was 65.05 years, and 32 (21.92%) experienced a postoperative myocardial injury. TTFM was independently associated with the incidence of postoperative myocardial injury (odds ratio = 0.34 [95% confidence interval = 0.15-0.78]; p = .01), and we found similar trends in regression analyses across subgroups of sex, age, number of bridging vessels, hypertension, diabetes mellitus, BMI, and percutaneous coronary revascularization. One hundred and six patients were followed for 1-year vessel permeability, and seven patients (6.6%) were occluded. CONCLUSIONS The use of TTFM is independently associated with a reduced incidence of postoperative myocardial lesions during off-pump coronary bypass surgery. The TTFM procedure in OPCABG deserves to be actively promoted to reduce the incidence of postoperative myocardial injury.
Collapse
Affiliation(s)
- Zheng Quan
- Department of Thoracic and Cardiovascular Surgery, Affiliated with Nanjing Medical University, Changzhou, Jiangsu, China.,Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyu Zhang
- Heart Center, Northwest Women and Children's Hospital, Xi'an, Shaanxi, China
| | - Xueyu Song
- Department of Thoracic and Cardiovascular Surgery, Affiliated with Nanjing Medical University, Changzhou, Jiangsu, China.,Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Pengyu Chen
- Department of Thoracic and Cardiovascular Surgery, Affiliated with Nanjing Medical University, Changzhou, Jiangsu, China.,Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Qiyong Wu
- Department of Thoracic and Cardiovascular Surgery, Affiliated with Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
94
|
Lozano J, Rai A, Lees JG, Fang H, Claridge B, Lim SY, Greening DW. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int J Mol Sci 2022; 23:14334. [PMID: 36430812 PMCID: PMC9696585 DOI: 10.3390/ijms232214334] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) from stem cells have shown significant therapeutic potential to repair injured cardiac tissues and regulate pathological fibrosis. However, scalable generation of stem cells and derived EVs for clinical utility remains a huge technical challenge. Here, we report a rapid size-based extrusion strategy to generate EV-like membranous nanovesicles (NVs) from easily sourced human iPSCs in large quantities (yield 900× natural EVs). NVs isolated using density-gradient separation (buoyant density 1.13 g/mL) are spherical in shape and morphologically intact and readily internalised by human cardiomyocytes, primary cardiac fibroblasts, and endothelial cells. NVs captured the dynamic proteome of parental cells and include pluripotency markers (LIN28A, OCT4) and regulators of cardiac repair processes, including tissue repair (GJA1, HSP20/27/70, HMGB1), wound healing (FLNA, MYH9, ACTC1, ILK), stress response/translation initiation (eIF2S1/S2/S3/B4), hypoxia response (HMOX2, HSP90, GNB1), and extracellular matrix organization (ITGA6, MFGE8, ITGB1). Functionally, NVs significantly promoted tubule formation of endothelial cells (angiogenesis) (p < 0.05) and survival of cardiomyocytes exposed to low oxygen conditions (hypoxia) (p < 0.0001), as well as attenuated TGF-β mediated activation of cardiac fibroblasts (p < 0.0001). Quantitative proteome profiling of target cell proteome following NV treatments revealed upregulation of angiogenic proteins (MFGE8, MYH10, VDAC2) in endothelial cells and pro-survival proteins (CNN2, THBS1, IGF2R) in cardiomyocytes. In contrast, NVs attenuated TGF-β-driven extracellular matrix remodelling capacity in cardiac fibroblasts (ACTN1, COL1A1/2/4A2/12A1, ITGA1/11, THBS1). This study presents a scalable approach to generating functional NVs for cardiac repair.
Collapse
Affiliation(s)
- Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jarmon G. Lees
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Bethany Claridge
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Shiang Y. Lim
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
95
|
Sweed E, Sweed D, Galal N, Abd-Elhafiz HI. Dapagliflozin Protection against Myocardial Ischemia by Modulating Sodium-glucose Transporter 2 Inhibitor, Silent Information Regulator 1, and Fatty Acid Synthase Expressions. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.10861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND: The emerging role of sodium-glucose transporter 2 (SGLT2) inhibitors drugs as potential therapeutic agents in myocardial ischemic (MI) injury treatment has raised the concern for possible mechanisms of action.
AIM: The current experimental study aimed to investigate the possible protective effects of dapagliflozin (DAPA) a SGLT2i, on isoproterenol (ISO)-induced MI in rats.
MATERIALS AND METHODS: Thirty Wistar rats were divided randomly and equally into three groups. Group 1 (control group): Received 1.0 mL of normal saline through an orogastric tube for 14 days. Group 2 (ISO group): Received 1.0 mL of normal saline orally through an orogastric tube for 14 days. In the last 2 days (days 13 and 14), ISO (100 mg/kg) was freshly dissolved in normal saline and injected subcutaneously once daily. Group 3 (ISO + DAPA-treated group): Received DAPA 1.0 mg/kg/day orally for 14 days. In the last 2 days (days 13 and 14), ISO (100 mg/kg) was introduced like that described in Group 2.
RESULTS: DAPA protects MI development by reversal of blood pressure changes, electrocardiographic alterations, stabilization of cardiac enzymes, inflammation restoration, oxidative stress, and lipid profile. SGLT2 was overexpressed in the ISO-induced MI, which declined in the ISO + DAPA group. Moreover, DAPA induced silent information regulator 1 (SIRT1)/fatty acid synthase (FASN) overexpression in ISO-induced MI. DAPA could have a potential protective role against acute MI.
CONCLUSION: DAPA protects against acute MI by modulating SIRT1 and FASN expression in cardiac muscles, suppressing oxidative stress, and downregulating inflammatory mediators.
Collapse
|
96
|
Hu Y, Lu H, Li H, Ge J. Molecular basis and clinical implications of HIFs in cardiovascular diseases. Trends Mol Med 2022; 28:916-938. [PMID: 36208988 DOI: 10.1016/j.molmed.2022.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022]
Abstract
Oxygen maintains the homeostasis of an organism in a delicate balance in different tissues and organs. Under hypoxic conditions, hypoxia-inducible factors (HIFs) are specific and dominant factors in the spatiotemporal regulation of oxygen homeostasis. As the most basic functional unit of the heart at the cellular level, the cardiomyocyte relies on oxygen and nutrients delivered by the microvasculature to keep the heart functioning properly. Under hypoxic stress, HIFs are involved in acute and chronic myocardial pathology because of their spatiotemporal specificity, thus granting them therapeutic potential. Most adult animals lack the ability to regenerate their myocardium entirely following injury, and complete regeneration has long been a goal of clinical treatment for heart failure. The precise manipulation of HIFs (considering their dynamic balance and transformation) and the development of HIF-targeted drugs is therefore an extremely attractive cardioprotective therapy for protecting against myocardial ischemic and hypoxic injury, avoiding myocardial remodeling and heart failure, and promoting recovery of cardiac function.
Collapse
Affiliation(s)
- Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hua Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
97
|
Zadory M, Lopez E, Babity S, Gravel SP, Brambilla D. Current knowledge on the tissue distribution of mRNA nanocarriers for therapeutic protein expression. Biomater Sci 2022; 10:6077-6115. [PMID: 36097955 DOI: 10.1039/d2bm00859a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exogenously delivered mRNA-based drugs are emerging as a new class of therapeutics with the potential to treat several diseases. Over the last decade, advancements in the design of non-viral delivery tools have enabled mRNA to be evaluated for several therapeutic purposes including protein replacement therapies, gene editing, and vaccines. However, in vivo delivery of mRNA to targeted organs and cells remains a critical challenge. Evaluation of the biodistribution of mRNA vehicles is of utmost importance for the development of effective pharmaceutical candidates. In this review, we discuss the recent advances in the design of nanoparticles loaded with mRNA and extrapolate the key factors influencing their biodistribution following administration. Finally, we highlight the latest developments in the preclinical and clinical translation of mRNA therapeutics for protein supplementation therapy.
Collapse
Affiliation(s)
- Matthias Zadory
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Elliot Lopez
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Samuel Babity
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Simon-Pierre Gravel
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| |
Collapse
|
98
|
Man W, Song X, Xiong Z, Gu J, Lin J, Gu X, Yu D, Li C, Jiang M, Zhang X, Yang Z, Cao Y, Zhang Y, Shu X, Wu D, Wang H, Ji G, Sun D. Exosomes derived from pericardial adipose tissues attenuate cardiac remodeling following myocardial infarction by Adipsin-regulated iron homeostasis. Front Cardiovasc Med 2022; 9:1003282. [PMID: 36172581 PMCID: PMC9510661 DOI: 10.3389/fcvm.2022.1003282] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
As a vital adipokine, Adipsin is closely associated with cardiovascular risks. Nevertheless, its role in the onset and development of cardiovascular diseases remains elusive. This study was designed to examine the effect of Adipsin on survival, cardiac dysfunction and adverse remodeling in the face of myocardial infarction (MI) injury. In vitro experiments were conducted to evaluate the effects of Adipsin on cardiomyocyte function in the face of hypoxic challenge and the mechanisms involved. Our results showed that Adipsin dramatically altered expression of proteins associated with iron metabolism and ferroptosis. In vivo results demonstrated that Adipsin upregulated levels of Ferritin Heavy Chain (FTH) while downregulating that of Transferrin Receptor (TFRC) in peri-infarct regions 1 month following MI. Adipsin also relieved post-MI-associated lipid oxidative stress as evidenced by decreased expression of COX2 and increased GPX4 level. Co-immunoprecipitation and immunofluorescence imaging prove a direct interaction between Adipsin and IRP2. As expected, cardioprotection provided by Adipsin depends on the key molecule of IRP2. These findings revealed that Adipsin could be efficiently delivered to the heart by exosomes derived from pericardial adipose tissues. In addition, Adipsin interacted with IRP2 to protect cardiomyocytes against ferroptosis and maintain iron homeostasis. Therefore, Adipsin-overexpressed exosomes derived from pericardial adipose tissues may be a promising therapeutic strategy to prevent adverse cardiac remodeling following ischemic heart injury.
Collapse
Affiliation(s)
- Wanrong Man
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinglong Song
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Gu
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Duan Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Haichang Wang
- Heart Hospital, Xi’an International Medical Center, Xi’an, China
- Haichang Wang,
| | - Gang Ji
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Gang Ji,
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Dongdong Sun,
| |
Collapse
|
99
|
Pioglitazone Mediates Cardiac Progenitor Formation through Increasing ROS Levels. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1480345. [PMID: 36124070 PMCID: PMC9482506 DOI: 10.1155/2022/1480345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/06/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
In order to achieve a sufficient population of cardiac-committed progenitor cells, it is crucial to know the mechanisms of cardiac progenitor formation. Previous studies suggested ROS effect on cardiac commitment events to play a key role in the cell signaling and activate cardiac differentiation of pluripotent stem cells. We previously reported that PPARγ activity is essential for cardiac progenitor cell commitment. Although several studies have conducted the involvement of PPARγ-related signaling pathways in cardiac differentiation, so far, the regulatory mechanisms of these signaling pathways have not been discussed and cleared. In this study, we focus on the role of PPARγ agonist in ROS generation and its further effects on the differentiation of cardiac cells from mESCs. The results of this study show that the presence of ROS is necessary for heart differentiation in the precursor stage of cardiac cells, and the coenzyme Q10 antioxidant precludes proper cardiac differentiation. In addition, this antioxidant prevents the action of pioglitazone in increasing oxygen radicals as well as beating cardiomyocyte differentiation properties. In this case, it can be concluded that PPARγ is required to modulate ROS levels during cardiac differentiation.
Collapse
|
100
|
Born LI, Andree T, Frank S, Hübner J, Link S, Langheine M, Charlet A, Esser JS, Brehm R, Moser M. eif4ebp3l-A New Affector of Zebrafish Angiogenesis and Heart Regeneration? Int J Mol Sci 2022; 23:ijms231710075. [PMID: 36077472 PMCID: PMC9456460 DOI: 10.3390/ijms231710075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
The eukaryotic initiation factor 4E binding protein (4E-BP) family is involved in translational control of cell proliferation and pro-angiogenic factors. The zebrafish eukaryotic initiation factor 4E binding protein 3 like (eif4ebp3l) is a member of the 4E-BPs and responsible for activity-dependent myofibrillogenesis, but whether it affects cardiomyocyte (CM) proliferation or heart regeneration is unclear. We examined eif4ebp3l during zebrafish vascular development and heart regeneration post cryoinjury in adult zebrafish. Using morpholino injections we induced silencing of eif4ebp3l in zebrafish embryos, which led to increased angiogenesis at 94 h post fertilization (hpf). For investigation of eif4ebp3l in cardiac regeneration, zebrafish hearts were subjected to cryoinjury. Regenerating hearts were analyzed at different time points post-cryoinjury for expression of eif4ebp3l by in situ hybridization and showed strongly decreased eif4ebp3l expression in the injured area. We established a transgenic zebrafish strain, which overexpressed eif4ebp3l under the control of a heat-shock dependent promotor. Overexpression of eif4ebp3l during zebrafish heart regeneration caused only macroscopically a reduced amount of fibrin at the site of injury. Overall, these findings demonstrate that silencing of eif4ebp3l has pro-angiogenic properties in zebrafish vascular development and when eif4ebp3l is overexpressed, fibrin deposition tends to be altered in zebrafish cardiac regeneration after cryoinjury.
Collapse
Affiliation(s)
- Lisa I. Born
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute of Anatomy, University of Veterinary Medicine of Hannover, Foundation, 30173 Hannover, Germany
- Correspondence:
| | - Theresa Andree
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Svenja Frank
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Judith Hübner
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sandra Link
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Marion Langheine
- Institute of Anatomy, University of Veterinary Medicine of Hannover, Foundation, 30173 Hannover, Germany
| | - Anne Charlet
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jennifer S. Esser
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Ralph Brehm
- Institute of Anatomy, University of Veterinary Medicine of Hannover, Foundation, 30173 Hannover, Germany
| | - Martin Moser
- Department of Cardiology and Angiology, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|