1
|
Hu H, Wang X, Yu H, Wang Z. Extracellular vesicular microRNAs and cardiac hypertrophy. Front Endocrinol (Lausanne) 2025; 15:1444940. [PMID: 39850481 PMCID: PMC11753959 DOI: 10.3389/fendo.2024.1444940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Cardiac hypertrophy is an adaptive response to pressure or volume overload such as hypertension and ischemic heart diseases. Sustained cardiac hypertrophy eventually leads to heart failure. The pathophysiological alterations of hypertrophy are complex, involving both cellular and molecular systems. Understanding the molecular events that inhibit or repress cardiac hypertrophy may help identify novel therapeutic strategies. Increasing evidence has indicated that extracellular vesicle (EV)-derived microRNAs (miRNAs) play a significant role in the development and progression of cardiac hypertrophy. In this review, we briefly review recent advancements in EV research, especially on biogenesis, cargoes and its role in cardiac hypertrophy. We then describe the latest findings regarding EV-derived miRNAs, highlighting their functions and regulatory mechanisms in cardiac hypertrophy. Finally, the potential role of EV-derived miRNAs as targets in the diagnosis and treatment of cardiac hypertrophy will be discussed.
Collapse
Affiliation(s)
- Hai Hu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xiulian Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
2
|
Ebrahimi F, Kumari A, Ghadami S, Al Abdullah S, Dellinger K. The Potential for Extracellular Vesicles in Nanomedicine: A Review of Recent Advancements and Challenges Ahead. Adv Biol (Weinh) 2024:e2400623. [PMID: 39739455 DOI: 10.1002/adbi.202400623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Indexed: 01/02/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising tools in diagnostics and therapy for chronic diseases, including cancer and Alzheimer's. Small EVs, also called exosomes, are lipid-bound particles (≈30-150 nm) that play a role in healthy and pathophysiological interactions, including intercellular communication, by transporting bioactive molecules, including proteins, lipids, and nucleic acids. Their ability to cross biological barriers, such as the blood-brain barrier, makes them ideal candidates for targeted therapeutic interventions. In the context of chronic diseases, exosomes can be engineered to deliver active agents, including small molecules and siRNAs to specific target cells, providing a novel approach to precision medicine. Moreover, exosomes show great promise as repositories for diagnostic biomarkers. Their cargo can reflect the physiological and pathological status of the parent cells, making them valuable indicators of disease progression and response to treatment. This paper presents a comprehensive review of the application of exosomes in four chronic diseases: cancer, cardiovascular disease, neurodegenerative disease, and orthopedic disease, which significantly impact global public health due to their high prevalence and associated morbidity and mortality rates. Furthermore, the potential of exosomes as valuable tools for theranostics and disease management is highlighted. Finally, the challenges associated with exosomes and their demonstrated potential for advancing future nanomedicine applications are discussed.
Collapse
Affiliation(s)
- Farbod Ebrahimi
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Anjali Kumari
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Samaneh Ghadami
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Saqer Al Abdullah
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| |
Collapse
|
3
|
Song R, Zhang L. MicroRNAs and therapeutic potentials in acute and chronic cardiac disease. Drug Discov Today 2024; 29:104179. [PMID: 39276921 DOI: 10.1016/j.drudis.2024.104179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
microRNAs (miRNAs) are small regulatory RNAs implicated in various cardiac disorders. In this review, the role of miRNAs is discussed in relation to acute myocardial infarction and chronic heart failure. In both settings, miRNAs are altered, contributing to injury and adverse remodeling. Notably, miRNA profiles differ between acute ischemic injury and progressive heart failure. Owing to miRNA variabilities between disease stages and delivery difficulties, translation of animal studies to the clinic remains challenging. The identification of distinct miRNA signatures could lead to the development of miRNA therapies tailored to different disease stages. Here, we summarize the current understanding of miRNAs in acute and chronic cardiac diseases, identify knowledge gaps and discuss progress in developing miRNA-based treatment strategies.
Collapse
Affiliation(s)
- Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
4
|
Toghiani R, Azimian Zavareh V, Najafi H, Mirian M, Azarpira N, Abolmaali SS, Varshosaz J, Tamaddon AM. Hypoxia-preconditioned WJ-MSC spheroid-derived exosomes delivering miR-210 for renal cell restoration in hypoxia-reoxygenation injury. Stem Cell Res Ther 2024; 15:240. [PMID: 39080774 PMCID: PMC11289969 DOI: 10.1186/s13287-024-03845-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Recent advancements in mesenchymal stem cell (MSC) technology have paved the way for innovative treatment options for various diseases. These stem cells play a crucial role in tissue regeneration and repair, releasing local anti-inflammatory and healing signals. However, challenges such as homing issues and tumorigenicity have led to exploring MSC-exosomes as a promising alternative. MSC-exosomes have shown therapeutic potential in conditions like renal ischemia-reperfusion injury, but low production yields hinder their clinical use. METHODS To address this limitation, we examined hypoxic preconditioning of Wharton jelly-derived MSCs (WJ-MSCs) 3D-cultured in spheroids on isolated exosome yields and miR-21 expression. We then evaluated their capacity to load miR-210 into HEK-293 cells and mitigate ROS production, consequently enhancing their survival and migration under hypoxia-reoxygenation conditions. RESULTS MiR-210 overexpression was significantly induced by optimized culture and preconditioning conditions, which also improved the production yield of exosomes from grown MSCs. The exosomes enriched with miR-210 demonstrated a protective effect by improving survival, reducing apoptosis and ROS accumulation in damaged renal cells, and ultimately promoting cell migration. CONCLUSION The present study underscores the possibility of employing advanced techniques to maximize the therapeutic attributes of exosomes produced from WJ-MSC spheroid for improved recovery outcomes in ischemia-reperfusion injuries.
Collapse
Affiliation(s)
- Reyhaneh Toghiani
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vajihe Azimian Zavareh
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Hanyieh Najafi
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Wang Y, Shi X. The potential mechanisms and treatment effects of stem cell-derived exosomes in cardiac reengineering. NANOTECHNOLOGY 2024; 35:362005. [PMID: 38834043 DOI: 10.1088/1361-6528/ad53d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Exosomes are extracellular vesicles of diverse compositions that are secreted by numerous cell types. Exosomes contain significant bioactive components, including lipids, proteins, mRNA, and miRNA. Exosomes play an important role in regulating cellular signaling and trafficking under both normal physiological and pathological circumstances. A multitude of factors, including thermal stress, ribosomal stress, endoplasmic reticulum stress, and oxidative stress influence the concentrations of exosomal mRNA, miRNA, proteins, and lipids. It has been stated that exosomes derived from stem cells (SCs) modulate a range of stresses by preventing or fostering cell balance. Exosomes derived from SCs facilitate recovery by facilitating cross-cellular communication via the transmission of information in the form of proteins, lipids, and other components. For this reason, exosomes are used as biomarkers to diagnose a wide variety of diseases. The focus of this review is the bioengineering of artificial exosomal cargoes. This process encompasses the control and transportation of particular exosomal cargoes, including but not limited to small molecules, recombinant proteins, immune modulators, and therapeutic medications. Therapeutic approaches of this nature have the potential to deliver therapeutic medications precisely to the intended site for the cure of a variety of disorders. Notably, our attention has been directed towards the therapeutic implementations of exosomes derived from SCs in the cure of cardiovascular ailments, including but not limited to ischemic heart disease, myocardial infarction, sepsis, heart failure, cardiomyopathy, and cardiac fibrosis. In general, researchers employ two methodologies when it comes to exosomal bioengineering. This review aims to explain the function of exosomes derived from SCs in the regulation of stress and present a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Cardiology, Hangzhou Ninth People's Hospital, Hangzhou 311225, People's Republic of China
| | - Xiulian Shi
- Emergency Department, Chun'an First People's Hospital, Hangzhou 311700, People's Republic of China
| |
Collapse
|
6
|
Chmielewski PP, Data K, Strzelec B, Farzaneh M, Anbiyaiee A, Zaheer U, Uddin S, Sheykhi-Sabzehpoush M, Mozdziak P, Zabel M, Dzięgiel P, Kempisty B. Human Aging and Age-Related Diseases: From Underlying Mechanisms to Pro-Longevity Interventions. Aging Dis 2024:AD.2024.0280. [PMID: 38913049 DOI: 10.14336/ad.2024.0280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Abstract
As human life expectancy continues to rise, becoming a pressing global concern, it brings into focus the underlying mechanisms of aging. The increasing lifespan has led to a growing elderly population grappling with age-related diseases (ARDs), which strains healthcare systems and economies worldwide. While human senescence was once regarded as an immutable and inexorable phenomenon, impervious to interventions, the emerging field of geroscience now offers innovative approaches to aging, holding the promise of extending the period of healthspan in humans. Understanding the intricate links between aging and pathologies is essential in addressing the challenges presented by aging populations. A substantial body of evidence indicates shared mechanisms and pathways contributing to the development and progression of various ARDs. Consequently, novel interventions targeting the intrinsic mechanisms of aging have the potential to delay the onset of diverse pathological conditions, thereby extending healthspan. In this narrative review, we discuss the most promising methods and interventions aimed at modulating aging, which harbor the potential to mitigate ARDs in the future. We also outline the complexity of senescence and review recent empirical evidence to identify rational strategies for promoting healthy aging.
Collapse
Affiliation(s)
- Piotr Pawel Chmielewski
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartłomiej Strzelec
- 2nd Department of General Surgery and Surgical Oncology, Medical University Hospital, Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Uzma Zaheer
- School of Biosciences, Faculty of Health Sciences and Medicine, The University of Surrey, United Kingdom
| | - Shahab Uddin
- Translational Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | | | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, Raleigh, NC 27695, USA
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Division of Anatomy and Histology, The University of Zielona Góra, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
- Center of Assisted Reproduction, Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| |
Collapse
|
7
|
Lin Q, He P, Tao J, Peng J. Role of Exosomes in Cardiovascular Diseases. Rev Cardiovasc Med 2024; 25:222. [PMID: 39076309 PMCID: PMC11270122 DOI: 10.31083/j.rcm2506222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 07/31/2024] Open
Abstract
Exosomes (EXOs) are a subgroup of extracellular vesicles (EVs) that contain numerous biologically active molecules. They exhibit an essential mode of cell communication, primarily between distinct cell populations, for the maintenance of tissue homeostasis and coordination of adaptive responses to various stresses. These intercellular communications are vital for the complex, multicellular cardiovascular system. In the last ten years, their potential role as effective tissue-to-tissue communicators has received increasing attention in cardiovascular physiology and pathology. There is growing evidence that repair of the heart and regeneration can be promoted by EXOs derived from cardiomyocytes or stem/progenitor cells. However, the underlying mechanisms remain unclear. EVs derived from different stem/progenitor cell populations have been used as cell-free therapies in different preclinical models involving cardiovascular diseases and have shown promising results. In this review, we have summarized the recent developments in EXOs research, the impact of EXOs derived from different cells on the cardiovascular system, their potential therapeutic roles as well as new diagnostic biomarkers, and the possible clinical translational outcomes.
Collapse
Affiliation(s)
- Qiumei Lin
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, 400014 Chongqing, China
| | - Pingfeng He
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, 400014 Chongqing, China
| | - Jing Tao
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, 400014 Chongqing, China
| | - Jing Peng
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, 400014 Chongqing, China
| |
Collapse
|
8
|
Wang C, Li H, Zhou H, Xu Y, Li S, Zhu M, Yu B, Feng Y. Intracranial aneurysm circulating exosome-derived LncRNA ATP1A1-AS1 promotes smooth muscle cells phenotype switching and apoptosis. Aging (Albany NY) 2024; 16:8320-8335. [PMID: 38728254 PMCID: PMC11132007 DOI: 10.18632/aging.205821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/26/2024] [Indexed: 05/12/2024]
Abstract
Exosomal long non-coding RNAs (LncRNAs) play a crucial role in the pathogenesis of cerebrovascular diseases. However, the expression profiles and functional significance of exosomal LncRNAs in intracranial aneurysms (IAs) remain poorly understood. Through high-throughput sequencing, we identified 1303 differentially expressed LncRNAs in the plasma exosomes of patients with IAs and healthy controls. Quantitative real-time polymerase chain reaction (qRT-PCR) verification confirmed the differential expression of LncRNAs, the majority of which aligned with the sequencing results. ATP1A1-AS1 showed the most significant upregulation in the disease group. Importantly, subsequent in vitro experiments validated that ATP1A1-AS1 overexpression induced a phenotype switching in vascular smooth muscle cells, along with promoting apoptosis and upregulating MMP-9 expression, potentially contributing to IAs formation. Furthermore, expanded-sample validation affirmed the high diagnostic value of ATP1A1-AS1. These findings suggest that ATP1A1-AS1 is a potential therapeutic target for inhibiting IAs progression and serves as a valuable clinical diagnostic marker.
Collapse
Affiliation(s)
- Chao Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Hong Li
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Han Zhou
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yifan Xu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Shifang Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Meng Zhu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Bing Yu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
9
|
Pinto AC, Tavares P, Neves B, Oliveira PF, Vitorino R, Moreira-Gonçalves D, Ferreira R. Exploiting the therapeutic potential of contracting skeletal muscle-released extracellular vesicles in cancer: Current insights and future directions. J Mol Med (Berl) 2024; 102:617-628. [PMID: 38451309 PMCID: PMC11055777 DOI: 10.1007/s00109-024-02427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
The health benefits of exercise training in a cancer setting are increasingly acknowledged; however, the underlying molecular mechanisms remain poorly understood. It has been suggested that extracellular vesicles (EVs) released from contracting skeletal muscles play a key role in mediating the systemic benefits of exercise by transporting bioactive molecules, including myokines. Nevertheless, skeletal muscle-derived vesicles account for only about 5% of plasma EVs, with the immune cells making the largest contribution. Moreover, it remains unclear whether the contribution of skeletal muscle-derived EVs increases after physical exercise or how muscle contraction modulates the secretory activity of other tissues and thus influences the content and profile of circulating EVs. Furthermore, the destination of EVs after exercise is unknown, and it depends on their molecular composition, particularly adhesion proteins. The cargo of EVs is influenced by the training program, with acute training sessions having a greater impact than chronic adaptations. Indeed, there are numerous questions regarding the role of EVs in mediating the effects of exercise, the clarification of which is critical for tailoring exercise training prescriptions and designing exercise mimetics for patients unable to engage in exercise programs. This review critically analyzes the current knowledge on the effects of exercise on the content and molecular composition of circulating EVs and their impact on cancer progression.
Collapse
Affiliation(s)
- Ana Carolina Pinto
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Patrícia Tavares
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Bruno Neves
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Daniel Moreira-Gonçalves
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
10
|
Yu T, Xu Q, Chen X, Deng X, Chen N, Kou MT, Huang Y, Guo J, Xiao Z, Wang J. Biomimetic nanomaterials in myocardial infarction treatment: Harnessing bionic strategies for advanced therapeutics. Mater Today Bio 2024; 25:100957. [PMID: 38322664 PMCID: PMC10844134 DOI: 10.1016/j.mtbio.2024.100957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Myocardial infarction (MI) and its associated poor prognosis pose significant risks to human health. Nanomaterials hold great potential for the treatment of MI due to their targeted and controlled release properties, particularly biomimetic nanomaterials. The utilization of biomimetic strategies based on extracellular vesicles (EVs) and cell membranes will serve as the guiding principle for the development of nanomaterial therapy in the future. In this review, we present an overview of research progress on various exosomes derived from mesenchymal stem cells, cardiomyocytes, or induced pluripotent stem cells in the context of myocardial infarction (MI) therapy. These exosomes, utilized as cell-free therapies, have demonstrated the ability to enhance the efficacy of reducing the size of the infarcted area and preventing ischaemic reperfusion through mechanisms such as oxidative stress reduction, polarization modulation, fibrosis inhibition, and angiogenesis promotion. Moreover, EVs can exert cardioprotective effects by encapsulating therapeutic agents and can be engineered to specifically target the infarcted myocardium. Furthermore, we discuss the use of cell membranes derived from erythrocytes, stem cells, immune cells and platelets to encapsulate nanomaterials. This approach allows the nanomaterials to camouflage themselves as endogenous substances targeting the region affected by MI, thereby minimizing toxicity and improving biocompatibility. In conclusion, biomimetic nano-delivery systems hold promise as a potentially beneficial technology for MI treatment. This review serves as a valuable reference for the application of biomimetic nanomaterials in MI therapy and aims to expedite the translation of NPs-based MI therapeutic strategies into practical clinical applications.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xu Chen
- Department of Clinical Pharmacy, Daqing Oilfield General Hospital, Daqing, 163000, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Man Teng Kou
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
11
|
Coughlan C, Lindenberger J, Jacot JG, Johnson NR, Anton P, Bevers S, Welty R, Graner MW, Potter H. Specific Binding of Alzheimer's Aβ Peptides to Extracellular Vesicles. Int J Mol Sci 2024; 25:3703. [PMID: 38612514 PMCID: PMC11011551 DOI: 10.3390/ijms25073703] [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: 01/23/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Alzheimer's disease (AD) is the fifth leading cause of death among adults aged 65 and older, yet the onset and progression of the disease is poorly understood. What is known is that the presence of amyloid, particularly polymerized Aβ42, defines when people are on the AD continuum. Interestingly, as AD progresses, less Aβ42 is detectable in the plasma, a phenomenon thought to result from Aβ becoming more aggregated in the brain and less Aβ42 and Aβ40 being transported from the brain to the plasma via the CSF. We propose that extracellular vesicles (EVs) play a role in this transport. EVs are found in bodily fluids such as blood, urine, and cerebrospinal fluid and carry diverse "cargos" of bioactive molecules (e.g., proteins, nucleic acids, lipids, metabolites) that dynamically reflect changes in the cells from which they are secreted. While Aβ42 and Aβ40 have been reported to be present in EVs, it is not known whether this interaction is specific for these peptides and thus whether amyloid-carrying EVs play a role in AD and/or serve as brain-specific biomarkers of the AD process. To determine if there is a specific interaction between Aβ and EVs, we used isothermal titration calorimetry (ITC) and discovered that Aβ42 and Aβ40 bind to EVs in a manner that is sequence specific, saturable, and endothermic. In addition, Aβ incubation with EVs overnight yielded larger amounts of bound Aβ peptide that was fibrillar in structure. These findings point to a specific amyloid-EV interaction, a potential role for EVs in the transport of amyloid from the brain to the blood, and a role for this amyloid pool in the AD process.
Collapse
Affiliation(s)
- Christina Coughlan
- University of Colorado Alzheimer’s and Cognition Center (CUACC), Linda Crnic Institute for Down Syndrome (LCI), Department of Neurology, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA (H.P.)
| | - Jared Lindenberger
- Structural Biology and Biophysics Core, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA (R.W.)
- Duke Human Vaccine Institute, Duke University, 2 Genome Ct., Durham, NC 27710, USA
| | - Jeffrey G. Jacot
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA
| | - Noah R. Johnson
- University of Colorado Alzheimer’s and Cognition Center (CUACC), Linda Crnic Institute for Down Syndrome (LCI), Department of Neurology, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA (H.P.)
| | - Paige Anton
- University of Colorado Alzheimer’s and Cognition Center (CUACC), Linda Crnic Institute for Down Syndrome (LCI), Department of Neurology, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA (H.P.)
| | - Shaun Bevers
- Structural Biology and Biophysics Core, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA (R.W.)
| | - Robb Welty
- Structural Biology and Biophysics Core, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA (R.W.)
| | - Michael W. Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA
| | - Huntington Potter
- University of Colorado Alzheimer’s and Cognition Center (CUACC), Linda Crnic Institute for Down Syndrome (LCI), Department of Neurology, University of Colorado Anschutz Medical Campus, 13001 E. 17th Pl, Aurora, CO 80045, USA (H.P.)
| |
Collapse
|
12
|
Jung SE, Kim SW, Choi JW. Exploring Cardiac Exosomal RNAs of Acute Myocardial Infarction. Biomedicines 2024; 12:430. [PMID: 38398032 PMCID: PMC10886708 DOI: 10.3390/biomedicines12020430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI), often a frequent symptom of coronary artery disease (CAD), is a leading cause of death and disability worldwide. Acute myocardial infarction (AMI), a major form of cardiovascular disease, necessitates a deep understanding of its complex pathophysiology to develop innovative therapeutic strategies. Exosomal RNAs (exoRNA), particularly microRNAs (miRNAs) within cardiac tissues, play a critical role in intercellular communication and pathophysiological processes of AMI. METHODS This study aimed to delineate the exoRNA landscape, focusing especially on miRNAs in animal models using high-throughput sequencing. The approach included sequencing analysis to identify significant miRNAs in AMI, followed by validation of the functions of selected miRNAs through in vitro studies involving primary cardiomyocytes and fibroblasts. RESULTS Numerous differentially expressed miRNAs in AMI were identified using five mice per group. The functions of 20 selected miRNAs were validated through in vitro studies with primary cardiomyocytes and fibroblasts. CONCLUSIONS This research enhances understanding of post-AMI molecular changes in cardiac tissues and investigates the potential of exoRNAs as biomarkers or therapeutic targets. These findings offer new insights into the molecular mechanisms of AMIs, paving the way for RNA-based diagnostics and therapeutics and therapies and contributing to the advancement of cardiovascular medicine.
Collapse
Affiliation(s)
- Seung Eun Jung
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Sang Woo Kim
- International St. Mary's Hospital, Incheon 22711, Republic of Korea
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Jung-Won Choi
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
13
|
Hussen BM, Abdullah ST, Abdullah SR, Younis YM, Hidayat HJ, Rasul MF, Mohamadtahr S. Exosomal non-coding RNAs: Blueprint in colorectal cancer metastasis and therapeutic targets. Noncoding RNA Res 2023; 8:615-632. [PMID: 37767111 PMCID: PMC10520679 DOI: 10.1016/j.ncrna.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) is ranked as the world's third-most prevalent cancer, and metastatic CRC considerably increases cancer-related fatalities globally. A number of complex mechanisms that are strictly controlled at the molecular level are involved in metastasis, which is the primary reason for death in people with CRC. Recently, it has become clear that exosomes, which are small extracellular vesicles released by non-tumorous and tumorigenic cells, play a critical role as communication mediators among tumor microenvironment (TME). To facilitate communication between the TME and cancer cells, non-coding RNAs (ncRNAs) play a crucial role and are recognized as potent regulators of gene expression and cellular processes, such as metastasis and drug resistance. NcRNAs are now recognized as potent regulators of gene expression and many hallmarks of cancer, including metastasis. Exosomal ncRNAs, like miRNAs, circRNAs, and lncRNAs, have been demonstrated to influence a number of cellular mechanisms that contribute to CRC metastasis. However, the molecular mechanisms that link exosomal ncRNAs with CRC metastasis are not well understood. This review highlights the essential roles that exosomal ncRNAs play in the progression of CRC metastatic disease and explores the therapeutic choices that are open to patients who have CRC metastases. However, exosomal ncRNA treatment strategy development is still in its early phases; consequently, additional investigation is required to improve delivery methods and find novel therapeutic targets as well as confirm the effectiveness and safety of these therapies in preclinical and clinical contexts.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Erbil, Kurdistan Region, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Sara Tharwat Abdullah
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Yousif Mohammed Younis
- Department of Nursing, College of Nursing, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Sayran Mohamadtahr
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| |
Collapse
|
14
|
Greco S, Made' A, Mutoli M, Zhang L, Piella SN, Vausort M, Lumley AI, Beltrami AP, Srivastava PK, Milani V, Boveri S, Ranucci M, Renna LV, Firat H, Bruno A, Spinetti G, Emanueli C, Devaux Y, Martelli F. HCG18, LEF1AS1 and lncCEACAM21 as biomarkers of disease severity in the peripheral blood mononuclear cells of COVID-19 patients. J Transl Med 2023; 21:758. [PMID: 37884975 PMCID: PMC10605335 DOI: 10.1186/s12967-023-04497-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/01/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Even after 3 years from SARS-CoV-2 identification, COVID-19 is still a persistent and dangerous global infectious disease. Significant improvements in our understanding of the disease pathophysiology have now been achieved. Nonetheless, reliable and accurate biomarkers for the early stratification of COVID-19 severity are still lacking. Long noncoding RNAs (LncRNAs) are ncRNAs longer than 200 nucleotides, regulating the transcription and translation of protein-coding genes and they can be found in the peripheral blood, thus holding a promising biomarker potential. Specifically, peripheral blood mononuclear cells (PBMCs) have emerged as a source of indirect biomarkers mirroring the conditions of tissues: they include monocytes, B and T lymphocytes, and natural killer T cells (NKT), being highly informative for immune-related events. METHODS We profiled by RNA-Sequencing a panel of 2906 lncRNAs to investigate their modulation in PBMCs of a pilot group of COVID-19 patients, followed by qPCR validation in 111 hospitalized COVID-19 patients. RESULTS The levels of four lncRNAs were found to be decreased in association with COVID-19 mortality and disease severity: HLA Complex Group 18-242 and -244 (HCG18-242 and HCG18-244), Lymphoid Enhancer Binding Factor 1-antisense 1 (LEF1-AS1) and lncCEACAM21 (i.e. ENST00000601116.5, a lncRNA in the CEACAM21 locus). Interestingly, these deregulations were confirmed in an independent patient group of hospitalized patients and by the re-analysis of publicly available single-cell transcriptome datasets. The identified lncRNAs were expressed in all of the PBMC cell types and inversely correlated with the neutrophil/lymphocyte ratio (NLR), an inflammatory marker. In vitro, the expression of LEF1-AS1 and lncCEACAM21 was decreased upon THP-1 monocytes exposure to a relevant stimulus, hypoxia. CONCLUSION The identified COVID-19-lncRNAs are proposed as potential innovative biomarkers of COVID-19 severity and mortality.
Collapse
Affiliation(s)
- Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy.
| | - Alisia Made'
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Martina Mutoli
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
| | - Lu Zhang
- Bioinformatics Platform, Data Integration and Analysis Unit, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Mélanie Vausort
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Andrew I Lumley
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | | | - Prashant Kumar Srivastava
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, W12 0NN, England, UK
| | - Valentina Milani
- Laboratory of Biostatistics and Data Management, Scientific Directorate, IRCCS Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Sara Boveri
- Laboratory of Biostatistics and Data Management, Scientific Directorate, IRCCS Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Marco Ranucci
- Department of Cardiovascular Anesthesia and ICU, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Laura Valentina Renna
- Biobank BioCor, IRCCS-Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | | | - Antonino Bruno
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Via Monte Generoso 71, 21100, Varese, Italy
| | - Gaia Spinetti
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, W12 0NN, England, UK.
- National Heart & Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK.
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy.
| |
Collapse
|
15
|
Zarà M, Baggiano A, Amadio P, Campodonico J, Gili S, Annoni A, De Dona G, Carerj ML, Cilia F, Formenti A, Fusini L, Banfi C, Gripari P, Tedesco CC, Mancini ME, Chiesa M, Maragna R, Marchetti F, Penso M, Tassetti L, Volpe A, Bonomi A, Marenzi G, Pontone G, Barbieri SS. Circulating Small Extracellular Vesicles Reflect the Severity of Myocardial Damage in STEMI Patients. Biomolecules 2023; 13:1470. [PMID: 37892152 PMCID: PMC10605123 DOI: 10.3390/biom13101470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Circulating small extracellular vesicles (sEVs) contribute to inflammation, coagulation and vascular injury, and have great potential as diagnostic markers of disease. The ability of sEVs to reflect myocardial damage assessed by Cardiac Magnetic Resonance (CMR) in ST-segment elevation myocardial infarction (STEMI) is unknown. To fill this gap, plasma sEVs were isolated from 42 STEMI patients treated by primary percutaneous coronary intervention (pPCI) and evaluated by CMR between days 3 and 6. Nanoparticle tracking analysis showed that sEVs were greater in patients with anterior STEMI (p = 0.0001), with the culprit lesion located in LAD (p = 0.045), and in those who underwent late revascularization (p = 0.038). A smaller sEV size was observed in patients with a low myocardial salvage index (MSI, p = 0.014). Patients with microvascular obstruction (MVO) had smaller sEVs (p < 0.002) and lower expression of the platelet marker CD41-CD61 (p = 0.039). sEV size and CD41-CD61 expression were independent predictors of MVO/MSI (OR [95% CI]: 0.93 [0.87-0.98] and 0.04 [0-0.61], respectively). In conclusion, we provide evidence that the CD41-CD61 expression in sEVs reflects the CMR-assessed ischemic damage after STEMI. This finding paves the way for the development of a new strategy for the timely identification of high-risk patients and their treatment optimization.
Collapse
Affiliation(s)
- Marta Zarà
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Andrea Baggiano
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Patrizia Amadio
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Jeness Campodonico
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Sebastiano Gili
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Andrea Annoni
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Gianluca De Dona
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | | | - Francesco Cilia
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Alberto Formenti
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Laura Fusini
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20156 Milan, Italy
| | - Cristina Banfi
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Paola Gripari
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | | | | | - Mattia Chiesa
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Riccardo Maragna
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Francesca Marchetti
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Marco Penso
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Luigi Tassetti
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Alessandra Volpe
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Alice Bonomi
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
| | - Gianluca Pontone
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (A.B.); (G.M.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | | |
Collapse
|
16
|
Lai Z, Liang J, Zhang J, Mao Y, Zheng X, Shen X, Lin W, Xu G. Exosomes as a delivery tool of exercise-induced beneficial factors for the prevention and treatment of cardiovascular disease: a systematic review and meta-analysis. Front Physiol 2023; 14:1190095. [PMID: 37841310 PMCID: PMC10570527 DOI: 10.3389/fphys.2023.1190095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023] Open
Abstract
Exercise-derived exosomes have been identified as novel players in mediating cell-to-cell communication in the beneficial effects of improving cardiovascular disease (CVD). This review aimed to systematically investigate exosomes as delivery tools for the benefits of exercise in the prevention and treatment of CVD and summarize these outcomes with an overview of their therapeutic implications. Among the 1417 articles obtained in nine database searches (PubMed, EBSCO, Embase, Web of Science, CENTRAL, Ovid, Science Direct, Scopus, and Wiley), 12 articles were included based on eligibility criteria. The results indicate that exercise increases the release of exosomes, increasing exosomal markers (TSG101, CD63, and CD81) and exosome-carried miRNAs (miR-125b-5p, miR-122-5p, miR-342-5p, miR-126, miR-130a, miR-138-5p, and miR-455). These miRNAs mainly regulate the expression of MAPK, NF-kB, VEGF, and Caspase to protect the cardiovascular system. Moreover, the outcome indicators of myocardial apoptosis and myocardial infarction volume are significantly reduced following exercise-induced exosome release, and angiogenesis, microvessel density and left ventricular ejection fraction are significantly increased, as well as alleviating myocardial fibrosis following exercise-induced exosome release. Collectively, these results further confirm that exercise-derived exosomes have a beneficial role in potentially preventing and treating CVD and support the use of exercise-derived exosomes in clinical settings.
Collapse
Affiliation(s)
- Zhijie Lai
- Department of School of Physical Education, Guangzhou College of Commerce, Guangzhou, China
| | - Jiling Liang
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jingfeng Zhang
- College of Humanities Education, Foshan University, Foshan, China
| | - Yuheng Mao
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Xinguang Zheng
- Department of School of Physical Education, Guangzhou College of Commerce, Guangzhou, China
| | - Xiang Shen
- Department of School of Physical Education, Guangzhou College of Commerce, Guangzhou, China
| | - Wentao Lin
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
- Department of School of Physical Education, Zhuhai College of Science and Techology, Zhuhai, China
| | - Guoqin Xu
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou, China
| |
Collapse
|
17
|
Mu N, Li J, Zeng L, You J, Li R, Qin A, Liu X, Yan F, Zhou Z. Plant-Derived Exosome-Like Nanovesicles: Current Progress and Prospects. Int J Nanomedicine 2023; 18:4987-5009. [PMID: 37693885 PMCID: PMC10492547 DOI: 10.2147/ijn.s420748] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023] Open
Abstract
Exosomes are small extracellular vesicles, ranging in size from 30-150nm, which can be derived from various types of cells. In recent years, mammalian-derived exosomes have been extensively studied and found to play a crucial role in regulating intercellular communication, thereby influencing the development and progression of numerous diseases. Traditional Chinese medicine has employed plant-based remedies for thousands of years, and an increasing body of evidence suggests that plant-derived exosome-like nanovesicles (PELNs) share similarities with mammalian-derived exosomes in terms of their structure and function. In this review, we provide an overview of recent advances in the study of PELNs and their potential implications for human health. Specifically, we summarize the roles of PELNs in respiratory, digestive, circulatory, and other diseases. Furthermore, we have extensively investigated the potential shortcomings and challenges in current research regarding the mechanism of action, safety, administration routes, isolation and extraction methods, characterization and identification techniques, as well as drug-loading capabilities. Based on these considerations, we propose recommendations for future research directions. Overall, our review highlights the potential of PELNs as a promising area of research, with broad implications for the treatment of human diseases. We anticipate continued interest in this area and hope that our summary of recent findings will stimulate further exploration into the implications of PELNs for human health.
Collapse
Affiliation(s)
- Nai Mu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Jie Li
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Li Zeng
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Juan You
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Rong Li
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Anquan Qin
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Xueping Liu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
| | - Fang Yan
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Zheng Zhou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
18
|
罗 欣, 苏 迪, 卢 娜, 万 源, 刘 桂, 罗 忠. [Physicochemical properties of a novel chiral self-assembling peptide R-LIFE-1 and its controlled release to exosomes]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2023; 40:770-777. [PMID: 37666768 PMCID: PMC10477380 DOI: 10.7507/1001-5515.202207056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 06/13/2023] [Indexed: 09/06/2023]
Abstract
This research aims to investigate the encapsulation and controlled release effect of the newly developed self-assembling peptide R-LIFE-1 on exosomes. The gelling ability and morphological structure of the chiral self-assembling peptide (CSAP) hydrogel were examined using advanced imaging techniques, including atomic force microscopy, transmission electron microscopy, and cryo-scanning electron microscopy. The biocompatibility of the CSAP hydrogel was assessed through optical microscopy and fluorescent staining. Exosomes were isolated via ultrafiltration, and their quality was evaluated using Western blot analysis, nanoparticle tracking analysis, and transmission electron microscopy. The controlled release effect of the CSAP hydrogel on exosomes was quantitatively analyzed using laser confocal microscopy and a BCA assay kit. The results revealed that the self-assembling peptide R-LIFE-1 exhibited spontaneous assembly in the presence of various ions, leading to the formation of nanofibers. These nanofibers were cross-linked, giving rise to a robust nanofiber network structure, which further underwent cross-linking to generate a laminated membrane structure. The nanofibers possessed a large surface area, allowing them to encapsulate a substantial number of water molecules, thereby forming a hydrogel material with high water content. This hydrogel served as a stable spatial scaffold and loading matrix for the three-dimensional culture of cells, as well as the encapsulation and controlled release of exosomes. Importantly, R-LIFE-1 demonstrated excellent biocompatibility, preserving the growth of cells and the biological activity of exosomes. It rapidly formed a three-dimensional network scaffold, enabling the stable loading of cells and exosomes, while exhibiting favorable biocompatibility and reduced cytotoxicity. In conclusion, the findings of this study support the notion that R-LIFE-1 holds significant promise as an ideal tissue engineering material for tissue repair applications.
Collapse
Affiliation(s)
- 欣怡 罗
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| | - 迪 苏
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| | - 娜 卢
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| | - 源 万
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| | - 桂岑 刘
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| | - 忠礼 罗
- 重庆医科大学 基础医学院 分子医学与肿瘤研究中心(重庆 400016)Molecular Medicine and Cancer Research Center, Collage of Basic Medicine, Chongqing Medical University, Chongqing 400016, P. R. China
| |
Collapse
|
19
|
Chen XW, Li QW, Wang H. Sequencing and bioinformatics analysis of miRNA from rat endplate chondrogenic exosomes. Exp Ther Med 2023; 25:267. [PMID: 37206570 PMCID: PMC10189748 DOI: 10.3892/etm.2023.11966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/16/2023] [Indexed: 05/21/2023] Open
Abstract
Exosomes have a key role in various diseases, such as arthritis, heart disease and respiratory disease. Exosomes from various sources have also been indicated to improve intervertebral disc degeneration. However, the role of endplate chondrogenic exosomes in intervertebral disc degeneration has remained largely elusive. The aim of the present study was to compare exosomal microRNA (miRNA) expression patterns in endplate chondrocytes before and after degeneration, and their potential roles in the pathogenesis of intervertebral disc degeneration (IVDD). Endplate chondrocytes were extracted from rats and cultured to obtain pre- and post-degeneration chondrocytes. Exosomes were obtained from the chondrocytes by centrifugation. The two groups of exosomes were subjected to small RNA sequencing, miRNA identification, novel miRNA prediction, quantitative analysis of miRNA expression and differentially expressed (DE) miRNA screening, in addition to miRNA target gene (TG) prediction and TG functional annotation and enrichment analysis. The percentage of miRNAs isolated from the exosomes before and after degeneration was found to differ. A total of 58 DE miRNAs were analyzed, the expression levels of which were significantly different post-degeneration compared with pre-degeneration. Cell experiments were also performed, in which the exosomes were co-cultured with nucleus pulposus (NP) cells. The results indicated that the chondrocyte-derived exosomes were taken up by the NP cells and influenced the expression of aggrecan and collagen 1A and 2A, suggesting that they may inhibit IVDD via their action on NP cells. The specific miRNAs present in exosomes during IVDD may be used to develop new targets for the treatment and diagnosis of this condition. DE exosomal miRNAs derived from endplate cartilage pre- and post-degeneration may be associated with the risk of IVDD and could help to distinguish patients with IVDD. Furthermore, the expression of certain miRNAs may be associated with disease progression, which may contribute to understanding the pathophysiology of IVDD from an epigenetic perspective.
Collapse
Affiliation(s)
- Xue-Wu Chen
- Department of Spinal Orthopedics, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Qiu-Wei Li
- Department of Spinal Orthopedics, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Hong Wang
- Department of Spinal Orthopedics, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
- Correspondence to: Professor Hong Wang, Department of Spinal Orthopedics, Yijishan Hospital of Wannan Medical College, 2 Zheshan West Road, Jinghu, Wuhu, Anhui 241000, P.R. China
| |
Collapse
|
20
|
Ding N, Yin Z, Chen C. Targeting non-coding RNAs in sEVs: The biological functions and potential therapeutic strategy of diabetic cardiomyopathy. Biomed Pharmacother 2023; 163:114836. [PMID: 37156118 DOI: 10.1016/j.biopha.2023.114836] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/15/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as abnormalities in myocardial structure and function in the setting of diabetes and in the absence of cardiovascular diseases, such as coronary artery disease, hypertension, and valvular heart disease. DCM is one of the leading causes of mortality in patients with diabetes. However, the underlying pathogenesis of DCM has not been fully elucidated. Recent studies have revealed that non-coding RNAs (ncRNAs) in small extracellular vesicles (sEVs) are closely associated with DCM and may act as potential diagnostic and therapeutic targets. Here, we introduced the role of sEV-ncRNAs in DCM, summarized the current therapeutic advancements and limitations of sEV-related ncRNAs against DCM, and discussed their potential improvement.
Collapse
Affiliation(s)
- Nan Ding
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zhongwei Yin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
21
|
Chen LY, Kao TW, Chen CC, Niaz N, Lee HL, Chen YH, Kuo CC, Shen YA. Frontier Review of the Molecular Mechanisms and Current Approaches of Stem Cell-Derived Exosomes. Cells 2023; 12:cells12071018. [PMID: 37048091 PMCID: PMC10093591 DOI: 10.3390/cells12071018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Exosomes are effective therapeutic vehicles that may transport their substances across cells. They are shown to possess the capacity to affect cell proliferation, migration, anti-apoptosis, anti-scarring, and angiogenesis, via the action of transporting molecular components. Possessing immense potential in regenerative medicine, exosomes, especially stem cell-derived exosomes, have the advantages of low immunogenicity, minimal invasiveness, and broad clinical applicability. Exosome biodistribution and pharmacokinetics may be altered, in response to recent advancements in technology, for the purpose of treating particular illnesses. Yet, prior to clinical application, it is crucial to ascertain the ideal dose and any potential negative consequences of an exosome. This review focuses on the therapeutic potential of stem cell-derived exosomes and further illustrates the molecular mechanisms that underpin their potential in musculoskeletal regeneration, wound healing, female infertility, cardiac recovery, immunomodulation, neurological disease, and metabolic regulation. In addition, we provide a summary of the currently effective techniques for isolating exosomes, and describe the innovations in biomaterials that improve the efficacy of exosome-based treatments. Overall, this paper provides an updated overview of the biological factors found in stem cell-derived exosomes, as well as potential targets for future cell-free therapeutic applications.
Collapse
|
22
|
Chen C, Zhang Z, Gu X, Sheng X, Xiao L, Wang X. Exosomes: New regulators of reproductive development. Mater Today Bio 2023; 19:100608. [PMID: 36969697 PMCID: PMC10034510 DOI: 10.1016/j.mtbio.2023.100608] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
Exosomes are a subtype of extracellular vesicles (EVs) with a size range between 30 and 150 nm, which can be released by the majority of cell types and circulate in body fluid. They function as a long-distance cell-to-cell communication mechanism that modulates the gene expression profile and fate of target cells. Increasing evidence has indicated exosomes' central role in regulating various complex reproductive processes. However, to our knowledge, a review that focally and vividly describes the role of exosomes in reproductive development is still lacking. This review highlights our knowledge about the contribution of exosomes to early mammalian reproduction, such as gametogenesis, fertilization, early embryonic development, implantation, placentation and pregnancy. The discussion is primarily drawn from literature pertaining to the mammalian lineage with emphasis on the roles of exosomes in human reproduction and laboratory and livestock models.
Collapse
|
23
|
Nassal DM, Shaheen R, Patel NJ, Yu J, Leahy N, Bibidakis D, Parinandi NL, Hund TJ. Spectrin-Based Regulation of Cardiac Fibroblast Cell-Cell Communication. Cells 2023; 12:748. [PMID: 36899883 PMCID: PMC10001335 DOI: 10.3390/cells12050748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Cardiac fibroblasts (CFs) maintain the fibrous extracellular matrix (ECM) that supports proper cardiac function. Cardiac injury induces a transition in the activity of CFs to promote cardiac fibrosis. CFs play a critical role in sensing local injury signals and coordinating the organ level response through paracrine communication to distal cells. However, the mechanisms by which CFs engage cell-cell communication networks in response to stress remain unknown. We tested a role for the action-associated cytoskeletal protein βIV-spectrin in regulating CF paracrine signaling. Conditioned culture media (CCM) was collected from WT and βIV-spectrin deficient (qv4J) CFs. WT CFs treated with qv4J CCM showed increased proliferation and collagen gel compaction compared to control. Consistent with the functional measurements, qv4J CCM contained higher levels of pro-inflammatory and pro-fibrotic cytokines and increased concentration of small extracellular vesicles (30-150 nm diameter, exosomes). Treatment of WT CFs with exosomes isolated from qv4J CCM induced a similar phenotypic change as that observed with complete CCM. Treatment of qv4J CFs with an inhibitor of the βIV-spectrin-associated transcription factor, STAT3, decreased the levels of both cytokines and exosomes in conditioned media. This study expands the role of the βIV-spectrin/STAT3 complex in stress-induced regulation of CF paracrine signaling.
Collapse
Affiliation(s)
- Drew M. Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Shaheen
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nehal J. Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jane Yu
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nick Leahy
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Dimitra Bibidakis
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Narasimham L. Parinandi
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas J. Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Almaghrbi H, Giordo R, Pintus G, Zayed H. Non-coding RNAs as biomarkers of myocardial infarction. Clin Chim Acta 2023; 540:117222. [PMID: 36627010 DOI: 10.1016/j.cca.2023.117222] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
Non-coding RNAs (ncRNAs) encompass a family of ubiquitous RNA molecules that lack protein-coding potential and have tissue-specific expression. A significant body of evidence indicates that ncRNA's aberrant expression plays a critical role in disease onset and development. NcRNAs' biochemical characteristics such as disease-associated concentration changes, structural stability, and high abundance in body fluids make them promising prognostic and diagnostic biomarkers. Myocardial infarction (MI) is a leading cause of mortality worldwide. Acute myocardial infarction (AMI), the term in use to describe MI's early phase, is generally diagnosed by physical examination, electrocardiogram (ECG), and the presence of specific biomarkers. In this regard, compared to standard MI biomarkers, such as the cardiac troponin isoforms (cTnT & cTnI) and the Creatinine Kinase (CK), ncRNAs appears to provide better sensitivity and specificity, ensuring a rapid and correct diagnosis, an earlier treatment, and consequently a good prognosis for the patients. This review aims to summarize and discuss the most promising and recent data on the potential clinical use of circulating ncRNAs as MI biomarkers. Specifically, we focused primarily on miRNAs and lncRNAs, highlighting their significant specificity and sensitivity, discussing their limitations, and suggesting possible overcoming approaches.
Collapse
Affiliation(s)
- Heba Almaghrbi
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, 505055 Dubai, United Arab Emirates
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100 Sassari, Italy; Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates.
| | - Hatem Zayed
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
25
|
Zhu Y, Wang S, Chen X. Extracellular Vesicles and Ischemic Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:57-68. [PMID: 37603272 DOI: 10.1007/978-981-99-1443-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Characterized by coronary artery obstruction or stenosis, ischemic cardiovascular diseases as advanced stages of coronary heart diseases commonly lead to left ventricular aneurysm, ventricular septal defect, and mitral insufficiency. Extracellular vesicles (EVs) secreted by diverse cells in the body exert roles in cell-cell interactions and intrinsic cellular regulations. With a lipid double-layer membrane and biological components such as DNA, protein, mRNA, microRNAs (miRNA), and siRNA inside, the EVs function as paracrine signaling for the pathophysiology of ischemic cardiovascular diseases and maintenance of the cardiac homeostasis. Unlike stem cell transplantation with the potential tumorigenicity and immunogenicity, the EV-based therapeutic strategy is proposed to satisfy the demand for cardiac repair and regeneration while the circulating EVs detected by a noninvasive approach can act as precious biomarkers. In this chapter, we extensively summarize the cardioprotective functions of native EVs and bioengineered EVs released from stem cells, cardiomyocytes, cardiac progenitor cells (CPCs), endothelial cells, fibroblast, smooth muscle cells, and immune cells. In addition, the potential of EVs as robust molecule biomarkers is discussed for clinical diagnosis of ischemic cardiovascular disease, attributed to the same pathology of EVs as that of their origin. Finally, we highlight EV-based therapy as a biocompatible alternative to direct cell-based therapy for ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Siqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xuerui Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China.
| |
Collapse
|
26
|
Chen L, Yang J, Xu G, Wu Y. Potential Value and Application of Liquid Biopsy in Tumor, Neurodegeneration, and Muscle Degenerative Diseases. Methods Mol Biol 2023; 2695:317-335. [PMID: 37450129 DOI: 10.1007/978-1-0716-3346-5_22] [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: 07/18/2023]
Abstract
Liquid biopsy provides a promising alternative for the detection of disease-specific markers due to its superior noninvasive and original tissue representativeness. Liquid biopsies have a wide range of health and disease applications involving components ranging from circulating cells to acellular nucleic acid molecules and other metabolites. Here, we review the different components of liquid biopsy and investigate the most advanced noninvasive methods for detecting these components as well as their existing problems and trends. In particular, we emphasize the importance of analyzing liquid biopsy data from extracellular vesicles and small nucleic acids in neurological and muscle degeneration, with the aim of using this technique to enhance personalized healthcare. Although previous reviews have focused on cancer, this review mainly emphasizes the potential application of extracellular vesicles and microRNAs in liquid biopsy in neurodegeneration and muscle degeneration.
Collapse
Affiliation(s)
- Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China
| | - Jun Yang
- Jianghan University Library, Wuhan, Hubei, People's Republic of China
| | - Guodong Xu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
27
|
Wronska A. The Role of microRNA in the Development, Diagnosis, and Treatment of Cardiovascular Disease: Recent Developments. J Pharmacol Exp Ther 2023; 384:123-132. [PMID: 35779862 DOI: 10.1124/jpet.121.001152] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/13/2023] Open
Abstract
Since their discovery in 1993, microRNAs (miRNAs) have emerged as important regulators of many crucial cellular processes, and their dysregulation have been shown to contribute to multiple pathologic conditions, including cardiovascular disease (CVD). miRNAs have been found to regulate the expression of various genes involved in cardiac development and function and in the development and progression of CVD. Many miRNAs are master regulators fine-tuning the expression of multiple, often interrelated, genes involved in inflammation, apoptosis, fibrosis, senescence, and other processes crucial for the development of different forms of CVD. This article presents a review of recent developments in our understanding of the role of miRNAs in the development of CVD and surveys their potential applicability as therapeutic targets and biomarkers to facilitate CVD diagnosis, prognosis, and treatment. There are currently multiple potential miRNA-based therapeutic agents in different stages of development, which can be grouped into two classes: miRNA mimics (replicating the sequence and activity of their corresponding miRNAs) and antagomiRs (antisense inhibitors of specific miRNAs). However, in spite of promising preliminary data and our ever-increasing knowledge about the mechanisms of action of specific miRNAs, miRNA-based therapeutics and biomarkers have yet to be approved for clinical applications. SIGNIFICANCE STATEMENT: Over the last few years microRNAs have emerged as crucial, specific regulators of the cardiovascular system and in the development of cardiovascular disease, by posttranscriptional regulation of their target genes. The minireview presents the most recent developments in this area of research, including the progress in diagnostic and therapeutic applications of microRNAs. microRNAs seem very promising candidates for biomarkers and therapeutic agents, although some challenges, such as efficient delivery and unwanted effects, need to be resolved.
Collapse
Affiliation(s)
- Anetta Wronska
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| |
Collapse
|
28
|
Su X, Shen Y, Kim IM, Weintraub NL, Hamrick M, Tang Y. Extracellular Vesicles for Muscle Atrophy Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:119-126. [PMID: 37603276 DOI: 10.1007/978-981-99-1443-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Skeletal muscle atrophy is a progressive chronic disease associated with various conditions, such as aging, cancer, and muscular dystrophy. Interleukin-6 (IL-6) is highly correlated with or plays a crucial role in inducing skeletal muscle atrophy. Extracellular vehicles (EVs), including exosomes, mediate cell-cell communication, and alterations in the genetic material contained in EVs during muscle atrophy may impair muscle cell signaling. Transplantation of muscle progenitor cell-derived EVs (MPC-EVs) is a promising approach for treating muscle diseases such as Duchenne muscular dystrophy (DMD). Moreover, stem cell-derived EVs with modification of microRNAs (e.g., miR-26 and miR-29) have been reported to attenuate muscle atrophy. Unbiased RNA-Seq analysis suggests that MPC-EVs may exert an inhibitory effect on IL-6 pathway. Here, we review the latest advances concerning the mechanisms of stem cell/progenitor cell-derived EVs in alleviating muscle atrophy, including anti-inflammatory and anti-fibrotic effects. We also discuss the clinical application of EVs in the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Xuan Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-Man Kim
- Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mark Hamrick
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
29
|
Li C, Li W, Pu G, Wu J, Qin F. Exosomes derived from miR-338-3p-modified adipose stem cells inhibited inflammation injury of chondrocytes via targeting RUNX2 in osteoarthritis. J Orthop Surg Res 2022; 17:567. [PMID: 36572886 PMCID: PMC9791748 DOI: 10.1186/s13018-022-03437-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease that is one of the main causes of disability in middle-aged and elderly people. Adipose stem cell (ASC)-derived exosomes (ASC-Exo) could repair cartilage damage and treat OA. MiRNA-338-3p expression was confirmed to play a role in inhibiting proinflammatory cytokines. Herein, we aimed to explore the mechanism by which exosomes derived from miR-338-3p overexpressing ASCs protects chondrocytes from interleukin (IL)-1β-induced chondrocyte change. METHODS Exosomes were extracted from ASCs transfected with miR-338-3p or its antisense inhibitor. The ASC-Exos (miR-338-3p silencing/overexpression) were incubated with IL-1β-induced ATDC5 cells, followed by evaluation of the chondrocyte proliferation, degradation, and inflammation injury. RESULTS In vitro results revealed that ASC-Exos inhibited the expression of prostaglandin E2 (PGE2), IL-6, IL-1β, and TNF-α, as well as promoted the proliferation of ATDC5 cells. Moreover, ASC-Exos inhibited inflammation injury and degradation of ATDC5 cells by transferring miR-338-3p. Luciferase reporter assays showed that RUNX2 was a target gene of miR-338-3p. Additionally, RUNX2 overexpression in ATDC5 cells reversed the protective effect of miR-338-3p on chondrocytes. Taken together, this study demonstrated that exosomes secreted from miR-338-3p-modified ASCs were effective in the repair of IL-1β-induced chondrocyte change by inhibiting RUNX2 expression. CONCLUSIONS Our result provided valuable data for understanding the mechanism of ASC-Exos in OA treatment.
Collapse
Affiliation(s)
- ChunLiang Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Wei Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - GengZang Pu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - JingWen Wu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Feng Qin
- grid.459333.bDepartment of Endocrinology, Qinghai University Affiliated Hospital, Chengxi District, No. 6, Xichuan South Road, Xining, 810006 Qinghai China
| |
Collapse
|
30
|
Three-Dimensional Bio-Printed Cardiac Patch for Sustained Delivery of Extracellular Vesicles from the Interface. Gels 2022; 8:gels8120769. [PMID: 36547293 PMCID: PMC9777613 DOI: 10.3390/gels8120769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac tissue engineering has emerged as a promising strategy to treat infarcted cardiac tissues by replacing the injured region with an ex vivo fabricated functional cardiac patch. Nevertheless, integration of the transplanted patch with the host tissue is still a burden, limiting its clinical application. Here, a bi-functional, 3D bio-printed cardiac patch (CP) design is proposed, composed of a cell-laden compartment at its core and an extracellular vesicle (EV)-laden compartment at its shell for better integration of the CP with the host tissue. Alginate-based bioink solutions were developed for each compartment and characterized rheologically, examined for printability and their effect on residing cells or EVs. The resulting 3D bio-printed CP was examined for its mechanical stiffness, showing an elastic modulus between 4-5 kPa at day 1 post-printing, suitable for transplantation. Affinity binding of EVs to alginate sulfate (AlgS) was validated, exhibiting dissociation constant values similar to those of EVs with heparin. The incorporation of AlgS-EVs complexes within the shell bioink sustained EV release from the CP, with 88% cumulative release compared with 92% without AlgS by day 4. AlgS also prolonged the release profile by an additional 2 days, lasting 11 days overall. This CP design comprises great potential at promoting more efficient patch assimilation with the host.
Collapse
|
31
|
Raghav A, Ashraf H, Jeong GB. Engineered Extracellular Vesicles in Treatment of Type 1 Diabetes Mellitus: A Prospective Review. Biomedicines 2022; 10:3042. [PMID: 36551798 PMCID: PMC9775549 DOI: 10.3390/biomedicines10123042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
Insulin replacement is an available treatment for autoimmune type 1 diabetes mellitus (T1DM). There are multiple limitations in the treatment of autoimmune diseases such as T1DM by immunosuppression using drugs and chemicals. The advent of extracellular vesicle (EV)-based therapies for the treatment of various diseases has attracted much attention to the field of bio-nanomedicine. Tolerogenic nanoparticles can induce immune tolerance, especially in autoimmune diseases. EVs can deliver cargo to specific cells without restrictions. Accordingly, EVs can be used to deliver tolerogenic nanoparticles, including iron oxide-peptide-major histocompatibility complex, polyethylene glycol-silver-2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester, and carboxylated poly (lactic-co-glycolic acid) nanoparticles coupled with or encapsulating an antigen, to effectively treat autoimmune T1DM. The present work highlights the advances in exosome-based delivery of tolerogenic nanoparticles for the treatment of autoimmune T1DM.
Collapse
Affiliation(s)
- Alok Raghav
- Multidisciplinary Research Unit, Sponsored by Department of Health Research, Ministry of Health and Family Welfare, GSVM Medical College, Kanpur 208002, India
| | - Hamid Ashraf
- Rajiv Gandhi Centre for Diabetes and Endocrinology, J.N. Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Goo-Bo Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Gachon University, 155 Getbeol-ro Yeonsu-gu, Incheon 21999, Republic of Korea
| |
Collapse
|
32
|
Zhou F, Li K, Yang K. Adipose-Derived Stem Cell Exosomes and Related microRNAs in Atherosclerotic Cardiovascular Disease. J Cardiovasc Transl Res 2022; 16:453-462. [PMID: 36223051 DOI: 10.1007/s12265-022-10329-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death from noncommunicable diseases worldwide. The pathological development of ASCVD begins with atherosclerosis, followed by the narrowing and occlusion of the vascular lumen and, subsequently, ischemic necrosis in coronary arteries. Preventing atherosclerosis development and delaying ischemia progression may be effective ways of pre-diagnosing and treating ASCVD. Studies have demonstrated that exosomes from adipose-derived stem cells play an increasingly important role in basic research on cardiovascular diseases in terms of the impact of macrophage polarization and the endothelial, anti-apoptosis, and angiogenesis effects. The related microRNAs play a significant role in ASCVD. This study was novel in reviewing the role of exosomes from adipose-derived stem cells and related microRNAs in ASCVD. Therapeutic potentials of adipose-derived stem cell exosomes in terms of their impact on macrophage polarization, endothelial effect, anti-apoptosis intervention, and angiogenesis.
Collapse
Affiliation(s)
- Fan Zhou
- Department of Cardiology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434023, China
| | - Ke Li
- Department of Cardiology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434023, China
| | - Keping Yang
- Department of Cardiology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434023, China.
| |
Collapse
|
33
|
Mao L, Li YD, Chen RL, Li G, Zhou XX, Song F, Wu C, Hu Y, Hong YX, Dang X, Li GR, Wang Y. Heart-targeting exosomes from human cardiosphere-derived cells improve the therapeutic effect on cardiac hypertrophy. J Nanobiotechnology 2022; 20:435. [PMID: 36195937 PMCID: PMC9531502 DOI: 10.1186/s12951-022-01630-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/09/2022] [Indexed: 12/04/2022] Open
Abstract
Exosomes of human cardiosphere-derived cells (CDCs) are very promising for treating cardiovascular disorders. However, the current challenge is inconvenient delivery methods of exosomes for clinical application. The present study aims to explore the potential to enhance the therapeutic effect of exosome (EXO) from human CDCs to myocardial hypertrophy. A heart homing peptide (HHP) was displayed on the surface of exosomes derived from CDCs that were forced to express the HHP fused on the N-terminus of the lysosomal-associated membrane protein 2b (LAMP2b). The cardiomyocyte-targeting capability of exosomes were analyzed and their therapeutic effects were evaluated in a mouse model of myocardial hypertrophy induced by transverse aorta constriction (TAC). The molecular mechanisms of the therapeutic effects were dissected in angiotensin II-induced neonatal rat cardiomyocyte (NRCMs) hypertrophy model using a combination of biochemistry, immunohistochemistry and molecular biology techniques. We found that HHP-exosomes (HHP-EXO) accumulated more in mouse hearts after intravenous delivery and in cultured NRCMs than control exosomes (CON-EXO). Cardiac function of TAC mice was significantly improved with intravenous HHP-EXO administration. Left ventricular hypertrophy was reduced more by HHP-EXO than CON-EXO via inhibition of β-MHC, BNP, GP130, p-STAT3, p-ERK1/2, and p-AKT. Similar results were obtained in angiotensin II-induced hypertrophy of NRCMs, in which the beneficial effects of HHP-EXO were abolished by miRNA-148a inhibition. Our results indicate that HHP-EXO preferentially target the heart and improve the therapeutic effect of CDCs-exosomes on cardiac hypertrophy. The beneficial therapeutic effect is most likely attributed to miRNA-148a-mediated suppression of GP130, which in turn inhibits STAT3/ERK1/2/AKT signaling pathway, leading to improved cardiac function and remodeling.
Collapse
Affiliation(s)
- Liang Mao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Yun-Da Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ruo-Lan Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xiao-Xia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Fei Song
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yu Hu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xitong Dang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.,Nanjing Amaigh Pharma Limited, Nanjing, 210032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
34
|
Exosome mediated Tom40 delivery protects against hydrogen peroxide-induced oxidative stress by regulating mitochondrial function. PLoS One 2022; 17:e0272511. [PMID: 35951602 PMCID: PMC9371349 DOI: 10.1371/journal.pone.0272511] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of neurodegeneration. The expression level of Tom40, a crucial mitochondrial membrane protein, is significantly reduced in neurodegenerative disease subjects. Tom40 overexpression studies have shown to protect the neurons against oxidative stress by improving mitochondrial function. Thus, successful delivery of Tom40 protein to the brain could lead to a novel therapy for neurodegenerative diseases. However, delivering protein to the cell may be difficult. Especially the blood-brain barrier (BBB) is a big hurdle to clear in order to deliver the protein to the brain. In the current study, we engineered exosomes, which are the extracellular vesicles of endosomal origin, and able to cross BBB as delivery vehicles packing human Tom40. We found Tom40 protein delivery by the exosome successfully protected the cells against hydrogen peroxide-induced oxidative stress. This result suggests that exosome-mediated delivery of Tom40 may potentially be useful in restoring mitochondrial functions and alleviating oxidative stress in neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases.
Collapse
|
35
|
Chandrasekera D, Katare R. Exosomal microRNAs in diabetic heart disease. Cardiovasc Diabetol 2022; 21:122. [PMID: 35778763 PMCID: PMC9250231 DOI: 10.1186/s12933-022-01544-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Diabetes is a metabolic disorder that affects millions of people worldwide. Diabetic heart disease (DHD) comprises coronary artery disease, heart failure, cardiac autonomic neuropathy, peripheral arterial disease, and diabetic cardiomyopathy. The onset and progression of DHD have been attributed to molecular alterations in response to hyperglycemia in diabetes. In this context, microRNAs (miRNAs) have been demonstrated to have a significant role in the development and progression of DHD. In addition to their effects on the host cells, miRNAs can be released into circulation after encapsulation within the exosomes. Exosomes are extracellular nanovesicles ranging from 30 to 180 nm in diameter secreted by all cell types. They carry diverse cargos that are altered in response to various conditions in their parent cells. Exosomal miRNAs have been extensively studied in recent years due to their role and therapeutic potential in DHD. This review will first provide an overview of exosomes, their biogenesis and function, followed by the role of exosomes in cardiovascular disease and then focuses on the known role of exosomes and associated miRNAs in DHD.
Collapse
Affiliation(s)
- Dhananjie Chandrasekera
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, 270, Great King Street, Dunedin, New Zealand.
| | - Rajesh Katare
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, 270, Great King Street, Dunedin, New Zealand.
| |
Collapse
|
36
|
Wang J, Ma W, Si C, Zhang M, Qian W, Park G, Zhou B, Luo D. Exosome-mediated miR-4655–3p contributes to UV radiation-induced bystander effects. Exp Cell Res 2022; 418:113247. [DOI: 10.1016/j.yexcr.2022.113247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/04/2022] [Accepted: 06/05/2022] [Indexed: 11/04/2022]
|
37
|
Hoffman JR, Park HJ, Bheri S, Jayaraman AR, Davis ME. Comparative computational RNA analysis of cardiac-derived progenitor cells and their extracellular vesicles. Genomics 2022; 114:110349. [PMID: 35346780 PMCID: PMC9510608 DOI: 10.1016/j.ygeno.2022.110349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/18/2022] [Accepted: 03/23/2022] [Indexed: 01/14/2023]
Abstract
Stem/progenitor cells, including cardiac-derived c-kit+ progenitor cells (CPCs), are under clinical evaluation for treatment of cardiac disease. Therapeutic efficacy of cardiac cell therapy can be attributed to paracrine signaling and the release of extracellular vesicles (EVs) carrying diverse cargo molecules. Despite some successes and demonstrated safety, large variation in cell populations and preclinical/clinical outcomes remains a problem. Here, we investigated this variability by sequencing coding and non-coding RNAs of CPCs and CPC-EVs from 30 congenital heart disease patients and used machine learning methods to determine potential mechanistic insights. CPCs retained RNAs related to extracellular matrix organization and exported RNAs related to various signaling pathways to CPC-EVs. CPC-EVs are enriched in miRNA clusters related to cell proliferation and angiogenesis. With network analyses, we identified differences in non-coding RNAs which give insight into age-dependent functionality of CPCs. By taking a quantitative computational approach, we aimed to uncover sources of CPC cell therapy variability.
Collapse
Affiliation(s)
- Jessica R. Hoffman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arun R. Jayaraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA.,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA.,Corresponding author at: Professor of Biomedical Engineering, 2015 Uppergate Drive, 310, Atlanta, GA 30322, USA,
| |
Collapse
|
38
|
Mesenchymal stem cell-derived exosomal microRNA-182-5p alleviates myocardial ischemia/reperfusion injury by targeting GSDMD in mice. Cell Death Dis 2022; 8:202. [PMID: 35422485 PMCID: PMC9010441 DOI: 10.1038/s41420-022-00909-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 01/03/2023]
Abstract
Recent evidence indicates that exosomes derived from mesenchymal stem cells (MSCs) confer protective effects against myocardial ischemia/reperfusion (I/R) injury. Exosomes are carriers of potentially protective endogenous molecules, including microRNAs (miRNAs/miRs). The current study set out to test the effects of transferring miR-182-5p from MSC-derived exosomes into myocardial cells on myocardial I/R injury. First, an I/R mouse model was developed by left anterior descending coronary artery occlusion, and myocardial cells were exposed to hypoxia/reoxygenation (H/R) for in vitro I/R model establishment. Loss- and gain-of-function experiments of miR-182-5p and GSDMD were conducted to explore the effects of miR-182-5p via MSC-derived exosomes on cell pyroptosis and viability. GSDMD was robustly expressed in I/R-injured myocardial tissues and H/R-exposed myocardial cells. GSDMD upregulation promoted H/R-induced myocardial cell pyroptosis and reduced viability, corresponding to increased lactate dehydrogenase release, reactive oxygen species production, and pyroptosis. A luciferase assay demonstrated GSDMD as a target of miR-182-5p. In addition, exosomal miR-182-5p was found to diminish GSDMD-dependent cell pyroptosis and inflammation induced by H/R. Furthermore, MSC-derived exosomes carrying miR-182-5p improved cardiac function and reduced myocardial infarction, accompanied with reduced inflammation and cell pyroptosis in vivo. Taken together, our findings suggest a cardioprotective effect of exosomal miR-182-5p against myocardial I/R injury, shedding light on an attractive therapeutic strategy.
Collapse
|
39
|
Li Z, Huo X, Chen K, Yang F, Tan W, Zhang Q, Yu H, Li C, Zhou D, Chen H, Zhao B, Wang Y, Chen Z, Du X. Profilin 2 and Endothelial Exosomal Profilin 2 Promote Angiogenesis and Myocardial Infarction Repair in Mice. Front Cardiovasc Med 2022; 9:781753. [PMID: 35479278 PMCID: PMC9036097 DOI: 10.3389/fcvm.2022.781753] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, wherein myocardial infarction (MI) is the most dangerous one. Promoting angiogenesis is a prospective strategy to alleviate MI. Our previous study indicated that profilin 2 (PFN2) may be a novel target associated with angiogenesis. Further results showed higher levels of serum PFN2 and exosomal PFN2 in patients, mice, and pigs with MI. In this study, we explored whether PFN2 and endothelial cell (EC)-derived exosomal PFN2 could increase angiogenesis and be beneficial for the treatment of MI. Serum PFN2, exosomes, and exosomal PFN2 were elevated in rats with MI. PFN2 and exosomes from PFN2-overexpressing ECs (OE-exo) enhanced EC proliferation, migration, and tube formation ability. OE-exo also significantly increased the vessel number in zebrafish and protected the ECs from inflammatory injury. Moreover, OE-exo-treated mice with MI showed improvement in motor ability, ejection fraction, left ventricular shortening fraction, and left ventricular mass, as well as increased vessel numbers in the MI location, and decreased infarction volume. Mechanistically, PI3K might be the upstream regulator of PFN2, while ERK might be the downstream regulator in the PI3K-PFN2-ERK axis. Taken together, our findings demonstrate that PFN2 and exosomal PFN2 promote EC proliferation, migration, and tube formation through the PI3K-PFN2-ERK axis. Exosomal PFN2 may be a valuable target in the repair of MI injury via angiogenesis.
Collapse
Affiliation(s)
- Zhenkun Li
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xueyun Huo
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Dalian, China
| | - Fenghua Yang
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Weijiang Tan
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Qi Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haixu Yu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Changlong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
| | - Deshan Zhou
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
| | - Hao Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhenwen Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Zhenwen Chen
| | - Xiaoyan Du
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- *Correspondence: Xiaoyan Du
| |
Collapse
|
40
|
Joladarashi D, Kishore R. Mesenchymal Stromal Cell Exosomes in Cardiac Repair. Curr Cardiol Rep 2022; 24:405-417. [PMID: 35092595 PMCID: PMC9885380 DOI: 10.1007/s11886-022-01660-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 02/01/2023]
Abstract
PURPOSE OF THE REVIEW Mesenchymal stromal cells (MSCs) are considered an attractive option for cell-based therapy because of their immune-privileged phenotype and paracrine activity. Substantial preclinical evidence indicates that MSC exosomes recapitulate MSC cellular function in cardiac regeneration and repair. Therefore, in this review, we briefly discuss the latest research progress of MSC exosomes in cardiac repair and regeneration. RECENT FINDINGS The recent revolutionary advance in controlling the contents of the exosomes by manipulating parental cells through bioengineering methods to alter specific signaling pathways in ischemic myocardium has proven to be beneficial in the treatment of heart failure. MSC Exosomes appear to be leading candidates to treat myocardial infarction and subsequent heart failure by carrying rich cargo from their parental cells. However, more clinical and pre-clinical studies on MSC exosomes will be required to confirm the beneficial effect to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Darukeshwara Joladarashi
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, MERB-953, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, MERB-953, 3500 N Broad Street, Philadelphia, PA 19140, USA,Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
41
|
Lin Z, Wong A, Alam S. Extraction of Exosomes and Exosomal miRNA from Mesenchymal Stem Cells. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2455:333-341. [PMID: 35213005 DOI: 10.1007/978-1-0716-2128-8_25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Exosomes, derived from stem cells, have great promise in regenerative medicine due to their capabilities of ameliorating inflammation, preventing tissue damage and promoting healing, which in part are associated with the exosomal RNA/miRNA. The application of mesenchymal stem cell exosomes in treating hepatic disorders including nonalcoholic fatty liver disease has drawn much attention. In this chapter, we describe our experience in culturing human mesenchymal stem cells and isolating their exosomes from culture medium through ultracentrifugation. Methods to extract exosomal RNA/miRNA are also discussed.
Collapse
Affiliation(s)
- Zhao Lin
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA.
| | - Andrew Wong
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Shiekh Alam
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
42
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles: Immunomodulatory Effects and Potential Applications in Intervertebral Disc Degeneration. Stem Cells Int 2022; 2022:7538025. [PMID: 35222648 PMCID: PMC8881131 DOI: 10.1155/2022/7538025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 11/25/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc (IVD) degenerative disease is a common health problem worldwide. Administration of mesenchymal stem cells (MSCs) in intervertebral disc degeneration (IVDD) has been widely explored in recent years. However, transplantation of MSCs is restricted by several factors. Currently, paracrine signaling is one of the main mechanisms by which MSCs play a therapeutic role in disc regeneration. Extracellular vehicles (EVs) are the main paracrine products of MSCs. They show great potential as an effective alternative to MSCs and play immunomodulation roles such as anti-inflammatory effects, antioxidative stress, antiapoptosis, and antiextracellular matrix (ECM) degradation during treatment of IVDD. This review focuses on the immunomodulatory effect of MSC EVs and their potential applications.
Collapse
|
43
|
Chen L, Shi Q, Ma X, Niu Y, Chong M, Ma L. WITHDRAWN: Serum exosomes mediate septic inflammation and liver and kidney injuries by up-regulating the expression of inflammatory factors. Biochem Biophys Res Commun 2022. [DOI: 10.1016/j.bbrc.2022.02.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
44
|
Gupta S, Mazumder P. Exosomes as diagnostic tools. Adv Clin Chem 2022; 110:117-144. [DOI: 10.1016/bs.acc.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Tastan B, Tarakcioglu E, Birinci Y, Park Y, Genc S. Role of Exosomal MicroRNAs in Cell-to-Cell Communication. Methods Mol Biol 2022; 2257:269-292. [PMID: 34432284 DOI: 10.1007/978-1-0716-1170-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Exosomes, a type of extracellular vesicle, are small vesicles (30-100 nm) secreted into extracellular space from almost all types of cells. Exosomes mediate cell-to-cell communication carrying various biologically active molecules including microRNAs. Studies have shown that exosomal microRNAs play fundamental roles in healthy and pathological conditions such as immunity, cancer, and inflammation. In this chapter, we introduce the current knowledge on exosome biogenesis, techniques used in exosome research, and exosomal miRNA and their functions in biological and pathological processes.
Collapse
Affiliation(s)
- Bora Tastan
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Emre Tarakcioglu
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Yelda Birinci
- Department of Molecular Biology and Genetics, Science Faculty, Koç University, Istanbul, Turkey
| | - Yongsoo Park
- Department of Molecular Biology and Genetics, Science Faculty, Koç University, Istanbul, Turkey
| | - Sermin Genc
- Department of Neuroscience, Institute of Health Science, University of Dokuz Eylul, Izmir, Turkey. .,Izmir Biomedicine and Genome Center, Izmir, Turkey.
| |
Collapse
|
46
|
Buja LM, Mitchell RN. Basic pathobiology of cell-based therapies and cardiac regenerative medicine. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
47
|
Jadli AS, Parasor A, Gomes KP, Shandilya R, Patel VB. Exosomes in Cardiovascular Diseases: Pathological Potential of Nano-Messenger. Front Cardiovasc Med 2021; 8:767488. [PMID: 34869682 PMCID: PMC8632805 DOI: 10.3389/fcvm.2021.767488] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVDs) represent a major global health problem, due to their continued high incidences and mortality. The last few decades have witnessed new advances in clinical research which led to increased survival and recovery in CVD patients. Nevertheless, elusive and multifactorial pathophysiological mechanisms of CVD development perplexed researchers in identifying efficacious therapeutic interventions. Search for novel and effective strategies for diagnosis, prevention, and intervention for CVD has shifted research focus on extracellular vesicles (EVs) in recent years. By transporting molecular cargo from donor to recipient cells, EVs modulate gene expression and influence the phenotype of recipient cells, thus EVs prove to be an imperative component of intercellular signaling. Elucidation of the role of EVs in intercellular communications under physiological conditions implied the enormous potential of EVs in monitoring and treatment of CVD. The EVs secreted from the myriad of cells in the cardiovascular system such as cardiomyocytes, cardiac fibroblasts, cardiac progenitor cells, endothelial cells, inflammatory cells may facilitate the communication in physiological and pathological conditions. Understanding EVs-mediated cellular communication may delineate the mechanism of origin and progression of cardiovascular diseases. The current review summarizes exosome-mediated paracrine signaling leading to cardiovascular disease. The mechanistic role of exosomes in cardiovascular disease will provide novel avenues in designing diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Anshul S Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Ananya Parasor
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Karina P Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Ruchita Shandilya
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
48
|
Alam P, Maliken BD, Jones SM, Ivey MJ, Wu Z, Wang Y, Kanisicak O. Cardiac Remodeling and Repair: Recent Approaches, Advancements, and Future Perspective. Int J Mol Sci 2021; 22:ijms222313104. [PMID: 34884909 PMCID: PMC8658114 DOI: 10.3390/ijms222313104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The limited ability of mammalian adult cardiomyocytes to proliferate following an injury to the heart, such as myocardial infarction, is a major factor that results in adverse fibrotic and myocardial remodeling that ultimately leads to heart failure. The continued high degree of heart failure-associated morbidity and lethality requires the special attention of researchers worldwide to develop efficient therapeutics for cardiac repair. Recently, various strategies and approaches have been developed and tested to extrinsically induce regeneration and restoration of the myocardium after cardiac injury have yielded encouraging results. Nevertheless, these interventions still lack adequate success to be used for clinical interventions. This review highlights and discusses both cell-based and cell-free therapeutic approaches as well as current advancements, major limitations, and future perspectives towards developing an efficient therapeutic method for cardiac repair.
Collapse
Affiliation(s)
- Perwez Alam
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Bryan D. Maliken
- Harrington Physician-Scientist Pathway, Department of Internal Medicine, University Hospitals Case Medical Center, Cleveland, OH 44106, USA;
| | - Shannon M. Jones
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Malina J. Ivey
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
- Correspondence: ; Tel.: +1-513-558-2029; Fax: +1-513-584-3892
| |
Collapse
|
49
|
Ma J, Zhang Z, Wang Y, Shen H. Investigation of miR-126-3p loaded on adipose stem cell-derived exosomes for wound healing of full-thickness skin defects. Exp Dermatol 2021; 31:362-374. [PMID: 34694648 DOI: 10.1111/exd.14480] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the function of miR-126-3p loaded on adipose stem cell (ADSC)-derived exosomes (ADSC-Exos) in wound healing of full-thickness skin defects. METHODS ADSCs transfected with miR-126-3p mimic, miR-126-3p inhibitor or pcDNA3.1-PIK3R2, or PKH26-marked ADSC-Exos were cultured with fibroblasts or human umbilical vein endothelial cells (HUVECs). The proliferation and migration rates of fibroblasts and angiogenesis of HUVECs were measured. Rats with full-thickness skin defects were injected with ADSC-Exos or exosomes extracted from ADSCs transfected with miR-126-3p inhibitor and the wound healing rates were measured. The wound bed, collagen deposition and angiogenesis in injured rats were assessed. RESULTS ADSC-Exos could be ingested by fibroblasts and HUVECs. Co-incubation with ADSCs or ADSC-Exos promoted the proliferation and migration of fibroblasts and angiogenesis of HUVECs, which was further enhanced by miR-126-3p overexpression. Inhibition of ADSC-Exos or miR-126-3p or PIK3R2 overexpression suppressed the proliferation and migration of fibroblasts and angiogenesis of HUVECs. ADSC-derived exosomal miR-126-3p increased wound healing rate, collagen deposition and newly formed vessels in injured rats. CONCLUSION ADSC-derived exosomal miR-126-3p promotes wound healing of full-thickness skin defects by targeting PIK3R2.
Collapse
Affiliation(s)
- Jie Ma
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Zhaofeng Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Yinmin Wang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Hua Shen
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| |
Collapse
|
50
|
García-Flores M, Sánchez-López CM, Ramírez-Calvo M, Fernández-Serra A, Marcilla A, López-Guerrero JA. Isolation and characterization of urine microvesicles from prostate cancer patients: different approaches, different visions. BMC Urol 2021; 21:137. [PMID: 34579682 PMCID: PMC8477576 DOI: 10.1186/s12894-021-00902-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Because of their specific and biologically relevant cargo, urine extracellular vesicles (EVs) constitute a valuable source of potential non-invasive biomarkers that could support the clinical decision-making to improve the management of prostate cancer (PCa) patients. Different EV isolation methods differ in terms of complexity and yield, conditioning, as consequence, the analytical result. METHODS The aim of this study was to compare three different isolation methods for urine EVs: ultracentrifugation (UC), size exclusion chromatography (SEC), and a commercial kit (Exolute® Urine Kit). Urine samples were collected from 6 PCa patients and 4 healthy donors. After filtered through 0.22 µm filters, urine was divided in 3 equal volumes to perform EVs isolation with each of the three approaches. Isolated EVs were characterized by spectrophotometric protein quantification, nanoparticle tracking analysis, transmission electron microscopy, AlphaScreen Technology, and whole miRNA Transcriptome. RESULTS Our results showed that UC and SEC provided better results in terms of EVs yield and purity than Exolute®, non-significant differences were observed in terms of EV-size. Interestingly, luminescent AlphaScreen assay demonstrated a significant enrichment of CD9 and CD63 positive microvesicles in SEC and UC methods compared with Exolute®. This heterogeneity was also demonstrated in terms of miRNA content indicating that the best correlation was observed between UC and SEC. CONCLUSIONS Our study highlights the importance of standardizing the urine EV isolation methods to guaranty the analytical reproducibility necessary for their implementation in a clinical setting.
Collapse
Affiliation(s)
- María García-Flores
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain.,IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Christian M Sánchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46000, Burjassot, Valencia, Spain.,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de Valencia, 46100, Valencia, Spain
| | - Marta Ramírez-Calvo
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain
| | - Antonio Fernández-Serra
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46000, Burjassot, Valencia, Spain. .,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de Valencia, 46100, Valencia, Spain.
| | - José Antonio López-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain. .,IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain. .,Department of Pathology, School of Medicine, Catholic University of Valencia "San Vicente Mártir", 46001, Valencia, Spain.
| |
Collapse
|