1
|
Liang L, Deng Y, Ao Z, Liao C, Tian J, Li C, Yu X. Recent progress in biomimetic nanomedicines based on versatile targeting strategy for atherosclerosis therapy. J Drug Target 2024; 32:606-623. [PMID: 38656224 DOI: 10.1080/1061186x.2024.2347353] [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: 01/30/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis (AS) is considered to be one of the major causes of cardiovascular disease. Its pathological microenvironment is characterised by increased production of reactive oxygen species, lipid oxides, and excessive inflammatory factors, which accumulate at the monolayer endothelial cells in the vascular wall to form AS plaques. Therefore, intervention in the pathological microenvironment would be beneficial in delaying AS. Researchers have designed biomimetic nanomedicines with excellent biocompatibility and the ability to avoid being cleared by the immune system through different therapeutic strategies to achieve better therapeutic effects for the characteristics of AS. Biomimetic nanomedicines can further enhance delivery efficiency and improve treatment efficacy due to their good biocompatibility and ability to evade clearance by the immune system. Biomimetic nanomedicines based on therapeutic strategies such as neutralising inflammatory factors, ROS scavengers, lipid clearance and integration of diagnosis and treatment are versatile approaches for effective treatment of AS. The review firstly summarises the targeting therapeutic strategy of biomimetic nanomedicine for AS in recent 5 years. Biomimetic nanomedicines using cell membranes, proteins, and extracellular vesicles as carriers have been developed for AS.
Collapse
Affiliation(s)
- Lijuan Liang
- Department of Pharmacy, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Changli Liao
- Science and Technology Department, Southwest Medical University, Luzhou, Sichuan, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Darbinian N, Hampe M, Martirosyan D, Bajwa A, Darbinyan A, Merabova N, Tatevosian G, Goetzl L, Amini S, Selzer ME. Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs). Int J Mol Sci 2024; 25:5826. [PMID: 38892014 PMCID: PMC11172088 DOI: 10.3390/ijms25115826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Fetal alcohol spectrum disorders (FASDs) are leading causes of neurodevelopmental disability but cannot be diagnosed early in utero. Because several microRNAs (miRNAs) are implicated in other neurological and neurodevelopmental disorders, the effects of EtOH exposure on the expression of these miRNAs and their target genes and pathways were assessed. In women who drank alcohol (EtOH) during pregnancy and non-drinking controls, matched individually for fetal sex and gestational age, the levels of miRNAs in fetal brain-derived exosomes (FB-Es) isolated from the mothers' serum correlated well with the contents of the corresponding fetal brain tissues obtained after voluntary pregnancy termination. In six EtOH-exposed cases and six matched controls, the levels of fetal brain and maternal serum miRNAs were quantified on the array by qRT-PCR. In FB-Es from 10 EtOH-exposed cases and 10 controls, selected miRNAs were quantified by ddPCR. Protein levels were quantified by ELISA. There were significant EtOH-associated reductions in the expression of several miRNAs, including miR-9 and its downstream neuronal targets BDNF, REST, Synapsin, and Sonic hedgehog. In 20 paired cases, reductions in FB-E miR-9 levels correlated strongly with reductions in fetal eye diameter, a prominent feature of FASDs. Thus, FB-E miR-9 levels might serve as a biomarker to predict FASDs in at-risk fetuses.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Monica Hampe
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Diana Martirosyan
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Ahsun Bajwa
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
- Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA;
| | - Shohreh Amini
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Michael E. Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
3
|
Qi L, Xing J, Yuan Y, Lei M. Noncoding RNAs in atherosclerosis: regulation and therapeutic potential. Mol Cell Biochem 2024; 479:1279-1295. [PMID: 37418054 PMCID: PMC11116212 DOI: 10.1007/s11010-023-04794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 06/18/2023] [Indexed: 07/08/2023]
Abstract
Atherosclerosis, a chronic disease of arteries, results in high mortality worldwide as the leading cause of cardiovascular disease. The development of clinically relevant atherosclerosis involves the dysfunction of endothelial cells and vascular smooth muscle cells. A large amount of evidence indicates that noncoding RNAs, such as microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), are involved in various physiological and pathological processes. Recently, noncoding RNAs were identified as key regulators in the development of atherosclerosis, including the dysfunction of endothelial cells, and vascular smooth muscle cells and it is pertinent to understand the potential function of noncoding RNAs in atherosclerosis development. In this review, the latest available research relates to the regulatory role of noncoding RNAs in the progression of atherosclerosis and the therapeutic potential for atherosclerosis is summarized. This review aims to provide a comprehensive overview of the regulatory and interventional roles of ncRNAs in atherosclerosis and to inspire new insights for the prevention and treatment of this disease.
Collapse
MESH Headings
- Humans
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/therapy
- Atherosclerosis/pathology
- Animals
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
Collapse
Affiliation(s)
- Luyao Qi
- Critical Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 200137, Shanghai, China
| | - Jixiang Xing
- Peripheral Vascular Department, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, 300150, Tianjin, China
| | - Yuesong Yuan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, 250014, Jinan, Shandong, China
| | - Ming Lei
- Critical Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 200137, Shanghai, China.
| |
Collapse
|
4
|
Lisi V, Senesi G, Balbi C. Converging protective pathways: Exploring the linkage between physical exercise, extracellular vesicles and oxidative stress. Free Radic Biol Med 2023; 208:718-727. [PMID: 37739138 DOI: 10.1016/j.freeradbiomed.2023.09.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy.
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| |
Collapse
|
5
|
Zhang H, Wan X, Tian J, An Z, Liu L, Zhao X, Zhou Y, Zhang L, Ge C, Song X. The therapeutic efficacy and clinical translation of mesenchymal stem cell-derived exosomes in cardiovascular diseases. Biomed Pharmacother 2023; 167:115551. [PMID: 37783149 DOI: 10.1016/j.biopha.2023.115551] [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: 06/19/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Exosomes, mainly derived from mesenchymal stem cells, provide a good reference for cardiac function repair and clinical application in cardiac and vascular diseases by regulating cardiomyocyte viability, inflammatory levels, angiogenesis, and ventricular remodeling after a heart injury. This review presents the cardioprotective efficacy of mesenchymal stem cell-originated exosomes and explores the underlying molecular mechanisms. Furthermore, we expound on several efficient approaches to transporting exosomes into the heart in clinical application and comment on the advantages and disadvantages of each method.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Xueqi Wan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China; The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, PR China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Yuquan Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Changjiang Ge
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
6
|
Liu J, Chen J, Zhang J, Fan Y, Zhao S, Wang B, Wang P. Mechanism of Resveratrol Improving Ischemia-Reperfusion Injury by Regulating Microglial Function Through microRNA-450b-5p/KEAP1/Nrf2 Pathway. Mol Biotechnol 2023; 65:1498-1507. [PMID: 36656498 DOI: 10.1007/s12033-022-00646-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/23/2022] [Indexed: 01/20/2023]
Abstract
Alterations in the M1/M2 polarization phenotype significantly affect disease progression. Antioxidant and anti-inflammatory protective effects of resveratrol (Res) have been demonstrated. This paper tested the hypothesis that Res could protect against cerebral ischemia-reperfusion injury (CI/RI) by modulating microglial polarization via the miR-450b-5p/KEAP1/Nrf2 pathway. Rats were first treated with Res and adenovirus that interfered with miR-450b-5p or KEAP1, and then established a middle cerebral artery occlusion-reperfusion model using modified nylon sutures. Rats were then evaluated for neurological and behavioral functions, and markers of M2 microglia were detected by immunofluorescence staining. Additionally, the signature patterns of miR-450b-5p, KEAP1, and Nrf2 were determined. The collected data demonstrated that Res exerted neuroprotective effects in CI/RI by promoting microglial M2 polarization. Additionally, Res could regulate the Nrf2 pathway by targeting KEAP1 by up-regulating miR-450b-5p. Up-regulating miR-450b-5p or down-regulating KEAP1 could further promote the protective effect of Res, while down-regulating miR-450b-5p or up-regulating KEAP1 worked oppositely. Our study demonstrates that Res exerts neuroprotective effects on microglial M2 polarization through the miR-450b-5p/KEAP1/Nrf2 pathway during CI/RI.
Collapse
Affiliation(s)
- JiaHui Liu
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - JinYu Chen
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - JinFeng Zhang
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Yu Fan
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - ShiJun Zhao
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - BaoJun Wang
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Po Wang
- Department of Neurology, Inner Mongolia Baotou Central Hospital, No. 61 Ring Roads, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
7
|
Ghafouri-Fard S, Shoorei H, Dong P, Poornajaf Y, Hussen BM, Taheri M, Akbari Dilmaghani N. Emerging functions and clinical applications of exosomal microRNAs in diseases. Noncoding RNA Res 2023; 8:350-362. [PMID: 37250456 PMCID: PMC10209650 DOI: 10.1016/j.ncrna.2023.05.004] [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: 02/13/2023] [Revised: 05/07/2023] [Accepted: 05/07/2023] [Indexed: 05/31/2023] Open
Abstract
Exosomes are an important group of extracellular vesicles that transfer several kinds of biomolecules and facilitate cell-cell communication. The content of exosomes, particularly the amounts of microRNA (miRNAs) inside these vesicles, demonstrates a disease-specific pattern reflecting pathogenic processes and may be employed as a diagnostic and prognostic marker. miRNAs may enter recipient cells through exosomes and generate a RISC complex that can cause degradation of the target mRNAs or block translation of their corresponding proteins. Therefore, exosome-derived miRNAs constitute an important mechanism of gene regulation in recipient cells. The miRNA content of exosomes can be used as an important tool in the detection of diverse disorders, particularly cancers. This research field has an important situation in cancer diagnosis. In addition, exosomal microRNAs offer a great deal of promise in the treatment of human disorders. However, there are still certain challenges to be resolved. The most important challenges are as follow: the detection of exosomal miRNAs should be standardized, exosomal miRNAs-associated studies should be conducted in large number of clinical samples, and experiment settings and detection criteria should be consistent across different labs. The goal of this article is to present an overview of the effects of exosome-derived microRNAs on a variety of diseases, including gastrointestinal, pulmonary, neurological, and cardiovascular diseases, with a particular emphasis on malignancies.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yadollah Poornajaf
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Akbari Dilmaghani
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Li J, Huang Y, Sun H, Yang L. Mechanism of mesenchymal stem cells and exosomes in the treatment of age-related diseases. Front Immunol 2023; 14:1181308. [PMID: 37275920 PMCID: PMC10232739 DOI: 10.3389/fimmu.2023.1181308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) from multiple tissues have the capability of multidirectional differentiation and self-renewal. Many reports indicated that MSCs exert curative effects on a variety of age-related diseases through regeneration and repair of aging cells and organs. However, as research has progressed, it has become clear that it is the MSCs derived exosomes (MSC-Exos) that may have a real role to play, and that they can be modified to achieve better therapeutic results, making them even more advantageous than MSCs for treating disease. This review generalizes the biological characteristics of MSCs and exosomes and their mechanisms in treating age-related diseases, for example, MSCs and their exosomes can treat age-related diseases through mechanisms such as oxidative stress (OS), Wnt/β-catenin signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, and so on. In addition, current in vivo and in vitro trials are described, and ongoing clinical trials are discussed, as well as the prospects and challenges for the future use of exosomes in disease treatment. This review will provide references for using exosomes to treat age-related diseases.
Collapse
Affiliation(s)
- Jia Li
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Yuling Huang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Lina Yang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Yang Y, Peng Y, Li Y, Shi T, Luan Y, Yin C. Role of stem cell derivatives in inflammatory diseases. Front Immunol 2023; 14:1153901. [PMID: 37006266 PMCID: PMC10062329 DOI: 10.3389/fimmu.2023.1153901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells of mesodermal origin with the ability of self-renewal and multidirectional differentiation, which have all the common characteristics of stem cells and the ability to differentiate into adipocytes, osteoblasts, neuron-like cells and other cells. Stem cell derivatives are extracellular vesicles(EVs) released from mesenchymal stem cells that are involved in the process of body’s immune response, antigen presentation, cell differentiation, and anti-inflammatory. EVs are further divided into ectosomes and exosomes are widely used in degenerative diseases, cancer, and inflammatory diseases due to their parental cell characteristics. However, most diseases are closely related to inflammation, and exosomes can mitigate the damage caused by inflammation in terms of suppressing the inflammatory response, anti-apoptosis and promoting tissue repair. Stem cell-derived exosomes have become an emerging modality for cell-free therapy because of their high safety and ease of preservation and transportation through intercellular communication. In this review, we highlight the characteristics and functions of MSCs-derived exosomes and discuss the regulatory mechanisms of MSCs-derived exosomes in inflammatory diseases and their potential applications in clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Yuxi Yang
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yiqiu Peng
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yingying Li
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tingjuan Shi
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yingyi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- *Correspondence: Yingyi Luan, ; Chenghong Yin,
| | - Chenghong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- *Correspondence: Yingyi Luan, ; Chenghong Yin,
| |
Collapse
|
10
|
Extracellular Vesicles and Cellular Ageing. Subcell Biochem 2023; 102:271-311. [PMID: 36600137 DOI: 10.1007/978-3-031-21410-3_11] [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: 01/06/2023]
Abstract
Ageing is a complex process characterized by deteriorated performance at multiple levels, starting from cellular dysfunction to organ degeneration. Stem cell-based therapies aim to administrate stem cells that eventually migrate to the injured site to replenish the damaged tissue and recover tissue functionality. Stem cells can be easily obtained and cultured in vitro, and display several qualities such as self-renewal, differentiation, and immunomodulation that make them suitable candidates for stem cell-based therapies. Current animal studies and clinical trials are being performed to assess the safety and beneficial effects of stem cell engraftments for regenerative medicine in ageing and age-related diseases.Since alterations in cell-cell communication have been associated with the development of pathophysiological processes, new research is focusing on the modulation of the microenvironment. Recent research has highlighted the important role of some microenvironment components that modulate cell-cell communication, thus spreading signals from damaged ageing cells to neighbor healthy cells, thereby promoting systemic ageing. Extracellular vesicles (EVs) are small-rounded vesicles released by almost every cell type. EVs cargo includes several bioactive molecules, such as lipids, proteins, and genetic material. Once internalized by target cells, their specific cargo can induce epigenetic modifications and alter the fate of the recipient cells. Also, EV's content is dependent on the releasing cells, thus, EVs can be used as biomarkers for several diseases. Moreover, EVs have been proposed to be used as cell-free therapies that focus on their administration to slow or even reverse some hallmarks of physiological ageing. It is not surprising that EVs are also under study as next-generation therapies for age-related diseases.
Collapse
|
11
|
Sanz-Ros J, Mas-Bargues C, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Therapeutic Potential of Extracellular Vesicles in Aging and Age-Related Diseases. Int J Mol Sci 2022; 23:ijms232314632. [PMID: 36498960 PMCID: PMC9735639 DOI: 10.3390/ijms232314632] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Aging is associated with an alteration of intercellular communication. These changes in the extracellular environment contribute to the aging phenotype and have been linked to different aging-related diseases. Extracellular vesicles (EVs) are factors that mediate the transmission of signaling molecules between cells. In the aging field, these EVs have been shown to regulate important aging processes, such as oxidative stress or senescence, both in vivo and in vitro. EVs from healthy cells, particularly those coming from stem cells (SCs), have been described as potential effectors of the regenerative potential of SCs. Many studies with different animal models have shown promising results in the field of regenerative medicine. EVs are now viewed as a potential cell-free therapy for tissue damage and several diseases. Here we propose EVs as regulators of the aging process, with an important role in tissue regeneration and a raising therapy for age-related diseases.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Correspondence:
| |
Collapse
|
12
|
Fu RH, Tsai CW, Liu SP, Chiu SC, Chen YC, Chiang YT, Kuo YH, Shyu WC, Lin SZ. Neuroprotective Capability of Narcissoside in 6-OHDA-Exposed Parkinson's Disease Models through Enhancing the MiR200a/Nrf-2/GSH Axis and Mediating MAPK/Akt Associated Signaling Pathway. Antioxidants (Basel) 2022; 11:2089. [PMID: 36358461 PMCID: PMC9686521 DOI: 10.3390/antiox11112089] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 09/29/2023] Open
Abstract
We assessed the antioxidant potential of narcissoside from Sambucus nigra flowers (elderflowers) in Parkinson's disease models in vitro and in vivo. The results showed that narcissoside lessened the 6-hydroxydopamine (6-OHDA)-induced increase in reactive oxygen species (ROS) and apoptosis in SH-SY5Y cells. In the 6-OHDA-exposed Caenorhabditis elegans model, narcissoside reduced degeneration of dopaminergic neurons and ROS generation, and also improved dopamine-related food-sensitive behavior and shortened lifespan. Moreover, NCS increased total glutathione (GSH) by increasing the expression of the catalytic subunit and modifier subunit of γ-glutamylcysteine ligase in cells and nematodes. Treatment with a GSH inhibitor partially abolished the anti-apoptotic ability of narcissoside. Furthermore, narcissoside diminished the 6-OHDA-induced phosphorylation of JNK and p38, while rising activities of ERK and Akt in resisting apoptosis. The antioxidant response element (ARE)-luciferase reporter activity analysis and electromobility gel shift assay showed that narcissoside promotes the transcriptional activity mediated by Nrf2. Finally, we found that narcissoside augmented the expression of miR200a, a translational inhibitor of the Nrf2 repressor protein Keap1. Downregulation of Nrf2 and miR200a by RNAi and anti-miR200a, respectively, reversed the neuroprotective ability of narcissoside. In summary, narcissoside can enhance the miR200a/Nrf2/GSH antioxidant pathway, alleviate 6-OHDA-induced apoptosis, and has the neuroprotective potential.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chia-Wen Tsai
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan
| | - Shih-Ping Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yen-Chuan Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yu-Ting Chiang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yun-Hua Kuo
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shinn-Zong Lin
- Buddhist Tzu Chi Bioinnovation Center, Tzu Chi Foundation, Hualien 97002, Taiwan
- Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien 97002, Taiwan
| |
Collapse
|
13
|
Ye C, Zheng F, Wu N, Zhu GQ, Li XZ. Extracellular vesicles in vascular remodeling. Acta Pharmacol Sin 2022; 43:2191-2201. [PMID: 35022541 PMCID: PMC9433397 DOI: 10.1038/s41401-021-00846-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular remodeling contributes to the development of a variety of vascular diseases including hypertension and atherosclerosis. Phenotypic transformation of vascular cells, oxidative stress, inflammation and vascular calcification are closely associated with vascular remodeling. Extracellular vesicles (EVs) are naturally released from almost all types of cells and can be detected in nearly all body fluids including blood and urine. EVs affect vascular oxidative stress, inflammation, calcification, and lipid plaque formation; and thereby impact vascular remodeling in a variety of cardiovascular diseases. EVs may be used as biomarkers for diagnosis and prognosis, and therapeutic strategies for vascular remodeling and cardiovascular diseases. This review includes a comprehensive analysis of the roles of EVs in the vascular remodeling in vascular diseases, and the prospects of EVs in the diagnosis and treatment of vascular diseases.
Collapse
Affiliation(s)
- Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Nan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China.
| | - Xiu-Zhen Li
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
14
|
de la Cruz-Ojeda P, Schmid T, Boix L, Moreno M, Sapena V, Praena-Fernández JM, Castell FJ, Falcón-Pérez JM, Reig M, Brüne B, Gómez-Bravo MA, Giráldez Á, Bruix J, Ferrer MT, Muntané J. miR-200c-3p, miR-222-5p, and miR-512-3p Constitute a Biomarker Signature of Sorafenib Effectiveness in Advanced Hepatocellular Carcinoma. Cells 2022; 11:cells11172673. [PMID: 36078082 PMCID: PMC9454520 DOI: 10.3390/cells11172673] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Sorafenib constitutes a suitable treatment alternative for patients with advanced hepatocellular carcinoma (HCC) in whom atezolizumab + bevacizumab therapy is contraindicated. The aim of the study was the identification of a miRNA signature in liquid biopsy related to sorafenib response. Methods: miRNAs were profiled in hepatoblastoma HepG2 cells and tested in animal models, extracellular vesicles (EVs), and plasma from HCC patients. Results: Sorafenib altered the expression of 11 miRNAs in HepG2 cells. miR-200c-3p and miR-27a-3p exerted an anti-tumoral activity by decreasing cell migration and invasion, whereas miR-122-5p, miR-148b-3p, miR-194-5p, miR-222-5p, and miR-512-3p exerted pro-tumoral properties by increasing cell proliferation, migration, or invasion, or decreasing apoptosis. Sorafenib induced a change in EVs population with an increased number of larger EVs, and promoted an accumulation of miR-27a-3p, miR-122-5p, miR-148b-3p, miR-193b-3p, miR-194-5p, miR-200c-3p, and miR-375 into exosomes. In HCC patients, circulating miR-200c-3p baseline levels were associated with increased survival, whereas high levels of miR-222-5p and miR-512-3p after 1 month of sorafenib treatment were related to poor prognosis. The RNA sequencing revealed that miR-200c-3p was related to the regulation of cell growth and death, whereas miR-222-5p and miR-512-3p were related to metabolic control. Conclusions: The study showed that Sorafenib regulates a specific miRNA signature in which miR-200c-3p, miR-222-5p, and miR-512-3p bear prognostic value and contribute to treatment response.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60528 Frankfurt, Germany
| | - Loreto Boix
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - Manuela Moreno
- Department of General Surgery, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Víctor Sapena
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | | | - Francisco J. Castell
- Department of Radiology, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Juan Manuel Falcón-Pérez
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Exosomes Lab, CIC bioGUNE, 48160 Derio, Spain
| | - María Reig
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60528 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60528 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60528 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60528 Frankfurt, Germany
| | - Miguel A. Gómez-Bravo
- Department of General Surgery, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Álvaro Giráldez
- Unit for the Clinical Management of Digestive Diseases, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Jordi Bruix
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - María T. Ferrer
- Unit for the Clinical Management of Digestive Diseases, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain
- Correspondence: ; Tel.: +34-955-923-122; Fax: +34-955-923-002
| |
Collapse
|
15
|
Tailored Extracellular Vesicles: Novel Tool for Tissue Regeneration. Stem Cells Int 2022; 2022:7695078. [PMID: 35915850 PMCID: PMC9338735 DOI: 10.1155/2022/7695078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Extracellular vesicles (EVs) play an essential part in multiple pathophysiological processes including tissue injury and regeneration because of their inherent characteristics of small size, low immunogenicity and toxicity, and capability of carrying a variety of bioactive molecules and mediating intercellular communication. Nevertheless, accumulating studies have shown that the application of EVs faces many challenges such as insufficient therapeutic efficacy, a lack of targeting capability, low yield, and rapid clearance from the body. It is known that EVs can be engineered, modified, and designed to encapsulate therapeutic cargos like proteins, peptides, nucleic acids, and drugs to improve their therapeutic efficacy. Targeted peptides, antibodies, aptamers, magnetic nanoparticles, and proteins are introduced to modify various cell-derived EVs for increasing targeting ability. In addition, extracellular vesicle mimetics (EMs) and self-assembly EV-mimicking nanocomplex are applied to improve production and simplify EV purification process. The combination of EVs with biomaterials like hydrogel, and scaffolds dressing endows EVs with long-term therapeutic efficacy and synergistically enhanced regenerative outcome. Thus, we will summarize recent developments of EV modification strategies for more extraordinary regenerative effect in various tissue injury repair. Subsequently, opportunities and challenges of promoting the clinical application of engineered EVs will be discussed.
Collapse
|
16
|
Liu C, Xiao K, Xie L. Advances in the Regulation of Macrophage Polarization by Mesenchymal Stem Cells and Implications for ALI/ARDS Treatment. Front Immunol 2022; 13:928134. [PMID: 35880175 PMCID: PMC9307903 DOI: 10.3389/fimmu.2022.928134] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common condition with high mortality. ALI/ARDS is caused by multiple etiologies, and the main clinical manifestations are progressive dyspnea and intractable hypoxemia. Currently, supportive therapy is the main ALI/ARDS treatment, and there remains a lack of targeted and effective therapeutic strategies. Macrophages are important components of innate immunity. M1 macrophages are pro-inflammatory, while M2 macrophages are anti-inflammatory and promote tissue repair. Mesenchymal stem cells (MSCs) are stem cells with broad application prospects in tissue regeneration due to their multi-directional differentiation potential along with their anti-inflammatory and paracrine properties. MSCs can regulate the balance of M1/M2 macrophage polarization to improve the prognosis of ALI/ARDS. In this paper, we review the mechanisms by which MSCs regulate macrophage polarization and the signaling pathways associated with polarization. This review is expected to provide new targets for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Kun Xiao
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| | - Lixin Xie
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| |
Collapse
|
17
|
Lee S, Ko JH, Kim SN. The Extracellular MicroRNAs on Inflammation: A Literature Review of Rodent Studies. Biomedicines 2022; 10:1601. [PMID: 35884901 PMCID: PMC9312877 DOI: 10.3390/biomedicines10071601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammation is an indispensable biological process stimulated by infection and injuries. Inflammatory mechanisms related to extracellular vesicles (EVs), which are small membrane structures carrying various molecules, were summarized in this review. Emerging evidence from animal studies has highlighted the role of EVs in modulating inflammatory responses, by transporting various molecules involved in host defense. In this review, we have discussed the role of EV miRNAs in inflammation. Rodent studies associated with extracellular miRNAs in inflammatory diseases, published from 2012 to 2022, were explored from PUBMED, EMBASE, and MEDLINE. A total of 95 studies were reviewed. In summary, EV-associated miRNAs play a key role in various diseases, including organ injury, immune dysfunction, neurological disease, metabolic syndrome, vesicular disease, arthritis, cancer, and other inflammatory diseases. Diverse EV-associated miRNAs regulate inflammasome activation and pro- and anti-inflammatory cytokine levels by targeting genes.
Collapse
Affiliation(s)
- Seri Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
- Graduate School, Dongguk University, Seoul 04620, Korea
| | - Jade Heejae Ko
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| |
Collapse
|
18
|
Ren H, Guo Z, Liu Y, Song C. Stem Cell-derived Exosomal MicroRNA as Therapy for Vascular Age-related Diseases. Aging Dis 2022; 13:852-867. [PMID: 35656114 PMCID: PMC9116915 DOI: 10.14336/ad.2021.1110] [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: 08/25/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular age-related diseases describe a group of age-related chronic diseases that result in a considerable healthcare burden to society. Vascular aging includes structural changes and dysfunctions of endothelial cells (ECs) and smooth muscle cells (SMCs) in blood vessels. Compared with conventional treatment for vascular age-related diseases, stem cell (SC) therapy elicits better anti-aging effects viathe inhibition/delay ECs and SMCs from entering senescence. Exosomal noncoding RNA (ncRNAs) in vascular aging and stem cell-derived exosomal microRNAs (SCEV-miRNAs), especially in mesenchymal stem cells, have an important role in the development of age-related diseases. This review summarizes SCEV-miRNAs of diverse origins that may play a vital role in treating subclinical and clinical stages of vascular age-related disorders. We further explored possible age-related pathways and molecular targets of SCEV-miRNA, which are associated with dysfunctions of ECs and SMCs in the senescent stage. Moreover, the perspectives and difficulties of SCEV-miRNA clinical translation are discussed. This review aims to provide greater understanding of the biology of vascular aging and to identify critical therapeutic targets for SCEV-miRNAs. Though still in its infancy, the potential value of SCEV-miRNAs for vascular age-related diseases is clear.
Collapse
Affiliation(s)
- Hang Ren
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Ziyuan Guo
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Chunli Song
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Yan F, Cui W, Chen Z. Mesenchymal Stem Cell-Derived Exosome-Loaded microRNA-129-5p Inhibits TRAF3 Expression to Alleviate Apoptosis and Oxidative Stress in Heart Failure. Cardiovasc Toxicol 2022; 22:631-645. [PMID: 35546649 DOI: 10.1007/s12012-022-09743-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/16/2022] [Indexed: 11/03/2022]
Abstract
Heart failure (HF) represents a main global healthy and economic burden with unacceptably high morbidity and mortality rates. In the current study, we evaluated the potential effect of mesenchymal stem cell (MSC)-derived exosomes (MSC-Exos) on oxygen-glucose deprivation (OGD)-induced damages to HL-1 cells and HF mice and searched for the possible mechanism. MSC-Exos ameliorated oxidative stress and reduced apoptosis in OGD-treated HL-1 cells. By microarray analysis, we found that MSC-Exos treatment significantly increased the microRNA (miR)-129-5p expression in HL-1 cells. miR-129-5p inhibitor attenuated the protective effect of MSC-Exos on OGD-treated HL-1 cells. miR-129-5p targeted tumor necrosis factor receptor-associated factor 3 (TRAF3), and TRAF3 loss reversed the effect of miR-129-5p inhibitor by blunting the NF-κB signaling. MSC-Exos injection alleviated ventricular dysfunction and suppressed oxidative stress, apoptosis, inflammation, and fibrosis in cardiomyocytes in mice with HF by inhibiting NF-κB signaling pathway through miR-129-5p/TRAF3. Our findings suggest that exosomal miR-129-5p from MSCs protects the heart from failure by targeting TRAF3 and the following NF-κB signaling. This regulatory axis may be a possible therapeutic target for HF.
Collapse
Affiliation(s)
- Fang Yan
- Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China.,Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Wei Cui
- Department of Cardiology, the Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Ziying Chen
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
20
|
Zhu B, Liu W, Xu Q, Liu HL. Clinical Values and Underlying Mechanism Analysis of Serum miR-455-5p in Carotid Artery Stenosis. J Inflamm Res 2022; 15:3207-3217. [PMID: 35668916 PMCID: PMC9165656 DOI: 10.2147/jir.s362774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/04/2022] [Indexed: 12/03/2022] Open
Abstract
Purpose Carotid artery stenosis (CAS) is a leading cause of cerebral infarction, its early diagnosis and intervention are necessary. In light of the important role of microRNAs (miRNAs) in cerebrovascular disease, this study aimed to investigate the expression pattern and clinical significance of serum miR-455-5p in the onset and development of CAS, as well as its underlying mechanism. Patients and Methods Seventy patients with asymptomatic CAS were recruited, and the development of cerebral ischemia events (CIEs) was recorded during the five-years follow-up. qRT-PCR was performed for the serum miR-455-5p detection. ROC curve was applied for the diagnostic ability evaluation. By constructing multivariable logistic or cox regression model, odds ratio (OR) or hazard ratio (HR) were calculated to assess the impact of each risk factor on independent variables. Human aortic endothelial cells (HAECs) were treated with ox-LDL to induce endothelial cell damage. The role of miR-455-5p in the cell viability, apoptosis, oxidative stress and inflammatory response was detected. Results Serum miR-455-5p showed low expression in cases with CAS, and had an independent influence on the degree of CAS. The diagnostic ability of serum miR-455-5p to diagnose CAS was determined via ROC curve, with the AUC of 0.927. During follow-up, patients with low miR-455-5p expression showed high incidence of CIEs. In multivariable cox regression model, degree of CAS and miR-455-5p were significant risk factors for the development of CIEs in the CAS patients. In vitro, miR-455-5p was at a low expression in HAECs cell models and can prevent cells from ox-LDL induced cell apoptosis, oxidative stress and inflammatory response. SOCS3 was a target gene of miR-455-5p and upregulated in ox-LDL treated cells. Conclusion Down-regulated expression of serum miR-455-5p is hopeful to be used as a biomarker for the early diagnosis of CAS. MiR-455-5p is an independent risk factor for the degree of CAS, and has a certain predictive value for the development of CIEs. That might be associated with the protective role of miR-455-5p against ox-LDL-induced endothelial cell injury via targeting SOCS3.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Neurosurgery, Renhe Hospital, Baoshan District, Shanghai, People’s Republic of China
| | - Wei Liu
- Department of Neurosurgery, Renhe Hospital, Baoshan District, Shanghai, People’s Republic of China
| | - Qiang Xu
- Department of Neurosurgery, Renhe Hospital, Baoshan District, Shanghai, People’s Republic of China
| | - Hong-liang Liu
- Department of Neurosurgery, Renhe Hospital, Baoshan District, Shanghai, People’s Republic of China
- Correspondence: Hong-liang Liu, Department of Neurosurgery, Renhe Hospital, Baoshan District, 1999 West Changjiang Road, Shanghai, 200431, People Republic of China, Tel +86-21-56731199-6083, Email
| |
Collapse
|
21
|
Cui Y, Zhou Y, Gan N, Xiang Q, Xia M, Liao W, Zheng XL, Peng J, Tang Z. The Role of Extracellular Non-coding RNAs in Atherosclerosis. J Cardiovasc Transl Res 2022; 15:477-491. [PMID: 35233720 DOI: 10.1007/s12265-022-10218-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
Atherosclerosis (AS) is a complex chronic inflammatory disease that leads to myocardial infarction, stroke, and disabling peripheral artery disease. Non-coding RNAs (ncRNAs) directly participate in various physiological processes and exhibit a wide range of biological functions. The present review discusses how different ncRNAs participate in the process of AS in various carrier forms. We focused on the role and potential mechanisms of extracellular ncRNAs in AS and examined their potential implications for clinical treatment.
Collapse
Affiliation(s)
- Yuting Cui
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
| | - Yating Zhou
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
| | - Ni Gan
- Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qiong Xiang
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
| | - Mengdie Xia
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
| | - Wei Liao
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
| | - Xi-Long Zheng
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Juan Peng
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhihan Tang
- Institute of Cardiovascular Disease Key Laboratory for Arteriosclerology of Hunan Province School of Basic Medical Sciences Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
22
|
Heo J, Kang H. Exosome-Based Treatment for Atherosclerosis. Int J Mol Sci 2022; 23:ijms23021002. [PMID: 35055187 PMCID: PMC8778342 DOI: 10.3390/ijms23021002] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is an inflammatory disease in which lipids accumulate on the walls of blood vessels, thickening and clogging these vessels. It is well known that cell-to-cell communication is involved in the pathogenesis of atherosclerosis. Exosomes are extracellular vesicles that deliver various substances (e.g., RNA, DNA, and proteins) from the donor cell to the recipient cell and that play an important role in intercellular communication. Atherosclerosis can be either induced or inhibited through cell-to-cell communication using exosomes. An understanding of the function of exosomes as therapeutic tools and in the pathogenesis of atherosclerosis is necessary to develop new atherosclerosis therapies. In this review, we summarize the studies on the regulation of atherosclerosis through exosomes derived from multiple cells as well as research on exosome-based atherosclerosis treatment.
Collapse
Affiliation(s)
- Jeongyeon Heo
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea
| |
Collapse
|
23
|
Sun L, He X, Zhang T, Han Y, Tao G. Knockdown of mesenchymal stem cell‑derived exosomal LOC100129516 suppresses the symptoms of atherosclerosis via upregulation of the PPARγ/LXRα/ABCA1 signaling pathway. Int J Mol Med 2021; 48:208. [PMID: 34608501 PMCID: PMC8510681 DOI: 10.3892/ijmm.2021.5041] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapy has potential applications in treating atherosclerosis and coronary heart disease (CAD). Previous studies have demonstrated that MSCs are the most preferable sources of therapeutic exosomes, which carry long non‑coding RNAs and participate in the progression of atherosclerosis. The results of our previous bioinformatics study demonstrated that the levels of LOC100129516 were significantly upregulated in peripheral blood mononuclear cells obtained from patients with CAD. However, the biological role of LOC100129516 in the development of atherosclerosis remains to be elucidated. In the present study, THP‑1 cells were treated with oxidized low‑density lipoproteins to induce foam cell formation in vitro. Reverse transcription‑quantitative PCR (RT‑qPCR) was performed to detect the levels of LOC100129516 in THP‑1 macrophage‑derived foam cells. In addition, an in vivo model of atherosclerosis was established using Apolipoprotein E (ApoE) knockout (ApoE‑/‑) mice. The results of the RT‑qPCR assays demonstrated that the levels of LOC100129516 were upregulated in THP‑1 macrophage‑derived foam cells. MSC‑derived exosomes were able to deliver small interfering (si)‑LOC100129516 to THP‑1 cells to reduce the levels of LOC100129516. Moreover, transfection of si‑LOC100129516 via exosomal delivery significantly decreased the levels of total cholesterol (TC), free cholesterol and cholesterol ester in THP‑1 macrophage‑derived foam cells. Exosomal‑mediated delivery of si‑LOC100129516 decreased TC levels and low‑density lipoprotein levels in the ApoE‑/‑ atherosclerosis mouse model. Mechanistically, exosomal‑mediated delivery of si‑LOC100129516 promoted cholesterol efflux by activating the peroxisome proliferator‑activated receptor γ (PPARγ)/liver X receptor α (LXRα)/phospholipid‑transporting ATPase ABCA1 (ABCA1) signaling pathway in vitro and in vivo. Collectively, these results suggested that exosomal‑mediated delivery of si‑LOC100129516, in which the exosomes were derived from MSCs, promoted cholesterol efflux and suppressed intracellular lipid accumulation, ultimately alleviating the progression of atherosclerosis via stimulation of the PPARγ/LXRα/ABCA1 signaling pathway.
Collapse
Affiliation(s)
- Limin Sun
- Department of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of General Practice, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xin He
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Tao Zhang
- Department of General Practice, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Guizhou Tao
- Department of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
24
|
Xu H, Ni YQ, Liu YS. Mechanisms of Action of MiRNAs and LncRNAs in Extracellular Vesicle in Atherosclerosis. Front Cardiovasc Med 2021; 8:733985. [PMID: 34692785 PMCID: PMC8531438 DOI: 10.3389/fcvm.2021.733985] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis, a complex chronic inflammatory disease, involves multiple alterations of diverse cells, including endothelial cells (ECs), vascular smooth muscle cells (VSMCs), monocytes, macrophages, dendritic cells (DCs), platelets, and even mesenchymal stem cells (MSCs). Globally, it is a common cause of morbidity as well as mortality. It leads to myocardial infarctions, stroke and disabling peripheral artery disease. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that secreted by multiple cell types and play a central role in cell-to-cell communication by delivering various bioactive cargos, especially microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Emerging evidence demonstrated that miRNAs and lncRNAs in EVs are tightly associated with the initiation and development of atherosclerosis. In this review, we will outline and compile the cumulative roles of miRNAs and lncRNAs encapsulated in EVs derived from diverse cells in the progression of atherosclerosis. We also discuss intercellular communications via EVs. In addition, we focused on clinical applications and evaluation of miRNAs and lncRNAs in EVs as potential diagnostic biomarkers and therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| | - Yu-Qing Ni
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| |
Collapse
|
25
|
Chang YJ, Wang KC. Therapeutic perspectives of extracellular vesicles and extracellular microRNAs in atherosclerosis. CURRENT TOPICS IN MEMBRANES 2021; 87:255-277. [PMID: 34696887 DOI: 10.1016/bs.ctm.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Extracellular signaling molecules, such as growth factors, cytokines, and hormones, regulate cell behaviors and fate through endocrine, paracrine, and autocrine actions and play essential roles in maintaining tissue homeostasis. MicroRNAs, an important class of posttranscriptional modulators, could stably present in extracellular space and body fluids and participate in intercellular communication in health and diseases. Indeed, recent studies demonstrated that microRNAs could be secreted through vesicular and non-vesicular routes, transported in body fluids, and then transmitted to recipient cells to regulate target gene expression and signaling events. Over the past decade, a great deal of effort has been made to investigate the functional roles of extracellular vesicles and extracellular microRNAs in pathological conditions. Emerging evidence suggests that altered levels of extracellular vesicles and extracellular microRNAs in body fluids, as part of the cellular responses to atherogenic factors, are associated with the development of atherosclerosis. This review article provides a brief overview of extracellular vesicles and perspectives of their applications as therapeutic tools for cardiovascular pathologies. In addition, we highlight the role of extracellular microRNAs in atherogenesis and offer a summary of circulating microRNAs in liquid biopsies associated with atherosclerosis.
Collapse
Affiliation(s)
- Ya-Ju Chang
- Department of Family Medicine and Public Health, School of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
26
|
Zingg JM, Vlad A, Ricciarelli R. Oxidized LDLs as Signaling Molecules. Antioxidants (Basel) 2021; 10:antiox10081184. [PMID: 34439432 PMCID: PMC8389018 DOI: 10.3390/antiox10081184] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Levels of oxidized low-density lipoproteins (oxLDLs) are usually low in vivo but can increase whenever the balance between formation and scavenging of free radicals is impaired. Under normal conditions, uptake and degradation represent the physiological cellular response to oxLDL exposure. The uptake of oxLDLs is mediated by cell surface scavenger receptors that may also act as signaling molecules. Under conditions of atherosclerosis, monocytes/macrophages and vascular smooth muscle cells highly exposed to oxLDLs tend to convert to foam cells due to the intracellular accumulation of lipids. Moreover, the atherogenic process is accelerated by the increased expression of the scavenger receptors CD36, SR-BI, LOX-1, and SRA in response to high levels of oxLDL and oxidized lipids. In some respects, the effects of oxLDLs, involving cell proliferation, inflammation, apoptosis, adhesion, migration, senescence, and gene expression, can be seen as an adaptive response to the rise of free radicals in the vascular system. Unlike highly reactive radicals, circulating oxLDLs may signal to cells at more distant sites and possibly trigger a systemic antioxidant defense, thus elevating the role of oxLDLs to that of signaling molecules with physiological relevance.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| | - Adelina Vlad
- Physiology Department, “Carol Davila” UMPh, 020021 Bucharest, Romania;
| | - Roberta Ricciarelli
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| |
Collapse
|