1
|
Long M, Cheng M. Small extracellular vesicles associated miRNA in myocardial fibrosis. Biochem Biophys Res Commun 2024; 727:150336. [PMID: 38959731 DOI: 10.1016/j.bbrc.2024.150336] [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: 04/16/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
Myocardial fibrosis involves the loss of cardiomyocytes, myocardial fibroblast proliferation, and a reduction in angiogenesis, ultimately leading to heart failure, Given its significant implications, it is crucial to explore novel therapies for myocardial fibrosis. Recently one emerging avenue has been the use of small extracellular vesicles (sEV)-carried miRNA. In this review, we summarize the regulatory role of sEV-carried miRNA in myocardial fibrosis. We explored not only the potential diagnostic value of circulating miRNA as biomarkers for heart disease but also the therapeutic implications of sEV-carried miRNA derived from various cellular sources and applications of modified sEV. This exploration is paramount for researchers striving to develop innovative, cell-free therapies as potential drug candidates for the management of myocardial fibrosis.
Collapse
Affiliation(s)
- Minwen Long
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Kufazvinei TTJ, Chai J, Boden KA, Channon KM, Choudhury RP. Emerging opportunities to target inflammation: myocardial infarction and type 2 diabetes. Cardiovasc Res 2024; 120:1241-1252. [PMID: 39027945 DOI: 10.1093/cvr/cvae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/05/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
After myocardial infarction (MI), patients with type 2 diabetes have an increased rate of adverse outcomes, compared to patients without. Diabetes confers a 1.5-2-fold increase in early mortality and, importantly, this discrepancy has been consistent over recent decades, despite advances in treatment and overall survival. Certain assumptions have emerged to explain this increased risk, such as differences in infarct size or coronary artery disease severity. Here, we re-evaluate that evidence and show how contemporary analyses using state-of-the-art characterization tools suggest that the received wisdom tells an incomplete story. Simultaneously, epidemiological and mechanistic biological data suggest additional factors relating to processes of diabetes-related inflammation might play a prominent role. Inflammatory processes after MI mediate injury and repair and are thus a potential therapeutic target. Recent studies have shown how diabetes affects immune cell numbers and drives changes in the bone marrow, leading to pro-inflammatory gene expression and functional suppression of healing and repair. Here, we review and re-evaluate the evidence around adverse prognosis in patients with diabetes after MI, with emphasis on how targeting processes of inflammation presents unexplored, yet valuable opportunities to improve cardiovascular outcomes in this vulnerable patient group.
Collapse
Affiliation(s)
- Tafadzwa T J Kufazvinei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Jason Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Katherine A Boden
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
3
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
4
|
Cui Z, Zhang L, Hu G, Zhang F. Extracellular Vesicles in Cardiovascular Pathophysiology: Communications, Biomarkers, and Therapeutic Potential. Cardiovasc Toxicol 2024; 24:711-726. [PMID: 38844744 DOI: 10.1007/s12012-024-09875-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/25/2024] [Indexed: 08/07/2024]
Abstract
Extracellular vesicles (EVs) are diverse, membrane-bound vesicles released from cells into the extracellular environment. They originate from either endosomes or the cell membrane and typically include exosomes and microvesicles. These EVs serve as crucial mediators of intercellular communication, carrying a variety of contents such as nucleic acids, proteins, and lipids, which regulate the physiological and pathological processes of target cells. Moreover, the molecular cargo of EVs can reflect critical information about the originating cells, making them potential biomarkers for the diagnosis and prognosis of diseases. Over the past decade, the role of EVs as key communicators between cell types in cardiovascular physiology and pathology has gained increasing recognition. EVs from different cellular sources, or from the same source under different cellular conditions, can have distinct impacts on the management, diagnosis, and prognosis of cardiovascular diseases. Furthermore, it is essential to consider the influence of cardiovascular-derived EVs on the metabolism of peripheral organs. This review aims to summarize recent advancements in the field of cardiovascular research with respect to the roles and implications of EVs. Our goal is to provide new insights and directions for the early prevention and treatment of cardiovascular diseases, with an emphasis on the therapeutic potential and diagnostic value of EVs.
Collapse
Affiliation(s)
- Zhe Cui
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Guangyu Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
5
|
Lin Z, Sun L. Research advances in the therapy of metabolic syndrome. Front Pharmacol 2024; 15:1364881. [PMID: 39139641 PMCID: PMC11319131 DOI: 10.3389/fphar.2024.1364881] [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: 01/05/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Metabolic syndrome refers to the pathological state of metabolic disorder of protein, fat, carbohydrate, and other substances in the human body. It is a syndrome composed of a group of complex metabolic disorders, whose pathogenesis includes multiple genetic and acquired entities falling under the category of insulin resistance and chronic low-grade inflammationand. It is a risk factor for increased prevalence and mortality from diabetes and cardiovascular disease. Cardiovascular diseases are the predominant cause of morbidity and mortality globally, thus it is imperative to investigate the impact of metabolic syndrome on alleviating this substantial disease burden. Despite the increasing number of scientists dedicating themselves to researching metabolic syndrome in recent decades, numerous aspects of this condition remain incompletely understood, leaving many questions unanswered. In this review, we present an epidemiological analysis of MetS, explore both traditional and novel pathogenesis, examine the pathophysiological repercussions of metabolic syndrome, summarize research advances, and elucidate the mechanisms underlying corresponding treatment approaches.
Collapse
Affiliation(s)
- Zitian Lin
- Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Zhejiang University-University of Edinburgh Institute, International Campus, Zhejiang University, Haining, China
| | - Luning Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
6
|
Liao HJ, Yang YP, Liu YH, Tseng HC, Huo TI, Chiou SH, Chang CH. Harnessing the potential of mesenchymal stem cells-derived exosomes in degenerative diseases. Regen Ther 2024; 26:599-610. [PMID: 39253597 PMCID: PMC11382214 DOI: 10.1016/j.reth.2024.08.001] [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: 06/28/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained attention as a promising therapeutic approach in both preclinical and clinical osteoarthritis (OA) settings. Various joint cell types, such as chondrocytes, synovial fibroblasts, osteoblasts, and tenocytes, can produce and release extracellular vesicles (EVs), which subsequently influence the biological activities of recipient cells. Recently, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have shown the potential to modulate various physiological and pathological processes through the modulation of cellular differentiation, immune responses, and tissue repair. This review explores the roles and therapeutic potential of MSC-EVs in OA and rheumatoid arthritis, cardiovascular disease, age-related macular degeneration, Alzheimer's disease, and other degenerative diseases. Notably, we provide a comprehensive summary of exosome biogenesis, microRNA composition, mechanisms of intercellular transfer, and their evolving role in the highlight of exosome-based treatments in both preclinical and clinical avenues.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Huan-Chin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ia Huo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, Taiwan
| |
Collapse
|
7
|
Graf S, Biemmi V, Arnold M, Segiser A, Müller A, Méndez‐Carmona N, Egle M, Siepe M, Barile L, Longnus S. Macrophage-derived extracellular vesicles alter cardiac recovery and metabolism in a rat heart model of donation after circulatory death. J Cell Mol Med 2024; 28:e18281. [PMID: 38652092 PMCID: PMC11037406 DOI: 10.1111/jcmm.18281] [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: 10/19/2023] [Revised: 02/11/2024] [Accepted: 03/14/2024] [Indexed: 04/25/2024] Open
Abstract
Conditions to which the cardiac graft is exposed during transplantation with donation after circulatory death (DCD) can trigger the recruitment of macrophages that are either unpolarized (M0) or pro-inflammatory (M1) as well as the release of extracellular vesicles (EV). We aimed to characterize the effects of M0 and M1 macrophage-derived EV administration on post-ischaemic functional recovery and glucose metabolism using an isolated rat heart model of DCD. Isolated rat hearts were subjected to 20 min aerobic perfusion, followed by 27 min global, warm ischaemia or continued aerobic perfusion and 60 min reperfusion with or without intravascular administration of EV. Four experimental groups were compared: (1) no ischaemia, no EV; (2) ischaemia, no EV; (3) ischaemia with M0-macrophage-dervied EV; (4) ischaemia with M1-macrophage-derived EV. Post-ischaemic ventricular and metabolic recovery were evaluated. During reperfusion, ventricular function was decreased in untreated ischaemic and M1-EV hearts, but not in M0-EV hearts, compared to non-ischaemic hearts (p < 0.05). In parallel with the reduced functional recovery in M1-EV versus M0-EV ischaemic hearts, rates of glycolysis from exogenous glucose and oxidative metabolism tended to be lower, while rates of glycogenolysis and lactate release tended to be higher. EV from M0- and M1-macrophages differentially affect post-ischaemic cardiac recovery, potentially by altering glucose metabolism in a rat model of DCD. Targeted EV therapy may be a useful approach for modulating cardiac energy metabolism and optimizing graft quality in the setting of DCD.
Collapse
Affiliation(s)
- Selianne Graf
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Graduate School of Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Vanessa Biemmi
- Laboratory for Cardiovascular TheranosticsCardiocentro Ticino Institute‐EOCLuganoSwitzerland
| | - Maria Arnold
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Adrian Segiser
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Anja Müller
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Natalia Méndez‐Carmona
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Manuel Egle
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Graduate School of Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Matthias Siepe
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
| | - Lucio Barile
- Laboratory for Cardiovascular TheranosticsCardiocentro Ticino Institute‐EOCLuganoSwitzerland
| | - Sarah Longnus
- Department of Cardiac SurgeryInselspital Bern University Hospital, University of BernBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| |
Collapse
|
8
|
Yao J, Zhou Y, Yao Z, Meng Y, Yu W, Yang X, Zhou D, Yang X, Zhou Y. A novel machine learning-derived four-gene signature predicts STEMI and post-STEMI heart failure. BIOMOLECULES & BIOMEDICINE 2024; 24:423-433. [PMID: 37715537 PMCID: PMC10950350 DOI: 10.17305/bb.2023.9629] [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: 08/08/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/17/2023]
Abstract
High mortality and morbidity rates associated with ST-elevation myocardial infarction (STEMI) and post-STEMI heart failure (HF) necessitate proper risk stratification for coronary artery disease (CAD). A prediction model that combines specificity and convenience is highly required. This study aimed to design a monocyte-based gene assay for predicting STEMI and post-STEMI HF. A total of 1,956 monocyte expression profiles and corresponding clinical data were integrated from multiple sources. Meta-results were obtained through the weighted gene co-expression network analysis (WGCNA) and differential analysis to identify characteristic genes for STEMI. Machine learning models based on the decision tree (DT), support vector machine (SVM), and random forest (RF) algorithms were trained and validated. Five genes overlapped and were subjected to the model proposal. The discriminative performance of the DT model outperformed the other two methods. The established four-gene panel (HLA-J, CFP, STX11, and NFYC) could discriminate STEMI and HF with an area under the curve (AUC) of 0.86 or above. In the gene set enrichment analysis (GSEA), several cardiac pathogenesis pathways and cardiovascular disorder signatures showed statistically significant, concordant differences between subjects with high and low expression levels of the four-gene panel, affirming the validity of the established model. In conclusion, we have developed and validated a model that offers the hope for accurately predicting the risk of STEMI and HF, leading to optimal risk stratification and personalized management of CAD, thereby improving individual outcomes.
Collapse
Affiliation(s)
- Jialu Yao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Institute for Hypertension of Soochow University, Jiangsu Engineering Laboratory of Novel Functional Polymeric Materials of Soochow University, Suzhou, Jiangsu Province, China
| | - Yujia Zhou
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhichao Yao
- Department of Vascular Surgery, Gusu School of Nanjing Medical University, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital (HQ), Suzhou, Jiangsu Province, China
| | - Ye Meng
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Wangjianfei Yu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Xinyu Yang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Dayong Zhou
- Department of Vascular Surgery, Gusu School of Nanjing Medical University, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital (HQ), Suzhou, Jiangsu Province, China
| | - Xiaoqin Yang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Institute for Hypertension of Soochow University, Jiangsu Engineering Laboratory of Novel Functional Polymeric Materials of Soochow University, Suzhou, Jiangsu Province, China
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Institute for Hypertension of Soochow University, Jiangsu Engineering Laboratory of Novel Functional Polymeric Materials of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
9
|
Omoto ACM, do Carmo JM, da Silva AA, Hall JE, Mouton AJ. Immunometabolism, extracellular vesicles and cardiac injury. Front Endocrinol (Lausanne) 2024; 14:1331284. [PMID: 38260141 PMCID: PMC10800986 DOI: 10.3389/fendo.2023.1331284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Recent evidence from our lab and others suggests that metabolic reprogramming of immune cells drives changes in immune cell phenotypes along the inflammatory-to-reparative spectrum and plays a critical role in mediating the inflammatory responses to cardiac injury (e.g. hypertension, myocardial infarction). However, the factors that drive metabolic reprogramming in immune cells are not fully understood. Extracellular vesicles (EVs) are recognized for their ability to transfer cargo such as microRNAs from remote sites to influence cardiac remodeling. Furthermore, conditions such as obesity and metabolic syndrome, which are implicated in the majority of cardiovascular disease (CVD) cases, can skew production of EVs toward pro-inflammatory phenotypes. In this mini-review, we discuss the mechanisms by which EVs may influence immune cell metabolism during cardiac injury and factors associated with obesity and the metabolic syndrome that can disrupt normal EV function. We also discuss potential sources of cardio-protective and anti-inflammatory EVs, such as brown adipose tissue. Finally, we discuss implications for future therapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
10
|
Qiuyue L, Gulin D, Hong X, Jiazhen Y, Rukui Y, Xinwu H, Guochun L. Zhilong Huoxue Tongyu Capsule Ameliorates Platelet Aggregation and Thrombus Induced by Aspirin in Rats by Regulating Lipid Metabolism and MicroRNA Pathway. Comb Chem High Throughput Screen 2024; 27:854-862. [PMID: 37438906 DOI: 10.2174/1386207326666230712110103] [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: 12/04/2022] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Zhilong Huoxue Tongyu capsule (ZLHX) is a traditional Chinese medicinal compound preparation, which exhibits obvious therapeutic effects on aspirin resistance (AR). However, the mechanism of ZLHX on AR is rarely reported. OBJECTIVES This study aimed to explore the therapeutic effects of AR and the underlying mechanisms of ZLHX on AR rats. METHODS An AR model was established through treatment with a high-fat, high-sugar, and highsalt diet for 12 weeks and oral administration of aspirin (27 mg/kg/day) and ibuprofen (36 mg/kg/day) in weeks 9-12. The rats were administrated with ZLHX (225, 450, and 900 mg/kg) from week 12 to week 16. Blood samples were collected after the experiment. Thromboelastography analysis was performed, and the levels of triglyceride (TG), total cholesterol (TC), lowdensity lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) were determined. Furthermore, the levels of thromboxane B2 (TXB2) and 6-keto-prostaglandin F1α (6- keto-PGF1α) were determined with commercial ELISA kits. Finally, the gene expressions of microRNA- 126-3p (miRNA-126-3p) and miRNA-34b-3p were detected through a real-time quantitative polymerase chain reaction. RESULTS Results demonstrated that ZLHX significantly inhibited platelet aggregation in the AR rats. Moreover, ZLHX markedly decreased the levels of TC, TG, and LDL-C and increased the level of HDL-C. Meanwhile, ELISA results confirmed that ZLHX can elevate the expression levels of TXB2 and 6-keto-PGF1α. Further studies suggested that ZLHX significantly downregulated the expression levels of miRNA-126-3p and miRNA-34b-3p. CONCLUSION This study revealed that the therapeutic effect of ZLHX might be related to the regulation of lipid metabolism and the miRNA pathway.
Collapse
Affiliation(s)
- Li Qiuyue
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Deng Gulin
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Xu Hong
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Yin Jiazhen
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Yuan Rukui
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Huang Xinwu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Li Guochun
- National Traditional Chinese Medicine Clinical Research Base and Pharmacy Intravenous Admixture Service of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
11
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
12
|
Xiong Z, An Q, Chen L, Xiang Y, Li L, Zheng Y. Cell or cell derivative-laden hydrogels for myocardial infarction therapy: from the perspective of cell types. J Mater Chem B 2023; 11:9867-9888. [PMID: 37751281 DOI: 10.1039/d3tb01411h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Myocardial infarction (MI) is a global cardiovascular disease with high mortality and morbidity. To treat acute MI, various therapeutic approaches have been developed, including cells, extracellular vesicles, and biomimetic nanoparticles. However, the clinical application of these therapies is limited due to low cell viability, inadequate targetability, and rapid elimination from cardiac sites. Injectable hydrogels, with their three-dimensional porous structure, can maintain the biomechanical stabilization of hearts and the transplantation activity of cells. However, they cannot regenerate cardiomyocytes or repair broken hearts. A better understanding of the collaborative relationship between hydrogel delivery systems and cell or cell-inspired therapy will facilitate advancing innovative therapeutic strategies against MI. Following that, from the perspective of cell types, MI progression and recent studies on using hydrogel to deliver cell or cell-derived preparations for MI treatment are discussed. Finally, current challenges and future prospects of cell or cell derivative-laden hydrogels for MI therapy are proposed.
Collapse
Affiliation(s)
- Ziqing Xiong
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yucheng Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China.
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Nagesh PT, Nishi H, Rawal S, Zahr T, Miano JM, Sorci-Thomas M, Xu H, Akbar N, Choudhury RP, Misra A, Fisher EA. HDL regulates TGFß-receptor lipid raft partitioning, restoring contractile features of cholesterol-loaded vascular smooth muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.562786. [PMID: 37905061 PMCID: PMC10614922 DOI: 10.1101/2023.10.19.562786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background Cholesterol-loading of mouse aortic vascular smooth muscle cells (mVSMCs) downregulates miR-143/145, a master regulator of the contractile state downstream of TGFβ signaling. In vitro, this results in transitioning from a contractile mVSMC to a macrophage-like state. This process likely occurs in vivo based on studies in mouse and human atherosclerotic plaques. Objectives To test whether cholesterol-loading reduces VSMC TGFβ signaling and if cholesterol efflux will restore signaling and the contractile state in vitro and in vivo. Methods Human coronary artery (h)VSMCs were cholesterol-loaded, then treated with HDL (to promote cholesterol efflux). For in vivo studies, partial conditional deletion of Tgfβr2 in lineage-traced VSMC mice was induced. Mice wild-type for VSMC Tgfβr2 or partially deficient (Tgfβr2+/-) were made hypercholesterolemic to establish atherosclerosis. Mice were then treated with apoA1 (which forms HDL). Results Cholesterol-loading of hVSMCs downregulated TGFβ signaling and contractile gene expression; macrophage markers were induced. TGFβ signaling positively regulated miR-143/145 expression, increasing Acta2 expression and suppressing KLF4. Cholesterol-loading localized TGFβ receptors into lipid rafts, with consequent TGFβ signaling downregulation. Notably, in cholesterol-loaded hVSMCs HDL particles displaced receptors from lipid rafts and increased TGFβ signaling, resulting in enhanced miR-145 expression and decreased KLF4-dependent macrophage features. ApoA1 infusion into Tgfβr2+/- mice restored Acta2 expression and decreased macrophage-marker expression in plaque VSMCs, with evidence of increased TGFβ signaling. Conclusions Cholesterol suppresses TGFβ signaling and the contractile state in hVSMC through partitioning of TGFβ receptors into lipid rafts. These changes can be reversed by promotion of cholesterol efflux, consistent with evidence in vivo.
Collapse
Affiliation(s)
- Prashanth Thevkar Nagesh
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Hitoo Nishi
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Shruti Rawal
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Tarik Zahr
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hao Xu
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| |
Collapse
|
14
|
Bernini Di Michele A, Onofri V, Pesaresi M, Turchi C. The Role of miRNA Expression Profile in Sudden Cardiac Death Cases. Genes (Basel) 2023; 14:1954. [PMID: 37895303 PMCID: PMC10606010 DOI: 10.3390/genes14101954] [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: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Sudden cardiac death (SCD) is one of the leading causes of death in the world and for this reason it has attracted the attention of numerous researchers in the field of legal medicine. It is not easy to determine the cause in a SCD case and the available methods used for diagnosis cannot always give an exhaustive answer. In addition, the molecular analysis of genes does not lead to a clear conclusion, but it could be interesting to focus attention on the expression level of miRNAs, a class of non-coding RNA of about 22 nucleotides. The role of miRNAs is to regulate the gene expression through complementary binding to 3'-untraslated regions of miRNAs, leading to the inhibition of translation or to mRNA degradation. In recent years, several studies were performed with the aim of exploring the use of these molecules as biomarkers for SCD cases, and to also distinguish the causes that lead to cardiac death. In this review, we summarize experiments, evidence, and results of different studies on the implication of miRNAs in SCD cases. We discuss the different biological starting materials with their respective advantages and disadvantages, studying miRNA expression on tissue (fresh-frozen tissue and FFPE tissue), circulating cell-free miRNAs in blood of patients affected by cardiac disease at high risk of SCD, and exosomal miRNAs analyzed from serum of people who died from SCD.
Collapse
Affiliation(s)
- Alessia Bernini Di Michele
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Valerio Onofri
- Legal Medicine Unit, AOU Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy;
| | - Mauro Pesaresi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Chiara Turchi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| |
Collapse
|
15
|
Shekari F, Alibhai FJ, Baharvand H, Börger V, Bruno S, Davies O, Giebel B, Gimona M, Salekdeh GH, Martin‐Jaular L, Mathivanan S, Nelissen I, Nolte‐’t Hoen E, O'Driscoll L, Perut F, Pluchino S, Pocsfalvi G, Salomon C, Soekmadji C, Staubach S, Torrecilhas AC, Shelke GV, Tertel T, Zhu D, Théry C, Witwer K, Nieuwland R. Cell culture-derived extracellular vesicles: Considerations for reporting cell culturing parameters. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e115. [PMID: 38939735 PMCID: PMC11080896 DOI: 10.1002/jex2.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/18/2023] [Accepted: 09/17/2023] [Indexed: 06/29/2024]
Abstract
Cell culture-conditioned medium (CCM) is a valuable source of extracellular vesicles (EVs) for basic scientific, therapeutic and diagnostic applications. Cell culturing parameters affect the biochemical composition, release and possibly the function of CCM-derived EVs (CCM-EV). The CCM-EV task force of the Rigor and Standardization Subcommittee of the International Society for Extracellular Vesicles aims to identify relevant cell culturing parameters, describe their effects based on current knowledge, recommend reporting parameters and identify outstanding questions. While some recommendations are valid for all cell types, cell-specific recommendations may need to be established for non-mammalian sources, such as bacteria, yeast and plant cells. Current progress towards these goals is summarized in this perspective paper, along with a checklist to facilitate transparent reporting of cell culturing parameters to improve the reproducibility of CCM-EV research.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP‐TDC), Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | | | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in BiologyUniversity of Science and CultureTehranIran
| | - Verena Börger
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Stefania Bruno
- Department of Medical Sciences and Molecular Biotechnology CenterUniversity of TorinoTurinItaly
| | - Owen Davies
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Mario Gimona
- GMP UnitSpinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS) and Research Program “Nanovesicular Therapies” Paracelsus Medical UniversitySalzburgAustria
| | | | - Lorena Martin‐Jaular
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneVICAustralia
| | - Inge Nelissen
- VITO (Flemish Institute for Technological Research), Health departmentBoeretangBelgium
| | - Esther Nolte‐’t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | - Francesca Perut
- Biomedical Science and Technologies and Nanobiotechnology LabIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Gabriella Pocsfalvi
- Institute of Biosciences and BioResourcesNational Research CouncilNaplesItaly
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
| | - Carolina Soekmadji
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | | | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)SPBrazil
| | - Ganesh Vilas Shelke
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Dandan Zhu
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVICAustralia
| | - Clotilde Théry
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Kenneth Witwer
- Departments of Molecular and Comparative Pathobiology and Neurology and Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, Amsterdam University Medical CentersLocation AMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
16
|
Tian C, Ziegler JN, Zucker IH. Extracellular Vesicle MicroRNAs in Heart Failure: Pathophysiological Mediators and Therapeutic Targets. Cells 2023; 12:2145. [PMID: 37681877 PMCID: PMC10486980 DOI: 10.3390/cells12172145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023] Open
Abstract
Extracellular vesicles (EVs) are emerging mediators of intracellular and inter-organ communications in cardiovascular diseases (CVDs), especially in the pathogenesis of heart failure through the transference of EV-containing bioactive substances. microRNAs (miRNAs) are contained in EV cargo and are involved in the progression of heart failure. Over the past several years, a growing body of evidence has suggested that the biogenesis of miRNAs and EVs is tightly regulated, and the sorting of miRNAs into EVs is highly selective and tightly controlled. Extracellular miRNAs, particularly circulating EV-miRNAs, have shown promising potential as prognostic and diagnostic biomarkers for heart failure and as therapeutic targets. In this review, we summarize the latest progress concerning the role of EV-miRNAs in HF and their application in a therapeutic strategy development for heart failure.
Collapse
Affiliation(s)
- Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA;
| | - Jessica N. Ziegler
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA;
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| |
Collapse
|
17
|
Spinetti G, Mutoli M, Greco S, Riccio F, Ben-Aicha S, Kenneweg F, Jusic A, de Gonzalo-Calvo D, Nossent AY, Novella S, Kararigas G, Thum T, Emanueli C, Devaux Y, Martelli F. Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems. Cardiovasc Diabetol 2023; 22:122. [PMID: 37226245 PMCID: PMC10206598 DOI: 10.1186/s12933-023-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by high levels of blood glucose caused by insulin defect or impairment, is a major risk factor for cardiovascular diseases and related mortality. Patients with diabetes experience a state of chronic or intermittent hyperglycemia resulting in damage to the vasculature, leading to micro- and macro-vascular diseases. These conditions are associated with low-grade chronic inflammation and accelerated atherosclerosis. Several classes of leukocytes have been implicated in diabetic cardiovascular impairment. Although the molecular pathways through which diabetes elicits an inflammatory response have attracted significant attention, how they contribute to altering cardiovascular homeostasis is still incompletely understood. In this respect, non-coding RNAs (ncRNAs) are a still largely under-investigated class of transcripts that may play a fundamental role. This review article gathers the current knowledge on the function of ncRNAs in the crosstalk between immune and cardiovascular cells in the context of diabetic complications, highlighting the influence of biological sex in such mechanisms and exploring the potential role of ncRNAs as biomarkers and targets for treatments. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.
Collapse
Affiliation(s)
- Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Martina Mutoli
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Federica Riccio
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Soumaya Ben-Aicha
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susana Novella
- Department of Physiology, University of Valencia - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Georgios Kararigas
- Department of Physiology, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Costanza Emanueli
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy.
| |
Collapse
|
18
|
Dutta B, Loo S, Kam A, Sze SK, Tam JP. Ginsentide TP1 Protects Hypoxia-Induced Dysfunction and ER Stress-Linked Apoptosis. Cells 2023; 12:1401. [PMID: 37408235 PMCID: PMC10216702 DOI: 10.3390/cells12101401] [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: 05/02/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Hypoxia-induced vascular endothelial dysfunction (VED) is a significant contributor to several severe human diseases, including heart disease, stroke, dementia, and cancer. However, current treatment options for VED are limited due to the lack of understanding of the underlying disease mechanisms and therapeutic leads. We recently discovered a heat-stable microprotein in ginseng, called ginsentide TP1, that has been shown to reduce vascular dysfunction in cardiovascular disease models. In this study, we use a combination of functional assays and quantitative pulsed SILAC proteomics to identify new proteins synthesized in hypoxia and to show that ginsentide TP1 provides protection for human endothelial cells against hypoxia and ER stress. Consistent with the reported findings, we also found that hypoxia activates various pathways related to endothelium activation and monocyte adhesion, which in turn, impairs nitric oxide (NO) synthase activity, reduces the bioavailability of NO, and increases the production of reactive oxygen species that contribute to VED. Additionally, hypoxia triggers endoplasmic reticulum stress and initiates apoptotic signaling pathways associated with cardiovascular pathology. Treatment with ginsentide TP1 reduced surface adhesion molecule expression, prevented activation of the endothelium and leukocyte adhesion, restored protein hemostasis, and reduced ER stress to protect against hypoxia-induced cell death. Ginsentide TP1 also restored NO signaling and bioavailability, reduced oxidative stress, and protected endothelial cells from endothelium dysfunction. In conclusion, this study shows that the molecular pathogenesis of VED induced by hypoxia can be mitigated by treatment with ginsentide TP1, which could be one of the key bioactive compounds responsible for the "cure-all" effect of ginseng. This research may lead to the development of new therapies for cardiovascular disorders.
Collapse
Affiliation(s)
- Bamaprasad Dutta
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
| | - Shining Loo
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Antony Kam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Department of Health Sciences, Brock University, Niagara Region, St. Catharines, ON L2S 3A1, Canada
| | - James P. Tam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
| |
Collapse
|
19
|
Jan Z, Hočevar M, Kononenko V, Michelini S, Repar N, Caf M, Kocjančič B, Dolinar D, Kralj S, Makovec D, Iglič A, Drobne D, Jenko M, Kralj-Iglič V. Inflammatory, Oxidative Stress and Small Cellular Particle Response in HUVEC Induced by Debris from Endoprosthesis Processing. MATERIALS (BASEL, SWITZERLAND) 2023; 16:ma16093287. [PMID: 37176169 PMCID: PMC10179554 DOI: 10.3390/ma16093287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
We studied inflammatory and oxidative stress-related parameters and cytotoxic response of human umbilical vein endothelial cells (HUVEC) to a 24 h treatment with milled particles simulating debris involved in sandblasting of orthopedic implants (OI). We used different abrasives (corundum-(Al2O3), used corundum retrieved from removed OI (u. Al2O3), and zirconia/silica composite (ZrO2/SiO2)). Morphological changes were observed by scanning electron microscopy (SEM). Concentration of Interleukins IL-6 and IL-1β and Tumor Necrosis Factor α (TNF)-α was assessed by enzyme-linked immunosorbent assay (ELISA). Activity of Cholinesterase (ChE) and Glutathione S-transferase (GST) was measured by spectrophotometry. Reactive oxygen species (ROS), lipid droplets (LD) and apoptosis were measured by flow cytometry (FCM). Detachment of the cells from glass and budding of the cell membrane did not differ in the treated and untreated control cells. Increased concentration of IL-1β and of IL-6 was found after treatment with all tested particle types, indicating inflammatory response of the treated cells. Increased ChE activity was found after treatment with u. Al2O3 and ZrO2/SiO2. Increased GST activity was found after treatment with ZrO2/SiO2. Increased LD quantity but not ROS quantity was found after treatment with u. Al2O3. No cytotoxicity was detected after treatment with u. Al2O3. The tested materials in concentrations added to in vitro cell lines were found non-toxic but bioactive and therefore prone to induce a response of the human body to OI.
Collapse
Affiliation(s)
- Zala Jan
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, SI-1000 Ljubljana, Slovenia
| | - Matej Hočevar
- Institute of Metals and Technology, SI-1000 Ljubljana, Slovenia
| | - Veno Kononenko
- University of Ljubljana, Biotechnical Faculty, Nanobiology Group, SI-1000 Ljubljana, Slovenia
| | - Sara Michelini
- University of Ljubljana, Biotechnical Faculty, Nanobiology Group, SI-1000 Ljubljana, Slovenia
| | - Neža Repar
- University of Ljubljana, Biotechnical Faculty, Nanobiology Group, SI-1000 Ljubljana, Slovenia
| | - Maja Caf
- Department for Materials Synthesis, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Boštjan Kocjančič
- University of Ljubljana, Faculty of Medicine, Chair of Orthopaedics, SI-1000 Ljubljana, Slovenia
- MD-RI Institute for Materials Research in Medicine, SI-1000 Ljubljana, Slovenia
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Drago Dolinar
- University of Ljubljana, Faculty of Medicine, Chair of Orthopaedics, SI-1000 Ljubljana, Slovenia
- MD-RI Institute for Materials Research in Medicine, SI-1000 Ljubljana, Slovenia
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Slavko Kralj
- Department for Materials Synthesis, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Darko Makovec
- Department for Materials Synthesis, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Aleš Iglič
- University of Ljubljana, Faculty of Electrical Engineering, Laboratory of Physics, SI-1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Medicine, Laboratory of Clinical Biophysics, SI-1000 Ljubljana, Slovenia
| | - Damjana Drobne
- University of Ljubljana, Biotechnical Faculty, Nanobiology Group, SI-1000 Ljubljana, Slovenia
| | - Monika Jenko
- MD-RI Institute for Materials Research in Medicine, SI-1000 Ljubljana, Slovenia
| | - Veronika Kralj-Iglič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
20
|
Panda R, Kubes P. Extracellular vesicles selectively mobilize splenic neutrophils. Cardiovasc Res 2023; 119:1-2. [PMID: 36691968 DOI: 10.1093/cvr/cvad015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 01/25/2023] Open
Affiliation(s)
- Rachita Panda
- Department of Physiology and Pharmacology, Health Science Centre, Room 1817, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, Health Science Centre, Room 1817, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
21
|
Davidson SM, Boulanger CM, Aikawa E, Badimon L, Barile L, Binder CJ, Brisson A, Buzas E, Emanueli C, Jansen F, Katsur M, Lacroix R, Lim SK, Mackman N, Mayr M, Menasché P, Nieuwland R, Sahoo S, Takov K, Thum T, Vader P, Wauben MHM, Witwer K, Sluijter JPG. Methods for the identification and characterization of extracellular vesicles in cardiovascular studies: from exosomes to microvesicles. Cardiovasc Res 2023; 119:45-63. [PMID: 35325061 PMCID: PMC10233250 DOI: 10.1093/cvr/cvac031] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized vesicles with a lipid bilayer that are released from cells of the cardiovascular system, and are considered important mediators of intercellular and extracellular communications. Two types of EVs of particular interest are exosomes and microvesicles, which have been identified in all tissue and body fluids and carry a variety of molecules including RNAs, proteins, and lipids. EVs have potential for use in the diagnosis and prognosis of cardiovascular diseases and as new therapeutic agents, particularly in the setting of myocardial infarction and heart failure. Despite their promise, technical challenges related to their small size make it challenging to accurately identify and characterize them, and to study EV-mediated processes. Here, we aim to provide the reader with an overview of the techniques and technologies available for the separation and characterization of EVs from different sources. Methods for determining the protein, RNA, and lipid content of EVs are discussed. The aim of this document is to provide guidance on critical methodological issues and highlight key points for consideration for the investigation of EVs in cardiovascular studies.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, WC1E 6HX London, UK
| | - Chantal M Boulanger
- Université Paris Cité, Paris-Cardiovascular Research Center, INSERM, Paris, France
| | - Elena Aikawa
- Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lina Badimon
- Cardiovascular Science Program-ICCC, IR-Hospital de la Santa Creu i Santa Pau-IIBSantPau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale and Faculty of Biomedical Sciences, Università Svizzera italiana, 6900 Lugano, Switzerland
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Alain Brisson
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Edit Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, HCEMM-SU and ELKH-SE Immune Proteogenomics Extracellular Vesicle Research Group, Budapest, Hungary
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Felix Jansen
- Department of Internal Medicine II, Heart Center, University Hospital Bonn, Bonn, Germany
| | - Miroslava Katsur
- The Hatter Cardiovascular Institute, University College London, WC1E 6HX London, UK
| | - Romaric Lacroix
- Aix Marseille University, INSERM 1263, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
- Department of Haematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Sai Kiang Lim
- Institute of Medical Biology and Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France
- Laboratory of Experimental Cardiology, Department of Cardiology, UMC Utrecht Regenerative Medicine Center and Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rienk Nieuwland
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaloyan Takov
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Pieter Vader
- Université Paris Cité, Paris-Cardiovascular Research Center, INSERM, Paris, France
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marca H M Wauben
- Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 2, Utrecht, The Netherlands
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, Department of Cardiology, UMC Utrecht Regenerative Medicine Center and Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
22
|
Akbar N, Braithwaite AT, Corr EM, Koelwyn GJ, van Solingen C, Cochain C, Saliba AE, Corbin A, Pezzolla D, Møller Jørgensen M, Bæk R, Edgar L, De Villiers C, Gunadasa-Rohling M, Banerjee A, Paget D, Lee C, Hogg E, Costin A, Dhaliwal R, Johnson E, Krausgruber T, Riepsaame J, Melling GE, Shanmuganathan M, Bock C, Carter DRF, Channon KM, Riley PR, Udalova IA, Moore KJ, Anthony DC, Choudhury RP. Rapid neutrophil mobilization by VCAM-1+ endothelial cell-derived extracellular vesicles. Cardiovasc Res 2023; 119:236-251. [PMID: 35134856 PMCID: PMC10022859 DOI: 10.1093/cvr/cvac012] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/28/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Acute myocardial infarction rapidly increases blood neutrophils (<2 h). Release from bone marrow, in response to chemokine elevation, has been considered their source, but chemokine levels peak up to 24 h after injury, and after neutrophil elevation. This suggests that additional non-chemokine-dependent processes may be involved. Endothelial cell (EC) activation promotes the rapid (<30 min) release of extracellular vesicles (EVs), which have emerged as an important means of cell-cell signalling and are thus a potential mechanism for communicating with remote tissues. METHODS AND RESULTS Here, we show that injury to the myocardium rapidly mobilizes neutrophils from the spleen to peripheral blood and induces their transcriptional activation prior to arrival at the injured tissue. Time course analysis of plasma-EV composition revealed a rapid and selective increase in EVs bearing VCAM-1. These EVs, which were also enriched for miRNA-126, accumulated preferentially in the spleen where they induced local inflammatory gene and chemokine protein expression, and mobilized splenic-neutrophils to peripheral blood. Using CRISPR/Cas9 genome editing, we generated VCAM-1-deficient EC-EVs and showed that its deletion removed the ability of EC-EVs to provoke the mobilization of neutrophils. Furthermore, inhibition of miRNA-126 in vivo reduced myocardial infarction size in a mouse model. CONCLUSIONS Our findings show a novel EV-dependent mechanism for the rapid mobilization of neutrophils to peripheral blood from a splenic reserve and establish a proof of concept for functional manipulation of EV-communications through genetic alteration of parent cells.
Collapse
Affiliation(s)
- Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Adam T Braithwaite
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Emma M Corr
- NYU Cardiovascular Research Center, Department of Medicine, Division of Cardiology, School of Medicine, New York University School of Medicine, 435 E 30th St. New York, NY 10016, USA
| | - Graeme J Koelwyn
- NYU Cardiovascular Research Center, Department of Medicine, Division of Cardiology, School of Medicine, New York University School of Medicine, 435 E 30th St. New York, NY 10016, USA
| | - Coen van Solingen
- NYU Cardiovascular Research Center, Department of Medicine, Division of Cardiology, School of Medicine, New York University School of Medicine, 435 E 30th St. New York, NY 10016, USA
| | - Clément Cochain
- Comprehensive Heart Failure Center, University Hospital Wurzburg, Anstalt des öffentlichen Rechts Josef-Schneider-Straße 2 97080 Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Inhoffenstraße 7 38124 Braunschweig, Würzburg, Germany
| | - Alastair Corbin
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7FY, UK
| | - Daniela Pezzolla
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Urbansgade 32-36, DK-9000, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Denmark
| | - Rikke Bæk
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Denmark
| | - Laurienne Edgar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Carla De Villiers
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building Parks Road, OX1 3PT, Oxford, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building Parks Road, OX1 3PT, Oxford, UK
| | - Abhirup Banerjee
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Daan Paget
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Charlotte Lee
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Eleanor Hogg
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
| | - Adam Costin
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Raman Dhaliwal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, Vienna, Austria
| | - Joey Riepsaame
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Genevieve E Melling
- Department of Biological and Medical Sciences, Oxford Brookes University, Headington Campus Oxford OX3 0BP, UK
- Institute of Clinical Sciences, School of Biomedical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Mayooran Shanmuganathan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
- The OxAMI Study is detailed in the Supplementary Acknowledgments
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Spitalgasse 23, BT88 1090, Vienna, Austria
| | - David R F Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, Headington Campus Oxford OX3 0BP, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
- The OxAMI Study is detailed in the Supplementary Acknowledgments
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building Parks Road, OX1 3PT, Oxford, UK
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7FY, UK
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, Department of Medicine, Division of Cardiology, School of Medicine, New York University School of Medicine, 435 E 30th St. New York, NY 10016, USA
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine University of Oxford Level 6, West Wing John Radcliffe Hospital Headington Oxford OX3 9DU, UK
- The OxAMI Study is detailed in the Supplementary Acknowledgments
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| |
Collapse
|
23
|
Shekari F, Abyadeh M, Meyfour A, Mirzaei M, Chitranshi N, Gupta V, Graham SL, Salekdeh GH. Extracellular Vesicles as reconfigurable therapeutics for eye diseases: Promises and hurdles. Prog Neurobiol 2023; 225:102437. [PMID: 36931589 DOI: 10.1016/j.pneurobio.2023.102437] [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: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
A large number of people worldwide suffer from visual impairment. However, most available therapies rely on impeding the development of a particular eye disorder. Therefore, there is an increasing demand for effective alternative treatments, specifically regenerative therapies. Extracellular vesicles, including exosomes, ectosomes, or microvesicles, are released by cells and play a potential role in regeneration. Following an introduction to EV biogenesis and isolation methods, this integrative review provides an overview of our current knowledge about EVs as a communication paradigm in the eye. Then, we focused on the therapeutic applications of EVs derived from conditioned medium, biological fluid, or tissue and highlighted some recent developments in strategies to boost the innate therapeutic potential of EVs by loading various kinds of drugs or being engineered at the level of producing cells or EVs. Challenges faced in the development of safe and effective translation of EV-based therapy into clinical settings for eye diseases are also discussed to pave the road toward reaching feasible regenerative therapies required for eye-related complications.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | | | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | | |
Collapse
|
24
|
Li T, Han X, Chen S, Wang B, Teng Y, Cheng W, Lu Z, Li Y, Wu X, Jiang Y, Wang L, Liu L, Zhao M. Effects of Exercise on Extracellular Vesicles in Patients with Metabolic Dysfunction: a Systematic Review. J Cardiovasc Transl Res 2023; 16:97-111. [PMID: 35655108 DOI: 10.1007/s12265-022-10282-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
Abstract
The aim of this study was to investigate the effect of exercise on extracellular vesicles (EVs) in patients with metabolic dysfunction. The literatures were searched until Apr 28, 2022, and 16 studies that met inclusion criteria were included in this review. The results showed that the concentrations of platelet-derived extracellular vesicles (PEVs) and endothelial cell-derived extracellular vesicles (EEVs) decreased after long-term exercise, especially for CD62E+ EEVs and CD105+ EEVs. Simultaneously, exercise improved the concentration of clinical evaluation indicators of metabolic diseases, and the changes in these indicators were positively correlated with the changes of EEVs and PEVs. The concentration of skeletal muscle-derived extracellular vesicles (SkEVs) increased after a single bout of exercise. The aforementioned results indicated that long-term exercise might improve endothelial function and hypercoagulability in patients with metabolic dysfunction. The changes in concentrations of EVs could assist in assessing effect of exercise on patients with metabolic dysfunction.
Collapse
Affiliation(s)
- Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Xiaowan Han
- Department of Cardiac Rehabilitation, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Shiqi Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Weiting Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Yangyang Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China
| | - Lisong Liu
- Department of Cardiac Rehabilitation, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China.
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People's Republic of China.
| |
Collapse
|
25
|
Eichner-Seitz N. Diagnosis of Extracellular Vesicles in Cardiovascular and Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:171-185. [PMID: 37603280 DOI: 10.1007/978-981-99-1443-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Early detection and identification of those with or at increased risk for cardiovascular disease (CVD) and metabolic dysfunction is crucial for improving disease management and prognosis. Given the complex, multifactorial nature of the pathogenesis of the CVD and metabolic dysfunction, it is essential to have biomarkers that encompass the multiple facets of disease development. The uniquely ubiquitous nature and functionality of extracellular vesicles (EVs) in various disease pathologies can provide novel insight into both diagnosis and prognosis while further improving assessments used in clinical and research practice. Herein we summarize the use of EV count and content (including miRNA and protein) in diagnosis of CVD, obesity, metabolic syndrome, and type 2 diabetes (T2D), as well as highlight the potential utility for enhancing determination of prognosis and long-term complications in these clinical populations. Although the results are promising, future work is needed in both methodology and in relation to other factors such as sex and medications, in order to apply these findings in clinical practice.
Collapse
Affiliation(s)
- N Eichner-Seitz
- Penn State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
26
|
Shen S. Prospective Advances of Extracellular Vesicles Investigation in Cardiovascular and Metabolic Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:231-240. [PMID: 37603283 DOI: 10.1007/978-981-99-1443-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Extracellular vesicles (EVs) play an important role in cardiovascular and metabolic diseases through intercellular communication. Although there has been extensive research on EVs, there are still some unsolved problems in the technologies of investigation of EVs. In this chapter, we reviewed the current knowledge of EVs functions in cardiovascular and metabolic pathophysiology and EVs as biomarkers and therapeutic agents in cardiovascular and metabolic diseases. We also addressed the challenges in isolation and identification of EVs as well as challenges in visualization and tracking of EVs. By addressing these challenges, we hope to have a more in-depth understanding of the biological functions of EVs.
Collapse
Affiliation(s)
- Shutong Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Yuan Y, Ma Y, Aili Z, Nijiati M. Reductions in extracellular vesicle-associated microRNA-126 levels in coronary blood after acute myocardial infarction: A retrospective study. Front Cardiovasc Med 2022; 9:1046839. [DOI: 10.3389/fcvm.2022.1046839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
BackgroundAcute Myocardial Infarction (AMI) is a kind of cardiovascular disease with high mortality and incidence. Extracellular vesicles (EVs) and microRNA-126 (miR-126) are known to play important role in the development and prognosis of several cardiovascular diseases. Therefore, this study aimed to investigate the changes in Extracellular vesicle (EV)-associated miR-126 levels in the coronary blood of patients with AMI to explore the relationship between miR-126 levels and AMI.Materials and methodsWe analyzed EV-associated miR-126 in the coronary blood of patients with AMI and stable coronary artery disease (SCAD) using quantitative reverse transcription polymerase chain reaction (qRT-PCR).ResultsWe tested the coronary blood of 20 patients with AMI and 20 with SCAD. The mean age of the patients was 58.8 ± 10.3 years and 32 (80%) were men. We observed that the EV-associated miR-126 levels were lower in patients with AMI [median = 0.13; interquartile range (IQR): 0.08–0.22] than in patients with SCAD (median = 0.37; IQR: 0.26–0.48) (P < 0.001). In addition, the levels of miR-126 were negatively associated with the Thrombolysis in Myocardial Infarction (TIMI) score (r = −0.66, P = 0.001).ConclusionReduction of EV-associated miR-126 levels in the coronary blood of patients with AMI may be involved in acute coronary thrombosis events.
Collapse
|
28
|
Stability of exosomes in the postmortem serum and preliminary study on exosomal miRNA expression profiling in serum from myocardial infarction cadavers. Int J Legal Med 2022; 137:825-834. [PMID: 36416963 DOI: 10.1007/s00414-022-02913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
Exosome-encapsulated miRNAs could potentially be sensitive biomarkers of human diseases. Since a lipid bilayer membrane surrounds exosomes, the exosomal miRNA may stably exist in body fluids with diseases as well as biological fluids. Therefore, exosomal miRNA may be helpful for autopsy diagnosis. Assuming cadaver blood would be most useful, we initially examined serum exosome stability with regard to storage temperatures and periods. Characteristic analyses of the exosome revealed that exosomes and the content, miRNA, were stably preserved until at least three days when stored at below 20 °C. Subsequently, exosomal miRNA expression profiling was performed on the serum of acute myocardial infarction (AMI, 4 cases) autopsy bodies and on hemorrhagic shock bodies used as the control (CT, 3 cases). Results showed that significant twofold up- and downregulations of expression of 18 and 16 miRNAs were detectable in AMI as compared to the CT, respectively. miR-126-3p, which has been reported to be increased in serum of AMI patients and a mouse model, was one of the significantly upregulated miRNAs. Furthermore, dysregulation of exosomal miRNAs, such as miR-145-5p, miR-143-3p, and miR-222-3p, which are involved in cardioprotection, may be associated with AMI pathogenesis. These findings provide a novel perspective on the potential role of exosomal miRNA in determining the cause of death.
Collapse
|
29
|
Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J Clin Med 2022; 11:jcm11164932. [PMID: 36013171 PMCID: PMC9410115 DOI: 10.3390/jcm11164932] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan E. El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-01-350-000 (ext. 4765)
| |
Collapse
|
30
|
Extracellular Vesicles, Inflammation, and Cardiovascular Disease. Cells 2022; 11:cells11142229. [PMID: 35883672 PMCID: PMC9320258 DOI: 10.3390/cells11142229] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. The underlying mechanisms of most cardiovascular disorders involve innate and adaptive immune responses, and extracellular vesicles are implicated in both. In this review, we describe the mechanistic role of extracellular vesicles at the intersection of inflammatory processes and cardiovascular disease. Our discussion focuses on atherosclerosis, myocardial ischemia and ischemic heart disease, heart failure, aortic aneurysms, and valvular pathology.
Collapse
|
31
|
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis. Cells 2022; 11:cells11111845. [PMID: 35681540 PMCID: PMC9180657 DOI: 10.3390/cells11111845] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality all over the world. Extracellular vesicles (EVs), small lipid-bilayer membrane vesicles released by most cellular types, exert pivotal and multifaceted roles in physiology and disease. Emerging evidence emphasizes the importance of EVs in intercellular communication processes with key effects on cell survival, endothelial homeostasis, inflammation, neoangiogenesis, and thrombosis. This review focuses on EVs as effective signaling molecules able to both derail vascular homeostasis and induce vascular dysfunction, inflammation, plaque progression, and thrombus formation as well as drive anti-inflammation, vascular repair, and atheroprotection. We provide a comprehensive and updated summary of the role of EVs in the development or regression of atherosclerotic lesions, highlighting the link between thrombosis and inflammation. Importantly, we also critically describe their potential clinical use as disease biomarkers or therapeutic agents in atherothrombosis.
Collapse
|
32
|
Sun P, Wang C, Mang G, Xu X, Fu S, Chen J, Wang X, Wang W, Li H, Zhao P, Li Y, Chen Q, Wang N, Tong Z, Fu X, Lang Y, Duan S, Liu D, Zhang M, Tian J. Extracellular vesicle-packaged mitochondrial disturbing miRNA exacerbates cardiac injury during acute myocardial infarction. Clin Transl Med 2022; 12:e779. [PMID: 35452193 PMCID: PMC9028097 DOI: 10.1002/ctm2.779] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Mounting evidence suggests that extracellular vesicles (EVs) are effective communicators in biological signalling in cardiac physiology and pathology. However, the role of EVs in cardiac injury, particularly in ischemic myocardial scenarios, has not been fully elucidated. Here, we report that acute myocardial infarction (AMI)‐induced EVs can impair cardiomyocyte survival and exacerbate cardiac injury. EV‐encapsulated miR‐503, which is enriched during the early phase of AMI, is a critical molecule that mediates myocardial injury. Functional studies revealed that miR‐503 promoted cardiomyocyte death by directly binding to peroxisome proliferator‐activated receptor gamma coactivator‐1β (PGC‐1β) and a mitochondrial deacetylase, sirtuin 3 (SIRT3), thereby triggering mitochondrial metabolic dysfunction and cardiomyocyte death. Mechanistically, we identified endothelial cells as the primary source of miR‐503 in EVs after AMI. Hypoxia induced rapid H3K4 methylation of the promoter of the methyltransferase‐like 3 gene (METTL3) and resulted in its overexpression. METTL3 overexpression evokes N6‐methyladenosine (m6A)‐dependent miR‐503 biogenesis in endothelial cells. In summary, this study highlights a novel endogenous mechanism wherein EVs aggravate myocardial injury during the onset of AMI via endothelial cell‐secreted miR‐503 shuttling.
Collapse
Affiliation(s)
- Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ge Mang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangli Xu
- Department of Ultrasound, The Second Hospital of Harbin city, Harbin, China
| | - Shuai Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jianfeng Chen
- Laboratory Animal Center, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqi Wang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiwei Wang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hairu Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Peng Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yifei Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Qi Chen
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Naixin Wang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhonghua Tong
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Lang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shasha Duan
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Dongmei Liu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Maomao Zhang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
33
|
Liu C, Fan Z, He D, Chen H, Zhang S, Guo S, Zheng B, Cen H, Zhao Y, Liu H, Wang L. Designer Functional Nanomedicine for Myocardial Repair by Regulating the Inflammatory Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14040758. [PMID: 35456592 PMCID: PMC9025700 DOI: 10.3390/pharmaceutics14040758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction is a major global health problem, and the repair of damaged myocardium is still a major challenge. Myocardial injury triggers an inflammatory response: immune cells infiltrate into the myocardium while activating myofibroblasts and vascular endothelial cells, promoting tissue repair and scar formation. Fragments released by cardiomyocytes become endogenous “danger signals”, which are recognized by cardiac pattern recognition receptors, activate resident cardiac immune cells, release thrombin factors and inflammatory mediators, and trigger severe inflammatory responses. Inflammatory signaling plays an important role in the dilation and fibrosis remodeling of the infarcted heart, and is a key event driving the pathogenesis of post-infarct heart failure. At present, there is no effective way to reverse the inflammatory microenvironment in injured myocardium, so it is urgent to find new therapeutic and diagnostic strategies. Nanomedicine, the application of nanoparticles for the prevention, treatment, and imaging of disease, has produced a number of promising applications. This review discusses the treatment and challenges of myocardial injury and describes the advantages of functional nanoparticles in regulating the myocardial inflammatory microenvironment and overcoming side effects. In addition, the role of inflammatory signals in regulating the repair and remodeling of infarcted hearts is discussed, and specific therapeutic targets are identified to provide new therapeutic ideas for the treatment of myocardial injury.
Collapse
Affiliation(s)
- Chunping Liu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhijin Fan
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China;
| | - Dongyue He
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huiqi Chen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Shihui Zhang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Sien Guo
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Bojun Zheng
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huan Cen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Yunxuan Zhao
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Hongxing Liu
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510230, China
- Correspondence: (H.L.); (L.W.)
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- Correspondence: (H.L.); (L.W.)
| |
Collapse
|
34
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part I: Health and Normal Physiology: Part I: Health and Normal Physiology. J Extracell Vesicles 2022; 11:e12151. [PMID: 35041249 PMCID: PMC8765331 DOI: 10.1002/jev2.12151] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Previously thought to be nothing more than cellular debris, extracellular vesicles (EVs) are now known to mediate physiological and pathological functions throughout the body. We now understand more about their capacity to transfer nucleic acids and proteins between distant organs, the interaction of their surface proteins with target cells, and the role of vesicle-bound lipids in health and disease. To date, most observations have been made in reductionist cell culture systems, or as snapshots from patient cohorts. The heterogenous population of vesicles produced in vivo likely act in concert to mediate both beneficial and detrimental effects. EVs play crucial roles in both the pathogenesis of diseases, from cancer to neurodegenerative disease, as well as in the maintenance of system and organ homeostasis. This two-part review draws on the expertise of researchers working in the field of EV biology and aims to cover the functional role of EVs in physiology and pathology. Part I will outline the role of EVs in normal physiology.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordJohn Radcliffe Hospital, HeadingtonOxfordUK
| |
Collapse
|
35
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part II: Pathology: Part II: Pathology. J Extracell Vesicles 2022; 11:e12190. [PMID: 35041301 PMCID: PMC8765328 DOI: 10.1002/jev2.12190] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
It is clear from Part I of this series that extracellular vesicles (EVs) play a critical role in maintaining the homeostasis of most, if not all, normal physiological systems. However, the majority of our knowledge about EV signalling has come from studying them in disease. Indeed, EVs have consistently been associated with propagating disease pathophysiology. The analysis of EVs in biofluids, obtained in the clinic, has been an essential of the work to improve our understanding of their role in disease. However, to interfere with EV signalling for therapeutic gain, a more fundamental understanding of the mechanisms by which they contribute to pathogenic processes is required. Only by discovering how the EV populations in different biofluids change-size, number, and physicochemical composition-in clinical samples, may we then begin to unravel their functional roles in translational models in vitro and in vivo, which can then feedback to the clinic. In Part II of this review series, the functional role of EVs in pathology and disease will be discussed, with a focus on in vivo evidence and their potential to be used as both biomarkers and points of therapeutic intervention.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
36
|
Coly PM, Loyer X. [Extracellular vesicles and cardiovascular diseases]. Med Sci (Paris) 2021; 37:1119-1124. [PMID: 34928215 DOI: 10.1051/medsci/2021204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death globally. There is therefore a need to develop new approaches for the treatment and early detection of these ailments. In the past decades, extracellular vesicles (EVs) have attracted significant attention as their role in intercellular communication has been brought to light. They have been shown to regulate pathways such as cellular inflammation or angiogenesis, and are therefore involved in key aspects of cardiovascular pathophysiology. Interestingly, EVs appear to have a multifaceted role which depends on their origin and cargo. Though at times deleterious, they have also been proposed as promising diagnostic tools and potential therapeutics. This review highlights recent advances in the role of extracellular vesicles in cardiovascular pathologies.
Collapse
Affiliation(s)
- Pierre-Michael Coly
- Université de Paris, Inserm UMR 970, Paris-Centre de recherche cardiovasculaire (Paris-Cardiovascular Research Center), 56 rue Leblanc, F-75015 Paris, France
| | - Xavier Loyer
- Université de Paris, Inserm UMR 970, Paris-Centre de recherche cardiovasculaire (Paris-Cardiovascular Research Center), 56 rue Leblanc, F-75015 Paris, France
| |
Collapse
|
37
|
Wagner KT, Radisic M. A New Role for Extracellular Vesicles in Cardiac Tissue Engineering and Regenerative Medicine. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100047. [PMID: 34927167 PMCID: PMC8680295 DOI: 10.1002/anbr.202100047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Discovering new therapies to treat heart disease requires improved understanding of cardiac physiology at a cellular level. Extracellular vesicles (EVs) are plasma membrane-bound nano- and microparticles secreted by cells and known to play key roles in intercellular communication, often through transfer of biomolecular cargo. Advances in EV research have established techniques for EV isolation from tissue culture media or biofluids, as well as standards for quantitation and biomolecular characterization. EVs released by cardiac cells are known to be involved in regulating cardiac physiology as well as in the progression of myocardial diseases. Due to difficulty accessing the heart in vivo, advanced in vitro cardiac 'tissues-on-a-chip' have become a recent focus for studying EVs in the heart. These physiologically relevant models are producing new insight into the role of EVs in cardiac physiology and disease while providing a useful platform for screening novel EV-based therapeutics for cardiac tissue regeneration post-injury. Numerous hurdles have stalled the clinical translation of EV therapeutics for heart patients, but tissue-on-a-chip models are playing an important role in bridging the translational gap, improving mechanistic understanding of EV signalling in cardiac physiology, disease, and repair.
Collapse
Affiliation(s)
- Karl T Wagner
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| |
Collapse
|
38
|
Scott A, Sueiro Ballesteros L, Bradshaw M, Tsuji C, Power A, Lorriman J, Love J, Paul D, Herman A, Emanueli C, Richardson RJ. In Vivo Characterization of Endogenous Cardiovascular Extracellular Vesicles in Larval and Adult Zebrafish. Arterioscler Thromb Vasc Biol 2021; 41:2454-2468. [PMID: 34261327 PMCID: PMC8384253 DOI: 10.1161/atvbaha.121.316539] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/30/2021] [Indexed: 01/08/2023]
Abstract
Objective Extracellular vesicles (EVs) facilitate molecular transport across extracellular space, allowing local and systemic signaling during homeostasis and in disease. Extensive studies have described functional roles for EV populations, including during cardiovascular disease, but the in vivo characterization of endogenously produced EVs is still in its infancy. Because of their genetic tractability and live imaging amenability, zebrafish represent an ideal but under-used model to investigate endogenous EVs. We aimed to establish a transgenic zebrafish model to allow the in vivo identification, tracking, and extraction of endogenous EVs produced by different cell types. Approach and Results Using a membrane-tethered fluorophore reporter system, we show that EVs can be fluorescently labeled in larval and adult zebrafish and demonstrate that multiple cell types including endothelial cells and cardiomyocytes actively produce EVs in vivo. Cell-type specific EVs can be tracked by high spatiotemporal resolution light-sheet live imaging and modified flow cytometry methods allow these EVs to be further evaluated. Additionally, cryo electron microscopy reveals the full morphological diversity of larval and adult EVs. Importantly, we demonstrate the utility of this model by showing that different cell types exchange EVs in the adult heart and that ischemic injury models dynamically alter EV production. Conclusions We describe a powerful in vivo zebrafish model for the investigation of endogenous EVs in all aspects of cardiovascular biology and pathology. A cell membrane fluorophore labeling approach allows cell-type specific tracing of EV origin without bias toward the expression of individual protein markers and will allow detailed future examination of their function.
Collapse
Affiliation(s)
- Aaron Scott
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Lorena Sueiro Ballesteros
- Flow Cytometry Facility, Faculty of Biomedical Sciences (L.S.B., A.H.)
- Now with Charles River Laboratories, Discovery House, Quays Office Park, Conference Avenue, Portishead, Bristol, United Kingdom (L.S.B.)
| | - Marston Bradshaw
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Chisato Tsuji
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Ann Power
- BioEconomy Centre, The Henry Wellcome Building for Biocatalysis, Biosciences, University of Exeter, United Kingdom (A.P., J.L.)
| | - James Lorriman
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - John Love
- BioEconomy Centre, The Henry Wellcome Building for Biocatalysis, Biosciences, University of Exeter, United Kingdom (A.P., J.L.)
| | - Danielle Paul
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Andrew Herman
- Flow Cytometry Facility, Faculty of Biomedical Sciences (L.S.B., A.H.)
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Science (C.E.), University of Bristol, United Kingdom
- National Heart and Lung Institute, Imperial College London, United Kingdom (C.E.)
| | - Rebecca J. Richardson
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| |
Collapse
|
39
|
Wagner KT, Nash TR, Liu B, Vunjak-Novakovic G, Radisic M. Extracellular Vesicles in Cardiac Regeneration: Potential Applications for Tissues-on-a-Chip. Trends Biotechnol 2021; 39:755-773. [PMID: 32958383 PMCID: PMC7969481 DOI: 10.1016/j.tibtech.2020.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/26/2022]
Abstract
Strategies to regenerate cardiac tissue postinjury are limited and heart transplantation remains the only 'cure' for a failing heart. Extracellular vesicles (EVs), membrane-bound cell secretions important in intercellular signaling, have been shown to play a crucial role in regulating heart function. A mechanistic understanding of the role of EVs in the heart remains elusive due to the challenges in studying the native human heart. Tissue-on-a-chip platforms, comprising functional, physiologically relevant human tissue models, are an emerging technology that has yet to be fully applied to the study of EVs. In this review, we summarize recent advances in cardiac tissue-on-a-chip (CTC) platforms and discuss how they are uniquely situated to advance our understanding of EVs in cardiac disease and regeneration.
Collapse
Affiliation(s)
- Karl T Wagner
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Trevor R Nash
- Department of Medicine, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bohao Liu
- Department of Medicine, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
40
|
Tavakol DN, Fleischer S, Vunjak-Novakovic G. Harnessing organs-on-a-chip to model tissue regeneration. Cell Stem Cell 2021; 28:993-1015. [PMID: 34087161 DOI: 10.1016/j.stem.2021.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue engineering has markedly matured since its early beginnings in the 1980s. In addition to the original goal to regenerate damaged organs, the field has started to explore modeling of human physiology "in a dish." Induced pluripotent stem cell (iPSC) technologies now enable studies of organ regeneration and disease modeling in a patient-specific context. We discuss the potential of "organ-on-a-chip" systems to study regenerative therapies with focus on three distinct organ systems: cardiac, respiratory, and hematopoietic. We propose that the combinatorial studies of human tissues at these two scales would help realize the translational potential of tissue engineering.
Collapse
Affiliation(s)
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Medicine, Columbia University, New York, NY.
| |
Collapse
|
41
|
Akbar N, Paget D, Choudhury RP. Extracellular Vesicles in Innate Immune Cell Programming. Biomedicines 2021; 9:biomedicines9070713. [PMID: 34201592 PMCID: PMC8301301 DOI: 10.3390/biomedicines9070713] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EV) are a heterogeneous group of bilipid-enclosed envelopes that carry proteins, metabolites, RNA, DNA and lipids from their parent cell of origin. They mediate cellular communication to other cells in local tissue microenvironments and across organ systems. EV size, number and their biologically active cargo are often altered in response to pathological processes, including infection, cancer, cardiovascular diseases and in response to metabolic perturbations such as obesity and diabetes, which also have a strong inflammatory component. Here, we discuss the broad repertoire of EV produced by neutrophils, monocytes, macrophages, their precursor hematopoietic stem cells and discuss their effects on the innate immune system. We seek to understand the immunomodulatory properties of EV in cellular programming, which impacts innate immune cell differentiation and function. We further explore the possibilities of using EV as immune targeting vectors, for the modulation of the innate immune response, e.g., for tissue preservation during sterile injury such as myocardial infarction or to promote tissue resolution of inflammation and potentially tissue regeneration and repair.
Collapse
Affiliation(s)
- Naveed Akbar
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
- Correspondence:
| | - Daan Paget
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Robin P. Choudhury
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
| |
Collapse
|
42
|
Azzimato V, Jager J, Chen P, Morgantini C, Levi L, Barreby E, Sulen A, Oses C, Willerbrords J, Xu C, Li X, Shen JX, Akbar N, Haag L, Ellis E, Wålhen K, Näslund E, Thorell A, Choudhury RP, Lauschke VM, Rydén M, Craige SM, Aouadi M. Liver macrophages inhibit the endogenous antioxidant response in obesity-associated insulin resistance. Sci Transl Med 2021; 12:12/532/eaaw9709. [PMID: 32102936 DOI: 10.1126/scitranslmed.aaw9709] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/06/2020] [Indexed: 12/22/2022]
Abstract
Obesity and insulin resistance are risk factors for nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disease worldwide. Because no approved medication nor an accurate and noninvasive diagnosis is currently available for NAFLD, there is a clear need to better understand the link between obesity and NAFLD. Lipid accumulation during obesity is known to be associated with oxidative stress and inflammatory activation of liver macrophages (LMs). However, we show that although LMs do not become proinflammatory during obesity, they display signs of oxidative stress. In livers of both humans and mice, antioxidant nuclear factor erythroid 2-related factor 2 (NRF2) was down-regulated with obesity and insulin resistance, yielding an impaired response to lipid accumulation. At the molecular level, a microRNA-targeting NRF2 protein, miR-144, was elevated in the livers of obese insulin-resistant humans and mice, and specific silencing of miR-144 in murine and human LMs was sufficient to restore NRF2 protein expression and the antioxidant response. These results highlight the pathological role of LMs and their therapeutic potential to restore the impaired endogenous antioxidant response in obesity-associated NAFLD.
Collapse
Affiliation(s)
- Valerio Azzimato
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Jennifer Jager
- Université Côte d'Azur, Inserm U1065, C3M, Team Cellular and Molecular Physiopathology of Obesity, 06000 Nice, France
| | - Ping Chen
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Cecilia Morgantini
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Laura Levi
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Emelie Barreby
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - André Sulen
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Carolina Oses
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Joost Willerbrords
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Connie Xu
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Xidan Li
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Joanne X Shen
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Solna, Sweden
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DU Oxford, UK
| | - Lars Haag
- Department of Laboratory Medicine, Laboratory Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Ewa Ellis
- Division of Transplantation Surgery, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Kerstin Wålhen
- Unit of Endocrinology Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 182 88 Stockholm, Sweden
| | - Anders Thorell
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 182 88 Stockholm, Sweden.,Department of Surgery, Ersta Hospital, Karolinska Institutet, 116 28 Stockholm, Sweden
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DU Oxford, UK
| | - Volker M Lauschke
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Solna, Sweden
| | - Mikael Rydén
- Unit of Endocrinology Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Siobhan M Craige
- Human Nutrition, Food, and Exercise Department, Virginia Tech, Blacksburg, VA 24060, USA
| | - Myriam Aouadi
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden.
| |
Collapse
|
43
|
Sansonetti M, De Windt LJ. Non-coding RNAs in cardiac inflammation: key drivers in the pathophysiology of heart failure. Cardiovasc Res 2021; 118:2058-2073. [PMID: 34097013 DOI: 10.1093/cvr/cvab192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Heart failure is among the most progressive diseases and a leading cause of morbidity. Despite several advances in cardiovascular therapies, pharmacological treatments are limited to relieve symptoms without curing cardiac injury. Multiple observations point to the involvement of immune cells as key drivers in the pathophysiology of heart failure. In particular, there is a growing recognition that heart failure is related to a prolonged and insufficiently repressed inflammatory response leading to molecular, cellular, and functional cardiac alterations. Over the last decades, non-coding RNAs are recognized as prominent mediators of the cardiac inflammation, affecting the function of several immune cells. In the current review, we explore the contribution of the diverse immune cells in the progression of heart failure, revealing mechanistic functions for non-coding RNAs in cardiac immune cells as a new and exciting field of investigation.
Collapse
Affiliation(s)
- Marida Sansonetti
- Department of Molecular Genetics, Faculty of Science and Engineering; Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Leon J De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering; Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
44
|
Akawi N, Checa A, Antonopoulos AS, Akoumianakis I, Daskalaki E, Kotanidis CP, Kondo H, Lee K, Yesilyurt D, Badi I, Polkinghorne M, Akbar N, Lundgren J, Chuaiphichai S, Choudhury R, Neubauer S, Channon KM, Torekov SS, Wheelock CE, Antoniades C. Fat-Secreted Ceramides Regulate Vascular Redox State and Influence Outcomes in Patients With Cardiovascular Disease. J Am Coll Cardiol 2021; 77:2494-2513. [PMID: 34016263 PMCID: PMC8141611 DOI: 10.1016/j.jacc.2021.03.314] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Obesity is associated with increased cardiovascular risk; however, the potential role of dysregulations in the adipose tissue (AT) metabolome is unknown. OBJECTIVES The aim of this study was to explore the role of dysregulation in the AT metabolome on vascular redox signaling and cardiovascular outcomes. METHODS A screen was conducted for metabolites differentially secreted by thoracic AT (ThAT) and subcutaneous AT in obese patients with atherosclerosis (n = 48), and these metabolites were then linked with dysregulated vascular redox signaling in 633 patients undergoing coronary bypass surgery. The underlying mechanisms were explored in human aortic endothelial cells, and their clinical value was tested against hard clinical endpoints. RESULTS Because ThAT volume was associated significantly with arterial oxidative stress, there were significant differences in sphingolipid secretion between ThAT and subcutaneous AT, with C16:0-ceramide and derivatives being the most abundant species released within adipocyte-derived extracellular vesicles. High ThAT sphingolipid secretion was significantly associated with reduced endothelial nitric oxide bioavailability and increased superoxide generated in human vessels. Circulating C16:0-ceramide correlated positively with ThAT ceramides, dysregulated vascular redox signaling, and increased systemic inflammation in 633 patients with atherosclerosis. Exogenous C16:0-ceramide directly increased superoxide via tetrahydrobiopterin-mediated endothelial nitric oxide synthase uncoupling and dysregulated protein phosphatase 2 in human aortic endothelial cells. High plasma C16:0-ceramide and its glycosylated derivative were independently related with increased risk for cardiac mortality (adjusted hazard ratios: 1.394; 95% confidence interval: 1.030 to 1.886; p = 0.031 for C16:0-ceramide and 1.595; 95% confidence interval: 1.042 to 2.442; p = 0.032 for C16:0-glycosylceramide per 1 SD). In a randomized controlled clinical trial, 1-year treatment of obese patients with the glucagon-like peptide-1 analog liraglutide suppressed plasma C16:0-ceramide and C16:0-glycosylceramide changes compared with control subjects. CONCLUSIONS These results demonstrate for the first time in humans that AT-derived ceramides are modifiable regulators of vascular redox state in obesity, with a direct impact on cardiac mortality in advanced atherosclerosis. (The Interaction Between Appetite Hormones; NCT02094183).
Collapse
Affiliation(s)
- Nadia Akawi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates. https://twitter.com/NadiaAkawi
| | - Antonio Checa
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Alexios S Antonopoulos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Evangelia Daskalaki
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Christos P Kotanidis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hidekazu Kondo
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Kirsten Lee
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dilan Yesilyurt
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ileana Badi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Murray Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie Lundgren
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Robin Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Signe S Torekov
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
45
|
Sahoo S, Adamiak M, Mathiyalagan P, Kenneweg F, Kafert-Kasting S, Thum T. Therapeutic and Diagnostic Translation of Extracellular Vesicles in Cardiovascular Diseases: Roadmap to the Clinic. Circulation 2021; 143:1426-1449. [PMID: 33819075 PMCID: PMC8021236 DOI: 10.1161/circulationaha.120.049254] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exosomes are small membrane-bound vesicles of endocytic origin that are actively secreted. The potential of exosomes as effective communicators of biological signaling in myocardial function has previously been investigated, and a recent explosion in exosome research not only underscores their significance in cardiac physiology and pathology, but also draws attention to methodological limitations of studying these extracellular vesicles. In this review, we discuss recent advances and challenges in exosome research with an emphasis on scientific innovations in isolation, identification, and characterization methodologies, and we provide a comprehensive summary of web-based resources available in the field. Importantly, we focus on the biology and function of exosomes, highlighting their fundamental role in cardiovascular pathophysiology to further support potential applications of exosomes as biomarkers and therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Marta Adamiak
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Prabhu Mathiyalagan
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
| | - Sabine Kafert-Kasting
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- REBIRTH Center for Translational Regenerative Medicine (T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| |
Collapse
|
46
|
Abstract
Extracellular vesicles (EVs) have received considerable attention in biological and clinical research due to their ability to mediate cell-to-cell communication. Based on their size and secretory origin, EVs are categorized as exosomes, microvesicles, and apoptotic bodies. Increasing number of studies highlight the contribution of EVs in the regulation of a wide range of normal cellular physiological processes, including waste scavenging, cellular stress reduction, intercellular communication, immune regulation, and cellular homeostasis modulation. Altered circulating EV level, expression pattern, or content in plasma of patients with cardiovascular disease (CVD) may serve as diagnostic and prognostic biomarkers in diverse cardiovascular pathologies. Due to their inherent characteristics and physiological functions, EVs, in turn, have become potential candidates as therapeutic agents. In this review, we discuss the evolving understanding of the role of EVs in CVD, summarize the current knowledge of EV-mediated regulatory mechanisms, and highlight potential strategies for the diagnosis and therapy of CVD. We also attempt to look into the future that may advance our understanding of the role of EVs in the pathogenesis of CVD and provide novel insights into the field of translational medicine.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| | - Xue Zou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, PR China
| | - Pedro A Jose
- Division of Renal Disease & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, PR China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, PR China.
| |
Collapse
|
47
|
Abstract
Cardiovascular pathologies are often induced by inflammation. The associated changes in the inflammatory response influence vascular endothelial biology; they complicate the extent of ischaemia and reperfusion injury, direct the migration of immune competent cells and activate platelets. The initiation and progression of inflammation is regulated by the classical paradigm through the system of cytokines and chemokines. Therapeutic approaches have previously used this knowledge to control the extent of cardiovascular changes with varying degrees of success. Neuronal guidance proteins (NGPs) have emerged in recent years and have been shown to be significantly involved in the control of tissue inflammation and the mechanisms of immune cell activation. Therefore, proteins of this class might be used in the future as targets to control the extent of inflammation in the cardiovascular system. In this review, we describe the role of NGPs during cardiovascular inflammation and highlight potential therapeutic options that could be explored in the future.
Collapse
|
48
|
Aljabali A. Meet Our Editorial Board Member. Curr Drug Metab 2020. [DOI: 10.2174/138920022112201207105350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Rejuvenation of Senescent Endothelial Progenitor Cells by Extracellular Vesicles Derived From Mesenchymal Stromal Cells. JACC Basic Transl Sci 2020; 5:1127-1141. [PMID: 33294742 PMCID: PMC7691285 DOI: 10.1016/j.jacbts.2020.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 02/08/2023]
Abstract
EVs derived from young, but not aged, MSCs rejuvenate senescent EPCs in vitro, recapitulating the effect of MSC transplantation. Aged MSCs can be genetically modified to produce tailored EVs with increased EPC rejuvenation capacity in vitro and increased angiogenesis capacity following ischemic event in vivo. EVs represent a promising platform to develop an acellular therapeutic approach in regenerative medicine for cardiovascular diseases.
Mesenchymal stromal cell (MSC) transplantation is a form of the stem-cell therapy that has shown beneficial effects for many diseases. The use of stem-cell therapy, including MSC transplantation, however, has limitations such as the tumorigenic potential of stem cells and the lack of efficacy of aged autologous cells. An ideal therapeutic approach would keep the beneficial effects of MSC transplantation while circumventing the limitations associated with the use of intact stem cells. This study provides proof-of-concept evidence that MSC-derived extracellular vesicles represent a promising platform to develop an acellular therapeutic approach that would just do that. Extracellular vesicles are membranous vesicles secreted by MSCs and contain bioactive molecules to mediate communication between different cells. Extracellular vesicles can be taken up by recipient cells, and once inside the recipient cells, the bioactive molecules are released to exert the beneficial effects on the recipient cells. This study, for the first time to our knowledge, shows that extracellular vesicles secreted by MSCs recapitulate the beneficial effects of MSCs on vascular repair and promote blood vessel regeneration after ischemic events. Furthermore, MSCs from aged donors can be engineered to produce extracellular vesicles with improved regenerative potential, comparable to MSCs from young donors, thus eliminating the need for allogenic young donors for elderly patients.
Collapse
Key Words
- BM, bone marrow
- CVD, cardiovascular disease
- EC, endothelial cell
- EPC, endothelial progenitor cell
- EV, extracellular vesicle
- FBS, fetal bovine serum
- MEM, minimum essential medium
- MI, myocardial infarction
- MSC, mesenchymal stromal cell
- NTA, nanotracking analysis
- PBS, phosphate-buffered saline
- TEV, tailored extracellular vesicle
- VEGF, vascular endothelial growth factor
- acellular
- angiogenesis
- extracellular vesicles
- lin− BMC, lineage negative bone marrow cell
- miR, microRNA
- qPCR, quantitative transcription polymerase chain reaction
- regeneration
- senescence
Collapse
|
50
|
Braun F, Rinschen M, Buchner D, Bohl K, Plagmann I, Bachurski D, Richard Späth M, Antczak P, Göbel H, Klein C, Lackmann J, Kretz O, Puelles VG, Wahba R, Hallek M, Schermer B, Benzing T, Huber TB, Beyer A, Stippel D, Kurschat CE, Müller R. The proteomic landscape of small urinary extracellular vesicles during kidney transplantation. J Extracell Vesicles 2020; 10:e12026. [PMID: 33304478 PMCID: PMC7710132 DOI: 10.1002/jev2.12026] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 09/05/2020] [Accepted: 10/10/2020] [Indexed: 12/24/2022] Open
Abstract
Kidney transplantation is the preferred renal replacement therapy available. Yet, long-term transplant survival is unsatisfactory, partially due to insufficient possibilities of longitudinal monitoring and understanding of the biological processes after transplantation. Small urinary extracellular vesicles (suEVs) - as a non-invasive source of information - were collected from 22 living donors and recipients. Unbiased proteomic analysis revealed temporal patterns of suEV protein signature and cellular processes involved in both early response and longer-term graft adaptation. Complement activation was among the most dynamically regulated components. This unique atlas of the suEV proteome is provided through an online repository allowing dynamic interrogation by the user. Additionally, a correlative analysis identified putative prognostic markers of future allograft function. One of these markers - phosphoenol pyruvate carboxykinase (PCK2) - could be confirmed using targeted MS in an independent validation cohort of 22 additional patients. This study sheds light on the impact of kidney transplantation on urinary extracellular vesicle content and allows the first deduction of early molecular processes in transplant biology. Beyond that our data highlight the potential of suEVs as a source of biomarkers in this setting.
Collapse
|