1
|
Feng Y, Wang Y, Li L, Yang Y, Tan X, Chen T. Exosomes Induce Crosstalk Between Multiple Types of Cells and Cardiac Fibroblasts: Therapeutic Potential for Remodeling After Myocardial Infarction. Int J Nanomedicine 2024; 19:10605-10621. [PMID: 39445157 PMCID: PMC11498042 DOI: 10.2147/ijn.s476995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Recanalization therapy can significantly improve the prognosis of patients with acute myocardial infarction (AMI). However, infarction or reperfusion-induced cardiomyocyte death, immune cell infiltration, fibroblast proliferation, and scarring formation lead to cardiac remodeling and gradually progress to heart failure or arrhythmia, resulting in a high mortality rate. Due to the inability of cardiomyocytes to regenerate, the healing of infarcted myocardium mainly relies on the formation of scars. Cardiac fibroblasts, as the main effector cells involved in repair and scar formation, play a crucial role in maintaining the structural integrity of the heart after MI. Recent studies have revealed that exosome-mediated intercellular communication plays a huge role in myocardial repair and signaling transduction after myocardial infarction (MI). Exosomes can regulate the biological behavior of fibroblasts by activating or inhibiting the intracellular signaling pathways through their contents, which are derived from cardiomyocytes, immune cells, endothelial cells, mesenchymal cells, and others. Understanding the interactions between fibroblasts and other cell types during cardiac remodeling will be the key to breakthrough therapies. This review examines the role of exosomes from different sources in the repair process after MI injury, especially the impacts on fibroblasts during myocardial remodeling, and explores the use of exosomes in the treatment of myocardial remodeling after MI.
Collapse
Affiliation(s)
- Yijuan Feng
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Li Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
2
|
Gu Y, Feng J, Shi J, Xiao G, Zhang W, Shao S, Liu B, Guo H. Global Research Trends on Exosome in Cardiovascular Diseases: A Bibliometric-Based Visual Analysis. Vasc Health Risk Manag 2024; 20:377-402. [PMID: 39188326 PMCID: PMC11346494 DOI: 10.2147/vhrm.s473520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024] Open
Abstract
Background Exosomes in cardiovascular diseases (CVDs) have attracted huge attention with substantial value and potential. Our bibliometrics is based on literature from the field of cardiovascular exosomes over the past 30 years, which has been visualized to display the development process, research hotspots, and cutting-edge trends of clinical practices, mechanisms, and management strategies related to psych cardiology. Methods We selected articles and reviews on exosomes in CVDs from the core collection of Web of Science, and generated visual charts by using CiteSpace and VOSviewer software. Results Our research included 1613 publications. The number of exosome articles in CVD fluctuates slightly, but overall shows an increasing trend. The main research institutions were Tongji University and Nanjing Medical University. The International Journal of Molecular Sciences has the highest publication volume, while the Journal of Cellular and Molecular Medicine has the highest citation count. Among all the authors, Eduardo Marban ranks first in terms of publication volume and H-index. The most common keywords are exosome, extracellular vesicles, and angiogenesis. Conclusion This is a bibliometric study on the research hotspots and trends of exosomes in CVD. Exosome research in the field of cardiovascular medicine is on the rise. Some exosome treatment methods may become the focus of future research.
Collapse
Affiliation(s)
- Yunxiao Gu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiayi Shi
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Guanyi Xiao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Weiwei Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Shuijin Shao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Haidong Guo
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Zhou W, Li H, Song J, Suo F, Gu M, Qi S. Healthy Plasma Exosomes Exert Potential Neuroprotective Effects against Methylmalonic Acid-Induced Hippocampal Neuron Injury. ACS Chem Neurosci 2024; 15:3022-3033. [PMID: 39026168 DOI: 10.1021/acschemneuro.4c00224] [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] [Indexed: 07/20/2024] Open
Abstract
Exosomes have shown good potential for alleviating neurological deficits and delaying memory deterioration, but the neuroprotective effects of exosomes remain unknown. Methylmalonic acidemia is a metabolic disorder characterized by the accumulation of methylmalonic acid (MMA) in various tissues that inhibits neuronal survival and function, leading to accelerated neurological deterioration. Effective therapies to mitigate these symptoms are lacking. The purpose of this study was to explore the neuroprotective effects of plasma exosomes on cells and a mouse model of MMA-induced injury. We evaluated the ability of plasma exosomes to reduce the neuronal apoptosis, cross the blood-brain barrier, and affect various parameters related to neuronal function. MMA promoted cell apoptosis, disrupted the metabolic balance, and altered the expression of B-cell lymphoma-2 (Bcl-2), Bcl2-associated X (Bax), and synaptophysin-1 (Syp-1), and these changes may be involved in MMA-induced neuronal apoptosis. Additionally, plasma exosomes normalized learning and memory and protected against MMA-induced neuronal apoptosis. Our findings indicate that neurological deficits are linked to the pathogenesis of methylmalonic acidemia, and healthy plasma exosomes may exert neuroprotective and therapeutic effects by altering the expression of exosomal microRNAs, facilitating neuronal functional recovery in the context of this inherited metabolic disease. Intravenous plasma-derived exosome treatment may be a novel clinical therapeutic strategy for methylmalonic acidemia.
Collapse
Affiliation(s)
- Wei Zhou
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou 221004, P.R China
- Newborn Screening Center, The Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou 221009, P.R China
| | - Huizhong Li
- Newborn Screening Center, The Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou 221009, P.R China
| | - Jinxiu Song
- Pharmacology College, Xuzhou Medical University, Xuzhou 221004, P.R China
| | - Feng Suo
- Newborn Screening Center, The Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou 221009, P.R China
| | - Maosheng Gu
- Newborn Screening Center, The Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou 221009, P.R China
| | - Suhua Qi
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou 221004, P.R China
- Pharmacology College, Xuzhou Medical University, Xuzhou 221004, P.R China
- Medical and Technology School, Xuzhou Medical University, Xuzhou 221004, P.R China
| |
Collapse
|
4
|
Rivera J, Sharma B, Torres MM, Kumar S. Factors affecting the GABAergic synapse function in Alzheimer's disease: Focus on microRNAs. Ageing Res Rev 2023; 92:102123. [PMID: 37967653 DOI: 10.1016/j.arr.2023.102123] [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: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurological disease characterized by the loss of cognitive function, confusion, and memory deficit. Accumulation of abnormal proteins, amyloid beta (Aß), and phosphorylated Tau (p-tau) forms plaques and tangles that deteriorate synapse function, resulting in neurodegeneration and cognitive decline in AD. The human brain is composed of different types of neurons and/or synapses that are functionally defective in AD. The GABAergic synapse, the most abundant inhibitory neuron in the human brain was found to be dysfunctional in AD and contributes to disrupting neurological function. This study explored the types of GABA receptors associated with neurological dysfunction and various biological and environmental factors that cause GABAergic neuron dysfunction in AD, such as Aβ, p-tau, aging, sex, astrocytes, microglia, APOE, mental disorder, diet, physical activity, and sleep. Furthermore, we explored the role of microRNAs (miRNAs) in the regulation of GABAergic synapse function in neurological disorders and AD states. We also discuss the molecular mechanisms underlying GABAergic synapse dysfunction with a focus on miR-27b, miR-30a, miR-190a/b, miR-33, miR-51, miR-129-5p, miR-376-3p, miR-376c, miR-30b and miR-502-3p. The purpose of our article is to highlight the recent research on miRNAs affecting the regulation of GABAergic synapse function and factors that contribute to the progression of AD.
Collapse
Affiliation(s)
- Jazmin Rivera
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Bhupender Sharma
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Melissa M Torres
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Subodh Kumar
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA; L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| |
Collapse
|
5
|
Harbuz I, Banciu DD, David R, Cercel C, Cotîrță O, Ciurea BM, Radu SM, Dinescu S, Jinga SI, Banciu A. Perspectives on Scaffold Designs with Roles in Liver Cell Asymmetry and Medical and Industrial Applications by Using a New Type of Specialized 3D Bioprinter. Int J Mol Sci 2023; 24:14722. [PMID: 37834167 PMCID: PMC10573170 DOI: 10.3390/ijms241914722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cellular asymmetry is an important element of efficiency in the compartmentalization of intracellular chemical reactions that ensure efficient tissue function. Improving the current 3D printing methods by using cellular asymmetry is essential in producing complex tissues and organs such as the liver. The use of cell spots containing at least two cells and basement membrane-like bio support materials allows cells to be tethered at two points on the basement membrane and with another cell in order to maintain cell asymmetry. Our model is a new type of 3D bioprinter that uses oriented multicellular complexes with cellular asymmetry. This novel approach is necessary to replace the sequential and slow processes of organogenesis with rapid methods of growth and 3D organ printing. The use of the extracellular matrix in the process of bioprinting with cells allows one to preserve the cellular asymmetry in the 3D printing process and thus preserve the compartmentalization of biological processes and metabolic efficiency.
Collapse
Affiliation(s)
- Iuliana Harbuz
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Daniel Dumitru Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Rodica David
- Institute for Research on the Quality of Society and the Sciences of Education, University Constantin Brancusi of Targu Jiu, Republicii 1, 210185 Targu Jiu, Romania;
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Cristina Cercel
- University of Medicine and Pharmacy “Carol Davila” Bucharest, 37 Dionisie Lupu Street, 020021 Bucharest, Romania;
| | - Octavian Cotîrță
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Bogdan Marius Ciurea
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Sorin Mihai Radu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Stela Dinescu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Sorin Ion Jinga
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Adela Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| |
Collapse
|
6
|
Jiang J, Ni L, Zhang X, Wang H, Liu L, Wei M, Li G, Bei Y. Platelet Membrane-Fused Circulating Extracellular Vesicles Protect the Heart from Ischemia/Reperfusion Injury. Adv Healthc Mater 2023; 12:e2300052. [PMID: 37097199 DOI: 10.1002/adhm.202300052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/10/2023] [Indexed: 04/26/2023]
Abstract
Myocardial ischemia/reperfusion injury (I/RI) may potentiate cardiac remodeling and heart failure, while effective therapies for I/RI remain lacking. Circulating human plasma-derived extracellular vesicles (hEV) have great potential to protect against I/RI. However, the effective delivery of hEV in vivo remains a limiting factor for clinical application. The present study constructs a biomimetic delivery system of platelet membrane-fused hEV (P-hEV), utilizing the natural affinity of platelets for hEV delivery to the injured vascular and myocardial sites. The results show that platelet membrane and hEV membrane fusion can be achieved through repeated extrusion. Compared to non-modified hEV, P-hEV uptake is greatly enhanced in human umbilical vein endothelial cells (HUVECs) stressed by oxygen-glucose deprivation/reperfusion (OGD/R). Functionally, P-hEV inhibits HUVEC and neonatal rat cardiomyocyte (NRCM) apoptosis and promotes HUVECs migration and tube formation under OGD/R stress in vitro. Intravenous delivery of P-hEV more effectively targets and accumulates at injury sites in the heart. Furthermore, P-hEV significantly enhances protection against acute I/RI and attenuates cardiac remodeling at three weeks post-I/RI. In conclusion, the platelet membrane-fused hEV delivery system enhances the target delivery of EV to protect against myocardial I/RI, presenting a novel drug delivery system for ischemic heart diseases.
Collapse
Affiliation(s)
- Jizong Jiang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Lingyan Ni
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Xinxin Zhang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Hongyun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Li Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Meng Wei
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Guoping Li
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yihua Bei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
7
|
Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer. Molecules 2023; 28:molecules28041802. [PMID: 36838790 PMCID: PMC9967195 DOI: 10.3390/molecules28041802] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/23/2022] [Accepted: 01/12/2023] [Indexed: 02/17/2023] Open
Abstract
Triple-negative breast cancer is the most potent metastatic type of breast cancer that can spread to other body parts. Chemotherapy and surgical intervention are the sole treatments for TNBC, owing to the scarcity of therapeutic targets. Manipulation of the membranes as per the desired targets of exosomes has recently gained much attention as a drug delivery method. Despite their known roles in different diseases, very few studies have focused on signalling that triggers the metastasis of triple-negative breast cancer to other body parts by exosomes. This article highlights the significant roles of exosomes associated with TNBC, the involvement of exosomes in breast cancer diagnosis, progression, and the treatment of triple-negative breast cancer by the exosomes as a drug delivery system. This review paper also illustrates the role of exosomes in initiating EMT in breast cancer, including novel signalling.
Collapse
|
8
|
Xu J, Wang W, Wang Y, Zhu Z, Li D, Wang T, Liu K. Progress in research on the role of exosomal miRNAs in the diagnosis and treatment of cardiovascular diseases. Front Genet 2022; 13:929231. [PMID: 36267409 PMCID: PMC9577319 DOI: 10.3389/fgene.2022.929231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022] Open
Abstract
Cardiovascular diseases are the most common diseases threatening the health of the elderly, and the incidence and mortality rates associated with cardiovascular diseases remain high and are increasing gradually. Studies on the treatment and prevention of cardiovascular diseases are underway. Currently, several research groups are studying the role of exosomes and biomolecules incorporated by exosomes in the prevention, diagnosis, and treatment of clinical diseases, including cardiovascular diseases. Now, based on the results of published studies, this review discusses the characteristics, separation, extraction, and identification of exosomes, specifically the role of exosomal miRNAs in atherosclerosis, myocardial injury and infarction, heart failure, aortic dissection, myocardial fibrosis, ischemic reperfusion, atrial fibrillation, and other diseases. We believe that the observations noted in this article will aid in the prevention, diagnosis, and treatment of cardiovascular diseases.
Collapse
|
9
|
Han C, Yang J, Zhang E, Jiang Y, Qiao A, Du Y, Zhang Q, An J, Sun J, Wang M, Nguyen T, Lal H, Krishnamurthy P, Zhang J, Qin G. Metabolic labeling of cardiomyocyte-derived small extracellular-vesicle (sEV) miRNAs identifies miR-208a in cardiac regulation of lung gene expression. J Extracell Vesicles 2022; 11:e12246. [PMID: 36250966 PMCID: PMC9575700 DOI: 10.1002/jev2.12246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/06/2022] Open
Abstract
Toxoplasma gondii uracil phosphoribosyltransferase (UPRT) converts 4-thiouracil (4TUc) into 4-thiouridine (4TUd), which is incorporated into nascent RNAs and can be biotinylated, then labelled with streptavidin conjugates or isolated via streptavidin-affinity methods. Here, we generated mice that expressed T. gondii UPRT only in cardiomyocytes (CM UPRT mice) and tested our hypothesis that CM-derived miRNAs (CM miRs) are transferred into remote organs after myocardial infarction (MI) by small extracellular vesicles (sEV) that are released from the heart into the peripheral blood (PB sEV). We found that 4TUd was incorporated with high specificity and sensitivity into RNAs isolated from the hearts and PB sEV of CM UPRT mice 6 h after 4TUc injection. In PB sEV, 4TUd was incorporated into CM-specific/enriched miRs including miR-208a, but not into miRs with other organ or tissue-type specificities. 4TUd-labelled miR208a was also present in lung tissues, especially lung endothelial cells (ECs), and CM-derived miR-208a (CM miR-208a) levels peaked 12 h after experimentally induced MI in PB sEV and 24 h after MI in the lung. Notably, miR-208a is expressed from intron 29 of α myosin heavy chain (αMHC), but αMHC transcripts were nearly undetectable in the lung. When PB sEV from mice that underwent MI (MI-PB sEV) or sham surgery (Sham-PB sEV) were injected into intact mice, the expression of Tmbim6 and NLK, which are suppressed by miR-208a and cooperatively regulate inflammation via the NF-κB pathway, was lower in the lungs of MI-PB sEV-treated animals than the lungs of animals treated with Sham-PB sEV or saline. In MI mice, Tmbim6 and NLK were downregulated, whereas endothelial adhesion molecules and pro-inflammatory cells were upregulated in the lung; these changes were significantly attenuated when the mice were treated with miR-208a antagomirs prior to MI surgery. Thus, CM UPRT mice enables us to track PB sEV-mediated transport of CM miRs and identify an miR-208a-mediated mechanism by which myocardial injury alters the expression of genes and inflammatory response in the lung.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Junjie Yang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Eric Zhang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Ying Jiang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Aijun Qiao
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yipeng Du
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Qinkun Zhang
- Department of MedicineDivision of Cardiovascular DiseaseSchool of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Junqing An
- Center for Molecular and Translational MedicineGeorgia State UniversityAtlantaGeorgiaUSA
| | - Jiacheng Sun
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Meimei Wang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Thanh Nguyen
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Hind Lal
- Department of MedicineDivision of Cardiovascular DiseaseSchool of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Prasanna Krishnamurthy
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jianyi Zhang
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Gangjian Qin
- Department of Biomedical EngineeringSchool of Medicine and School of EngineeringUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
10
|
Meng L, Song K, Li S, Kang Y. Exosomes: Small Vesicles with Important Roles in the Development, Metastasis and Treatment of Breast Cancer. MEMBRANES 2022; 12:membranes12080775. [PMID: 36005690 PMCID: PMC9414313 DOI: 10.3390/membranes12080775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 05/12/2023]
Abstract
Breast cancer (BC) has now overtaken lung cancer as the most common cancer, while no biopredictive marker isolated from biological fluids has yet emerged clinically. After traditional chemotherapy, with the huge side effects brought by drugs, patients also suffer from the double affliction of drugs to the body while fighting cancer, and they often quickly develop drug resistance after the drug, leading to a poor prognosis. And the treatment of some breast cancer subtypes, such as triple negative breast cancer (TNBC), is even more difficult. Exosomes (Exos), which are naturally occurring extracellular vesicles (EVs) with nanoscale acellular structures ranging in diameter from 40 to 160 nm, can be isolated from various biological fluids and have been widely studied because they are derived from the cell membrane, have extremely small diameter, and are widely involved in various biological activities of the body. It can be used directly or modified to make derivatives or to make some analogs for the treatment of breast cancer. This review will focus on the involvement of exosomes in breast cancer initiation, progression, invasion as well as metastasis and the therapeutic role of exosomes in breast cancer.
Collapse
Affiliation(s)
- Ling’ao Meng
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| |
Collapse
|
11
|
Zhou D, Dai Z, Ren M, Yang M. Adipose-Derived Stem Cells-Derived Exosomes with High Amounts of Circ_0001747 Alleviate Hypoxia/Reoxygenation-Induced Injury in Myocardial Cells by Targeting MiR-199b-3p/MCL1 Axis. Int Heart J 2022; 63:356-366. [PMID: 35354755 DOI: 10.1536/ihj.21-441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent studies demonstrated that circular RNAs play important roles in exosome-mediated cardio-protective effects after acute myocardial infarction (AMI). A previous study reported that circ_0001747 level is down-regulated in mouse hypoxia/reoxygenation (H/R) injury model. However, its biological role and working mechanism in AMI remain largely unknown.Exosomes were isolated from the culture supernatant of adipose-derived stem cells (ADSCs) using an ExoQuick precipitation kit. We treated mouse myocardial cells HL-1 with H/R to explore the role of exosomal circ_0001747 in AMI pathology. Cell viability, proliferation, apoptosis, and inflammation were analyzed by Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine assay, flow cytometry, and enzyme-linked immunosorbent assay. Dual-luciferase reporter assay and RNA immunoprecipitation assay were conducted to confirm the interaction between microRNA-199b-3p (miR-199b-3p) and circ_0001747 or MCL1 apoptosis regulator, BCL2 family member (MCL1).H/R-induced HL-1 dysfunction was attenuated by the incubation of exosomes derived from ADSCs, especially the exosomes with high amounts of circ_0001747. Circ_0001747 directly targeted miR-199b-3p in HL-1 cells. miR-199b-3p overexpression partly overturned exosomal circ_0001747-mediated protective effects in H/R-induced HL-1 cells. MCL1 was a direct target of miR-199b-3p in HL-1 cells. miR-199b-3p silencing alleviated H/R-induced damage in HL-1 cells partly by up-regulating MCL1. Circ_0001747 can elevate the messenger RNA and protein levels of MCL1 by sequestering miR-199b-3p.Overall, these results indicated that ADSCs-derived exosomes with high amounts of circ_0001747 attenuated H/R-induced HL-1 dysfunction partly by targeting miR-199b-3p/MCL1 signaling.
Collapse
Affiliation(s)
- Duohui Zhou
- Department of Cardiology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital
| | - Zhongli Dai
- Department of Cardiology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital
| | - Mingde Ren
- Department of Cardiology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital
| | | |
Collapse
|
12
|
Huang LY, Song JX, Cai H, Wang PP, Yin QL, Zhang YD, Chen J, Li M, Song JJ, Wang YL, Luo L, Wang W, Qi SH. Healthy Serum-Derived Exosomes Improve Neurological Outcomes and Protect Blood–Brain Barrier by Inhibiting Endothelial Cell Apoptosis and Reversing Autophagy-Mediated Tight Junction Protein Reduction in Rat Stroke Model. Front Cell Neurosci 2022; 16:841544. [PMID: 35308117 PMCID: PMC8927286 DOI: 10.3389/fncel.2022.841544] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2022] [Indexed: 02/03/2023] Open
Abstract
Blood–brain barrier (BBB) dysfunction causing edema and hemorrhagic transformation is one of the pathophysiological characteristics of stroke. Protection of BBB integrity has shown great potential in improving stroke outcome. Here, we assessed the efficacy of exosomes extracted from healthy rat serum in protection against ischemic stroke in vivo and in vitro. Exosomes were isolated by gradient centrifugation and ultracentrifugation and exosomes were characterized by transmission electron microscopy (TEM) and nanoparticle tracking video microscope. Exosomes were applied to middle cerebral artery occlusion (MCAO) rats or brain microvascular endothelial cell line (bEnd.3) subjected to oxygen-glucose deprivation (OGD) injury. Serum-derived exosomes were injected intravenously into adult male rats 2 h after transient MCAO. Infarct volume and gross cognitive function were assessed 24 h after reperfusion. Poststroke rats treated with serum-derived exosomes exhibited significantly reduced infarct volumes and enhanced neurological function. Apoptosis was assessed via terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) staining and the expression of B-cell lymphoma-2 (Bcl-2), Bax, and cleaved caspase-3 24 h after injury. Our data showed that serum exosomes treatment strikingly decreased TUNEL+ cells in the striatum, enhanced the ratio of Bcl-2 to Bax, and inhibited cleaved caspase-3 production in MCAO rats and OGD/reoxygenation insulted bEnd.3 cells. Under the consistent treatment, the expression of microtubule-associated protein 1 light chain 3B-II (LC3B-II), LC3B-I, and Sequestosome-1 (SQSTM1)/p62 was detected by Western blotting. Autolysosomes were observed via TEM. We found that serum exosomes reversed the ratio of LC3B-II to LC3B-I, prevented SQSTM1/p62 degradation, autolysosome formation, and autophagic flux. Together, these results indicated that exosomes isolated from healthy serum provided neuroprotection against experimental stroke partially via inhibition of endothelial cell apoptosis and autophagy-mediated BBB breakdown. Intravenous serum-derived exosome treatment may, therefore, provide a novel clinical therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Lin-Yan Huang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Jin-Xiu Song
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Heng Cai
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Pei-Pei Wang
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Qi-Long Yin
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Yi-De Zhang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Jie Chen
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Ming Li
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Jia-Jia Song
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Yan-Ling Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Lan Luo
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Wan Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Su-Hua Qi
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Su-Hua Qi,
| |
Collapse
|
13
|
Huang L, Shi Y, Li M, Wang T, Zhao L. Plasma Exosomes Loaded pH-Responsive Carboxymethylcellulose Hydrogel Promotes Wound Repair by Activating the Vascular Endothelial Growth Factor Signaling Pathway in Type 1 Diabetic Mice. J Biomed Nanotechnol 2021; 17:2021-2033. [PMID: 34706802 DOI: 10.1166/jbn.2021.3165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chronic wound healing plagues thousands of diabetic patients and brings social and economic burdens. Plasma exosomes (P-Exos), regarded as nanosized therapeutic agents, have shown therapeutic efficacy in promoting diabetic wound healing. The present work prepared the P-Exos-loaded pH-responsive carboxymethylcellulose (P-Exos-loaded CMC) hydrogel to investigate its ability to accelerate diabetic wound healing and to explore its underlying mechanisms. The results showed that the P-Exos-loaded CMC hydrogel was an effective therapeutic agent for accelerating diabetic wound repair. It promoted the local wound healing process in diabetic type 1 mice and enhanced angiogenesis and re-epithelialization via activating angiogenesis-related pathways mediated by vascular endothelial growth factor (VEGF).
Collapse
Affiliation(s)
- Lijuan Huang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Mengdie Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Tao Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| |
Collapse
|
14
|
Li B, Cai X, Wang Y, Zhu H, Zhang P, Jiang P, Yang X, Sun J, Hong L, Shao L. Circ-SKA3 Enhances Doxorubicin Toxicity in AC16 Cells Through miR-1303/TLR4 Axis. Int Heart J 2021; 62:1112-1123. [PMID: 34544967 DOI: 10.1536/ihj.20-809] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Doxorubicin (DOX) is a widely used anticancer drug, but its cardiotoxicity largely limits its clinical utilization. Circular RNA spindle and kinetochore-associated protein 3 (circ-SKA3) were found to be differentially expressed in heart failure patients. In this study, we investigated the role and mechanism of circ-SKA3 in DOX-induced cardiotoxicity.The quantitative real-time polymerase chain reaction and western blot assays were applied to measure the expression of circ-SKA3, microRNA (miR) -1303, and toll-like receptor 4 (TLR4). The viability and apoptosis of AC16 cells were analyzed using cell counting kit-8, flow cytometry, and western blot assays. The interaction between miR-1303 and circ-SKA3 or TLR4 was verified using dual-luciferase reporter and RNA immunoprecipitation assays. Exosomes were collected from culture media by the use of commercial kits and then qualified by transmission electron microscopy.The expression of circ-SKA3 and TLR4 was increased, whereas miR-1303 expression was decreased in DOX-treated AC16 cells. DOX treatment promoted cell apoptosis and inhibited cell viability in AC16 cells in vitro, which was partially reversed by circ-SKA3 knockdown, TLR4 silencing, or miR-1303 overexpression. Mechanistically, circ-SKA3 served as a sponge for miR-1303 to upregulate TLR4, which was confirmed to be a target of miR-1303. Additionally, circ-SKA3 contributed to DOX-induced cardiotoxicity through the miR-1303/TLR4 axis. Further studies suggested that circ-SKA3 was overexpressed in exosomes extracted from DOX-mediated AC16 cells, which could be internalized by surrounding untreated AC16 cells.Circ-SKA3 enhanced DOX-induced toxicity in AC16 cells through the miR-1303/TLR4 axis. Extracellular circ-SKA3 was packaged into exosomes, and exosomal circ-SKA3 could function as a mediator in intercellular communication between AC16 cells.
Collapse
Affiliation(s)
- Bin Li
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | - Yunxia Wang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | - Hongmin Zhu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | | | - Xu Yang
- Shenzhen Realomics (Biotech), Co. Ltd
| | - Jianhua Sun
- Department of Cardiology, The People's Hospital of Yudu County
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University
| |
Collapse
|
15
|
Isaac R, Reis FCG, Ying W, Olefsky JM. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab 2021; 33:1744-1762. [PMID: 34496230 PMCID: PMC8428804 DOI: 10.1016/j.cmet.2021.08.006] [Citation(s) in RCA: 339] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/07/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
Exosomes are nanoparticles secreted by all cell types and are a large component of the broader class of nanoparticles termed extracellular vesicles (EVs). Once secreted, exosomes gain access to the interstitial space and ultimately the circulation, where they exert local paracrine or distal systemic effects. Because of this, exosomes are important components of an intercellular and intraorgan communication system capable of carrying biologic signals from one cell type or tissue to another. The exosomal cargo consists of proteins, lipids, miRNAs, and other RNA species, and many of the biologic effects of exosomes have been attributed to miRNAs. Exosomal miRNAs have also been used as disease biomarkers. The field of exosome biology and metabolism is rapidly expanding, with new discoveries and reports appearing on a regular basis, and it is possible that potential therapeutic approaches for the use of exosomes or miRNAs in metabolic diseases will be initiated in the near future.
Collapse
Affiliation(s)
- Roi Isaac
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Felipe Castellani Gomes Reis
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Wei Ying
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
16
|
Das Gupta A, Krawczynska N, Nelson ER. Extracellular Vesicles-The Next Frontier in Endocrinology. Endocrinology 2021; 162:6310412. [PMID: 34180968 PMCID: PMC8294678 DOI: 10.1210/endocr/bqab133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs), including exosomes, are emerging as important carriers of signals in normal and pathological physiology. As EVs are a long-range communication or signaling modality-just like hormones are-the field of endocrinology is uniquely poised to offer insight into their functional biology and regulation. EVs are membrane-bound particles secreted by many different cell types and can have local or systemic effects, being transported in body fluids. They express transmembrane proteins, some of which are shared between EVs and some being specific to the tissue of origin, that can interact with target cells directly (much like hormones can). They also contain cargo within them that includes DNA, RNA, miRNA, and various metabolites. They can fuse with target cells to empty their cargo and alter their target cell physiology in this way also. Similar to the endocrine system, the EV system is likely to be under homeostatic control, making the regulation of their biogenesis and secretion important aspects to study. In this review, we briefly highlight select examples of how EVs are implicated in normal physiology and disease states. We also discuss what is known about their biogenesis and regulation of secretion. We hope that this paper inspires the endocrinology field to use our collective expertise to explore these new multimodal "hormones."
Collapse
Affiliation(s)
- Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence: Erik R. Nelson, Ph.D., University of Illinois at Urbana-Champaign. 407 S Goodwin Ave (MC-114), Urbana, IL, 61801, USA.
| |
Collapse
|
17
|
Pulmonary arterial hypertension induces the release of circulating extracellular vesicles with oxidative content and alters redox and mitochondrial homeostasis in the brains of rats. Hypertens Res 2021; 44:918-931. [PMID: 33875858 DOI: 10.1038/s41440-021-00660-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 02/03/2023]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by increased resistance of the pulmonary vasculature and afterload imposed on the right ventricle (RV). Two major contributors to the worsening of this disease are oxidative stress and mitochondrial impairment. This study aimed to explore the effects of monocrotaline (MCT)-induced PAH on redox and mitochondrial homeostasis in the RV and brain and how circulating extracellular vesicle (EV) signaling is related to these phenomena. Wistar rats were divided into control and MCT groups (60 mg/kg, intraperitoneal), and EVs were isolated from blood on the day of euthanasia (21 days after MCT injections). There was an oxidative imbalance in the RV, brain, and EVs of MCT rats. PAH impaired mitochondrial function in the RV, as seen by a decrease in the activities of mitochondrial complex II and citrate synthase and manganese superoxide dismutase (MnSOD) protein expression, but this function was preserved in the brain. The key regulators of mitochondrial biogenesis, namely, proliferator-activated receptor gamma coactivator 1-alpha and sirtuin 1, were poorly expressed in the EVs of MCT rats, and this result was positively correlated with MnSOD expression in the RV and negatively correlated with MnSOD expression in the brain. Based on these findings, we can conclude that the RV is severely impacted by the development of PAH, but this pathological injury may signal the release of circulating EVs that communicate with different organs, such as the brain, helping to prevent further damage through the upregulation of proteins involved in redox and mitochondrial function.
Collapse
|
18
|
Ionescu RF, Cretoiu SM. MicroRNAs as monitoring markers for right-sided heart failure and congestive hepatopathy. J Med Life 2021; 14:142-147. [PMID: 34104236 PMCID: PMC8169151 DOI: 10.25122/jml-2021-0071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The last decades showed a worrying increase in the evolution of cardiovascular diseases towards different stages of heart failure (HF), as a stigma of the western lifestyle. MicroRNAs (miRNAs), non-coding RNAs, which are approximately 22-nucleotide long, were shown to regulate gene expression at the post-transcriptional level and play a role in the pathogenesis and progression of HF. miRNAs research is of high interest nowadays, as these molecules display mechanisms of action that can influence the course of evolution of common chronic diseases, including HF. The potential of post-transcriptional regulation by miRNAs concerning the diagnosis, management, and therapy for HF represents a new promising approach in the accurate assessment of cardiovascular diseases. This review aims to assess the current knowledge of miRNAs in cardiovascular diseases, especially right-sided heart failure and hepatomegaly. Moreover, attention is focused on their role as potential molecular biomarkers and more promising aspects involving miRNAs as future therapeutic targets in the pathophysiology of HF.
Collapse
Affiliation(s)
- Ruxandra Florentina Ionescu
- Department of Cardiology I, Central Military Emergency University Hospital Dr. Carol Davila, Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
19
|
Yang T, He R, Li G, Liang J, Zhao L, Zhao X, Li L, Wang P. Growth arrest and DNA damage-inducible protein 34 (GADD34) contributes to cerebral ischemic injury and can be detected in plasma exosomes. Neurosci Lett 2021; 758:136004. [PMID: 34098025 DOI: 10.1016/j.neulet.2021.136004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
Growth arrest and DNA damage-inducible protein 34 (GADD34), one of the key effectors of negative feedback loops, is induced by stress and subsequently attempts to restore homeostasis. It plays a critical role in response to DNA damage and endoplasmic reticulum stress. GADD34 has opposing effects on different stimulus-induced cell apoptosis events in many nervous system diseases, but its role in ischemic stroke is unclear. In this study, we evaluated the role of GADD34 and its distribution in a rat cerebral ischemic model. The results showed that GADD34 was increased in the cortex and contributed to brain injury in ischemic rats. Furthermore, treatment with a GADD34 inhibitor reduced the infarct volume, improved functional outcomes, and inhibited neuronal apoptosis in the cortical penumbra after ischemia. The role of GADD34 in ischemic stroke was associated with the dephosphorylation of eukaryotic translation initiation factor 2α (eIF2α) and phosphorylation of p53. In addition, the GADD34 level was increased in plasma exosomes of cerebral ischemic rats. These findings indicate that GADD34 could be a potential therapeutic target and biomarker for ischemic stroke.
Collapse
Affiliation(s)
- Tianhui Yang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ruyi He
- College of Pharmacy, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Gongzhe Li
- College of Pharmacy, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jia Liang
- Institution of Life Science, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Liang Zhao
- College of Pharmacy, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xing Zhao
- Department of Ophthalmology and Otolaryngology, Liaoning Provincial Corps Hospital of Chinese People's Armed Police Forces, Shenyang, Liaoning, China
| | - Liyang Li
- Department of Ophthalmology and Otolaryngology, Liaoning Provincial Corps Hospital of Chinese People's Armed Police Forces, Shenyang, Liaoning, China
| | - Peng Wang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
20
|
Zhu Z, Shen Y, Chen Y, Shi H, Shi Y. The exosome of platelet endothelial cell adhesion molecule-1 (PECAM1) protein: A potential risking star in high blood pressure patients (HBPP). Medicine (Baltimore) 2021; 100:e21370. [PMID: 33530152 PMCID: PMC7850734 DOI: 10.1097/md.0000000000021370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/18/2020] [Indexed: 01/05/2023] Open
Abstract
A number of studies have demonstrated that exosomes were involved in important physiological and pathological processes through cell-to-cell communication in cardiovascular disease, which contained nucleic acids, proteins, and lipid contents. In our study, we found that the protein platelet endothelial cell adhesion molecule-1 (PECAM1) was an extracellular vesicle in the blood of high blood pressure patients (HBPP).Isolated the vesicles from the blood of HBPP and health examiners and detected its size and morphology with nanoparticle tracking analysis, then we identified its surface protein CD63, CD81, and the protein expression of PECAM1 in the exosome with western blot. Furthermore, we analyzed the correlation between the expression of PECAM1 and the high blood degree with linear regression analysis.Our results showed that the morphology of extracellular vesicles was more evident in high blood pressure groups than healthy controls, and the protein expression of PECAM1 was also abundant in the vesicles of HBPP, however, there were no extracellular vesicles in the blood samples of healthy controls. Besides, linear regression showed the linear correlation coefficient R = 0.901, P < .01 between the expression of PECAM1 and the systolic blood pressure of the high blood patients. Therefore, the exosome of protein of PECAM1 was a potential risking star in HBPP.
Collapse
Affiliation(s)
- Zhidong Zhu
- Department of Cardiology, North Hospital of Huashan Hospital Affiliated to Fudan University
| | | | | | - Haiming Shi
- Department of Cardiology, Huashan Sub-Hospital of Fudan University
| | - Yun Shi
- Department of Cardiovascular Medicine, Kong Jiang Hospital of Yangpu District, Shuangyang Road, Shanghai, China
| |
Collapse
|
21
|
Khan AQ, Akhtar S, Prabhu KS, Zarif L, Khan R, Alam M, Buddenkotte J, Ahmad A, Steinhoff M, Uddin S. Exosomes: Emerging Diagnostic and Therapeutic Targets in Cutaneous Diseases. Int J Mol Sci 2020; 21:ijms21239264. [PMID: 33291683 PMCID: PMC7730213 DOI: 10.3390/ijms21239264] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Skin is the largest human organ and is continuously exposed to various exogenous and endogenous trigger factors affecting body homeostasis. A number of mechanisms, including genetic, inflammatory and autoimmune ones, have been implicated in the pathogenesis of cutaneous diseases. Recently, there has been considerable interest in the role that extracellular vesicles, particularly exosomes, play in human diseases, through their modulation of multiple signaling pathways. Exosomes are nano-sized vesicles secreted by all cell types. They function as cargo carriers shuttling proteins, nucleic acids, lipids etc., thus impacting the cell-cell communications and transfer of vital information/moieties critical for skin homeostasis and disease pathogenesis. This review summarizes the available knowledge on how exosomes affect pathogenesis of cutaneous diseases, and highlights their potential as future targets for the therapy of various skin diseases.
Collapse
Affiliation(s)
- Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
| | - Sabah Akhtar
- Department of Biological and Environmental Sciences, Qatar University, Doha 2713, Qatar; (S.A.); (L.Z.)
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
| | - Lubna Zarif
- Department of Biological and Environmental Sciences, Qatar University, Doha 2713, Qatar; (S.A.); (L.Z.)
| | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India;
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Correspondence: (A.A.); (M.S.); (S.U.); Tel.: +974-40253220 (S.U.)
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar
- Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- College of Medicine, Qatar University, Doha 2713, Qatar
- Correspondence: (A.A.); (M.S.); (S.U.); Tel.: +974-40253220 (S.U.)
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
- Correspondence: (A.A.); (M.S.); (S.U.); Tel.: +974-40253220 (S.U.)
| |
Collapse
|
22
|
Henning RJ. Cardiovascular Exosomes and MicroRNAs in Cardiovascular Physiology and Pathophysiology. J Cardiovasc Transl Res 2020; 14:195-212. [PMID: 32588374 DOI: 10.1007/s12265-020-10040-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022]
Abstract
Cardiac exosomes mediate cell-to-cell communication, stimulate or inhibit the activities of target cells, and affect myocardial hypertrophy, injury and infarction, ventricular remodeling, angiogenesis, and atherosclerosis. The exosomes that are released in the heart from cardiomyocytes, vascular cells, fibroblasts, and resident stem cells are hypoimmunogenic, are physiologically more stable than cardiac cells, can circulate in the body, and are able to cross the blood-brain barrier. Exosomes utilize three mechanisms for cellular communication: (1) internalization by cells, (2) direct fusion to the cell membrane, and (3) receptor-ligand interactions. Cardiac exosomes transmit proteins, mRNA, and microRNAs to other cells during both physiological and pathological process. Cardiac-specific exosome miRNAs can regulate the expression of sarcomeric genes, ion channel genes, autophagy, anti-apoptotic and anti-fibrotic activity, and angiogenesis. This review discusses the role of exosomes and microRNAs in normal myocardium, myocardial injury and infarction, atherosclerosis, and the importance of circulating microRNAs as biomarkers of cardiac disease. Graphical Abstract.
Collapse
Affiliation(s)
- Robert J Henning
- University of South Florida, 13201 Bruce B. Downs Blvd., Tampa, FL, 33612-3805, USA.
| |
Collapse
|
23
|
Jiang Y, He R, Shi Y, Liang J, Zhao L. Plasma exosomes protect against cerebral ischemia/reperfusion injury via exosomal HSP70 mediated suppression of ROS. Life Sci 2020; 256:117987. [PMID: 32569778 DOI: 10.1016/j.lfs.2020.117987] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022]
Abstract
AIMS Ischemic stroke is the leading cause of severe disability and death worldwide. As the pathogenesis of stroke has not been clearly elucidated and the ability of current therapeutic drugs on crossing the blood-brain barrier (BBB) is extremely low, there is no effective strategy to treat stroke. We aim at investigating the specific advantages of using plasma exosomes (Pla-Exo) for targeting ischemic brain and exploring its underlying mechanism in neuroprotection. MAIN METHODS Pla-Exo was obtained by a gradient ultracentrifugation of fresh plasma. The quantification of penetrated Pla-Exo through BBB was investigated in vitro BBB model, furthermore, the effects of Pla-Exo and exosomal HSP70 on cerebral ischemia/reperfusion injury were evaluated. KEY FINDINGS Pla-Exo enhanced BBB crossing by specific interaction between Pla-Exo inherited heat shock protein 70 (HSP70) and endothelial Toll-like receptor 4 (TLR4). As expected, Pla-Exo increased HSP70 expression in the ischemic region through the transfer of HSP70, and led to HSP70 mediated suppression of ROS, thus alleviating cerebral ischemia/reperfusion (I/R) injury by attenuating the deterioration of BBB and preventing mitochondria damage. SIGNIFICANCE These findings indicated that Pla-Exo can provide protection against ischemia-reperfusion injury via the regulation of HSP70 and it should be further studied as a potential candidate for protection against ischemic injury.
Collapse
Affiliation(s)
- Yibing Jiang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ruyi He
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jia Liang
- Life Science Institution, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| |
Collapse
|
24
|
Chinnappan M, Srivastava A, Amreddy N, Razaq M, Pareek V, Ahmed R, Mehta M, Peterson JE, Munshi A, Ramesh R. Exosomes as drug delivery vehicle and contributor of resistance to anticancer drugs. Cancer Lett 2020; 486:18-28. [PMID: 32439419 DOI: 10.1016/j.canlet.2020.05.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
Abstract
Exosomes are small membranous vesicles implicated in intercellular signalling. Through their uncanny ability to carry and deliver donor cellular cargo (biomolecules) to target cells, they exert a profound effect on the regular functioning of healthy cells and play a significant role in pathogenesis and progression of several diseases, including cancer. The composition and number of endogenously circulating exosomes frequently vary, which is often reflective of the pathophysiological status of the cell. Applicability of exosomes derived from normal cells as a drug carrier with or without modifying their intraluminal and surface components are generally tested. Conversely, exosomes also are reported to contribute to resistance towards several anti-cancer therapies. Therefore, it is necessary to carefully evaluate the role of exosomes in cancer progression, resistance and the potential use of exosomes as a delivery vehicle of cancer therapeutics. In this review, we summarize the recent advancements in the exploitation of exosomes as a drug delivery vehicle. We also discuss the role of exosomes in conferring resistance to anti-cancer therapeutics. While this review is focused on cancer, the exosome-based drug delivery and resistance is also applicable to other human diseases.
Collapse
Affiliation(s)
- Mahendran Chinnappan
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Akhil Srivastava
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Narsireddy Amreddy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohammad Razaq
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Vipul Pareek
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rebaz Ahmed
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Meghna Mehta
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jo Elle Peterson
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
25
|
Vechetti IJ, Valentino T, Mobley CB, McCarthy JJ. The role of extracellular vesicles in skeletal muscle and systematic adaptation to exercise. J Physiol 2020; 599:845-861. [PMID: 31944292 DOI: 10.1113/jp278929] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022] Open
Abstract
Regular exercise has a central role in human health by reducing the risk of type 2 diabetes, obesity, stroke and cancer. How exercise is able to promote such systemic benefits has remained somewhat of a mystery but has been thought to be in part mediated by the release of myokines, skeletal muscle-specific cytokines, in response to exercise. Recent studies have revealed skeletal muscle can also release extracellular vesicles (EVs) into circulation following a bout of exercise. EVs are small membrane-bound vesicles capable of delivering biomolecules to recipient cells and subsequently altering their metabolism. The notion that EVs may have a role in both skeletal muscle and systemic adaptation to exercise has generated a great deal of excitement within a number of different fields including exercise physiology, neuroscience and metabolism. The purpose of this review is to provide an introduction to EV biology and what is currently known about skeletal muscle EVs and their potential role in the response of muscle and other tissues to exercise.
Collapse
Affiliation(s)
- Ivan J Vechetti
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Taylor Valentino
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - C Brooks Mobley
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
26
|
Luo Y, Huang L, Luo W, Ye S, Hu Q. Genomic analysis of lncRNA and mRNA profiles in circulating exosomes of patients with rheumatic heart disease. Biol Open 2019; 8:bio.045633. [PMID: 31784421 PMCID: PMC6918777 DOI: 10.1242/bio.045633] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Rheumatic heart disease (RHD) remains one of the most common cardiovascular conditions in developing countries. Accumulating evidence suggests that circulating exosomes and their cargoes, including mRNA and long noncoding RNA (lncRNA), play essential roles in many cardiovascular diseases. However, their specific roles in RHD remain unexplored. In the present study, we identified 231 lncRNAs and 179 mRNAs differentially expressed in the circulating exosomes harvested from RHD patients compared to healthy controls. We performed gene ontology (GO) and KEGG pathway analysis, and identified five pairs of lncRNAs and their flanking coding genes simultaneously dysregulated in the circulating exosomes. Collectively, we provide the first transcriptome analysis identifying differentially expressed lncRNAs and mRNAs in circulating exosomes of RHD patients, which may bring valuable insights for the discovery of potential biomarkers and therapeutic targets for RHD.
Collapse
Affiliation(s)
- Yanli Luo
- Department of Anesthesiology, Xiangya Hospital, Central-South University, Changsha, Hunan Province, China 410008
| | - Lingjin Huang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central-South University, Changsha, Hunan Province, China 410008
| | - Wanjun Luo
- Department of Cardiovascular Surgery, Xiangya Hospital, Central-South University, Changsha, Hunan Province, China 410008
| | - Shu Ye
- Department of Dermatology, Hunan Children's Hospital, Changsha, Hunan Province, China 410007
| | - Qinghua Hu
- Department of Cardiovascular Surgery, Xiangya Hospital, Central-South University, Changsha, Hunan Province, China 410008
| |
Collapse
|
27
|
Chen L, Han Y, Li Y, Chen B, Bai X, Belguise K, Wang X, Chen Y, Yi B, Lu K. Hepatocyte-derived exosomal MiR-194 activates PMVECs and promotes angiogenesis in hepatopulmonary syndrome. Cell Death Dis 2019; 10:853. [PMID: 31700002 PMCID: PMC6838168 DOI: 10.1038/s41419-019-2087-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/27/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022]
Abstract
Hepatopulmonary syndrome (HPS) is a serious vascular complication in the setting of liver disease. Factors produced by the liver are essential to regulate pulmonary angiogenesis in the pathogenesis of HPS; however, the pathogenic mechanisms of pulmonary angiogenesis are not fully understood. We investigated the role of HPS rat serum exosomes (HEs) and sham-operated rat serum exosomes (SEs) in the regulation of angiogenesis. We found that HEs significantly enhance PMVEC proliferation, migration, and tube formation. We further identified miR-194 was the most notably increased miRNA in HEs compared to SEs. Once released, hepatocyte-derived exosomal miR-194 was internalized by PMVECs, leading to the promotion of PMVEC proliferation, migration, and tube formation through direct targeting of THBS1, STAT1, and LIF. Importantly, the pathogenic role of exosomal miR-194 in initiating angiogenesis was reversed by P53 inhibition, exosome secretion inhibition or miR-194 inhibition. Additionally, high levels of miR-194 were found in serum exosomes and were positively correlated with P(A-a)O2 in HPS patients and rats. Thus, our results highlight that the exosome/miR-194 axis plays a critical pathologic role in pulmonary angiogenesis, representing a new therapeutic target for HPS.
Collapse
Affiliation(s)
- Lin Chen
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yi Han
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yujie Li
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Bing Chen
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Xuehong Bai
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Karine Belguise
- LBCMCP, ×tégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Xiaobo Wang
- LBCMCP, ×tégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yang Chen
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| | - Bin Yi
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| | - Kaizhi Lu
- Department of Anaesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
28
|
Wang S, Min J, Yu Y, Yin L, Wang Q, Shen H, Yang J, Zhang P, Xiao J, Wang Z. Differentially expressed miRNAs in circulating exosomes between atrial fibrillation and sinus rhythm. J Thorac Dis 2019; 11:4337-4348. [PMID: 31737319 DOI: 10.21037/jtd.2019.09.50] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Exosomes are small (30-150 nm) membrane vesicles released by cells that transmit intercellular information. As one of the contents of exosomes, microRNAs (miRNAs) may play an important role in the pathogenesis of atrial fibrillation (AF). Exosomal miRNAs potentially function as biomarkers in AF, as shown in many other diseases. Methods To identify the different expression level of plasma exosomal miRNAs between persistent AF and sinus rhythm (SR) patients, we performed high-throughput sequencing of small RNAs in the exosomes of AF (n=4) and SR (n=4) patients. Target genes of the DE miRNAs were predicted and put into gene ontology analysis and pathway analysis. In the validation phase, we performed quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) of 6 of the DE miRNAs in AF (n=40) and SR (n=20) patients. Univariate and multivariate logistic analysis were used to analyze risk factors of AF. Results With high-throughput sequencing, we revealed 39 differentially expressed (DE) miRNAs in circulating exosomes. We validated 4 of the DE plasma exosomal miRNAs (miR-483-5p, miR-142-5p, miR-223-3p, miR-223-5p) using qRT-PCR. Univariate logistic analysis shows miR-483-5p, miR-142-5p, miR-223-3p are related with AF, while multivariate logistic analysis suggests miR-483-5p is independently in correlation with AF. Conclusions This discovery opens up a new perspective in the complicated mechanism underlying AF and the DE plasma exosomal miRNAs exert enormous potential of acting as biomarkers in assessing severity or prognostic of AF and help selecting therapeutic strategy.
Collapse
Affiliation(s)
- Suyu Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Jie Min
- Department of Cardiothoracic Surgery, Bethune international peace hospital, Shijiazhuang 50082, China
| | - Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Liang Yin
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Qing Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Hua Shen
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Jie Yang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Peng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai 200003, China
| |
Collapse
|
29
|
Yu H, Wang Z. Cardiomyocyte-Derived Exosomes: Biological Functions and Potential Therapeutic Implications. Front Physiol 2019; 10:1049. [PMID: 31481897 PMCID: PMC6710398 DOI: 10.3389/fphys.2019.01049] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022] Open
Abstract
Exosomes, which are membrane-enclosed nanovesicles released by almost all cell types, have been recognized to play important roles in mediating cell-cell communication. In recent years, the physiological and pathological effects of exosomes on cardiovascular disease have been extensively studied. Exosomes can transfer proteins, mRNAs, microRNAs, and other bioactive molecules to recipient cells to influence their biological properties. In recent years, accumulating evidence has suggested that cardiomyocyte-derived exosomes play an important role in the progression of cardiovascular disease. Here, we summarize the functional roles of cardiomyocyte-derived exosomes in cardiovascular physiology and pathology.
Collapse
Affiliation(s)
- Hui Yu
- The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
30
|
Dong R, Liu Y, Yang Y, Wang H, Xu Y, Zhang Z. MSC-Derived Exosomes-Based Therapy for Peripheral Nerve Injury: A Novel Therapeutic Strategy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6458237. [PMID: 31531362 PMCID: PMC6719277 DOI: 10.1155/2019/6458237] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/06/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Although significant advances have been made in synthetic nerve conduits and surgical techniques, complete regeneration following peripheral nerve injury (PNI) remains far from optimized. The repair of PNI is a highly heterogeneous process involving changes in Schwann cell phenotypes, the activation of macrophages, and the reconstruction of the vascular network. At present, the efficacy of MSC-based therapeutic strategies for PNI can be attributed to paracrine secretion. Exosomes, as a product of paracrine secretion, are considered to be an important regulatory mediator. Furthermore, accumulating evidence has demonstrated that exosomes from mesenchymal stem cells (MSCs) can shuttle bioactive components (proteins, lipids, mRNA, miRNA, lncRNA, circRNA, and DNA) that participate in almost all of the abovementioned processes. Thus, MSC exosomes may represent a novel therapeutic tool for PNI. In this review, we discuss the current understanding of MSC exosomes related to peripheral nerve repair and provide insights for developing a cell-free MSC therapeutic strategy for PNI.
Collapse
Affiliation(s)
- Ruiqi Dong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Yuxiang Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Haojie Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Yaolu Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, China
| |
Collapse
|
31
|
On the Choice of the Extracellular Vesicles for Therapeutic Purposes. Int J Mol Sci 2019; 20:ijms20020236. [PMID: 30634425 PMCID: PMC6359369 DOI: 10.3390/ijms20020236] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/29/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid membrane vesicles released by all human cells and are widely recognized to be involved in many cellular processes, both in physiological and pathological conditions. They are mediators of cell-cell communication, at both paracrine and systemic levels, and therefore they are active players in cell differentiation, tissue homeostasis, and organ remodeling. Due to their ability to serve as a cargo for proteins, lipids, and nucleic acids, which often reflects the cellular source, they should be considered the future of the natural nanodelivery of bio-compounds. To date, natural nanovesicles, such as exosomes, have been shown to represent a source of disease biomarkers and have high potential benefits in regenerative medicine. Indeed, they deliver both chemical and bio-molecules in a way that within exosomes drugs are more effective that in their exosome-free form. Thus, to date, we know that exosomes are shuttle disease biomarkers and probably the most effective way to deliver therapeutic molecules within target cells. However, we do not know exactly which exosomes may be used in therapy in avoiding side effects as well. In regenerative medicine, it will be ideal to use autologous exosomes, but it seems not ideal to use plasma-derived exosomes, as they may contain potentially dangerous molecules. Here, we want to present and discuss a contradictory relatively unmet issue that is the lack of a general agreement on the choice for the source of extracellular vesicles for therapeutic use.
Collapse
|
32
|
Das CK, Jena BC, Banerjee I, Das S, Parekh A, Bhutia SK, Mandal M. Exosome as a Novel Shuttle for Delivery of Therapeutics across Biological Barriers. Mol Pharm 2018; 16:24-40. [PMID: 30513203 DOI: 10.1021/acs.molpharmaceut.8b00901] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The effective delivery of target-specific siRNA to the brain by exploiting the exosomes derived from dendritic cells renders the paradigm shift for the prospective use of nanosized exosomes as a delivery system. Although the in vivo targeting strategies by other nanovesicles like liposomes exist, still this novel exosome-based delivery approach holds an inclusive dominance of in vivo security and reduced immunogenicity. Achieving promising exosome-based delivery strategies warrants more desirable exploration of their biology. Over the years, the invention of novel production, characterization, targeting strategies, and cargo loading techniques of exosome improved its ability to reach clinics. Essentially, exosome-based delivery of therapeutics assures to conquer the major hurdles, like delivery of cargos across impermeable biological barriers, like the blood-brain barrier, biocompatibility, increased solubility, metabolic stability, improved circulation time, target specific delivery, and pharmacokinetics, and thereby enhanced the efficacy of loaded therapeutic agents. In this article, we cover the current status of exosome as a delivery vehicle for therapeutics and the challenges that need to be overcome, and we also discuss future perspectives of this exciting field of research to transform it from bench to clinical reality.
Collapse
Affiliation(s)
- Chandan Kanta Das
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Bikash Chandra Jena
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Indranil Banerjee
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Subhayan Das
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Aditya Parekh
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Sujit Kumar Bhutia
- Department of Life Science , National Institute of Technology , Rourkela , India
| | - Mahitosh Mandal
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| |
Collapse
|
33
|
Pan W, Zhu Y, Meng X, Zhang C, Yang Y, Bei Y. Immunomodulation by Exosomes in Myocardial Infarction. J Cardiovasc Transl Res 2018; 12:28-36. [PMID: 30374796 DOI: 10.1007/s12265-018-9836-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
Exosomes are important carriers of biological information that facilitate intercellular communication and participate in the pathophysiology of different cardiovascular diseases. Myocardial infarction is among the leading causes of death worldwide. Upon myocardial infarction, massive cardiomyocyte death triggers a strong inflammatory response which is a vital process of cardiac injury, repair, and remodeling. Increasing evidence has unveiled that exosomes are involved in the inflammatory response and immune regulation after myocardial infarction. In this review, we will summarize the biological function of exosomes in the pathophysiology of myocardial infarction, especially focusing on their roles in the modulation of inflammation and immune response after myocardial infarction which further influences myocardial repair and remodeling. We will also discuss the immunomodulation by exosomes derived from stem and progenitor cells in the treatment of myocardial infarction. A deep understanding of immunomodulation by exosomes may represent a promising therapeutic option for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Wen Pan
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.,Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xiangmin Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Chenlin Zhang
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Yihua Bei
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
| |
Collapse
|
34
|
Tao L, Shi J, Yang X, Yang L, Hua F. The Exosome: a New Player in Diabetic Cardiomyopathy. J Cardiovasc Transl Res 2018; 12:62-67. [PMID: 30251219 DOI: 10.1007/s12265-018-9825-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022]
Abstract
Diabetic cardiomyopathy (DCM) or diabetes-induced cardiac dysfunction is a direct consequence of uncontrolled metabolic syndrome and occurs worldwide. However, the underlying cellular and molecular mechanisms remain poorly understood. Recently, exosomes have attracted considerable interest for their use as efficient, targeted, and non-immunogenic delivery systems for biological molecules or pharmacotherapies. This review will summarize the fast-developing field of the regulation and function of exosomes in DCM, affording valuable insights and therapeutic opportunities in combatting diabetes-related cardiac disorder for modern human health.
Collapse
Affiliation(s)
- Lichan Tao
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Xiaoyu Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China.
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China.
| |
Collapse
|