1
|
Schröder S, Epple R, Fischer A, Schettler VJJ. Effective exosomes reduction in hypercholesterinemic patients suffering from cardiovascular diseases by lipoprotein apheresis: Exosomes apheresis. Ther Apher Dial 2024; 28:863-870. [PMID: 38837319 DOI: 10.1111/1744-9987.14172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/21/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION Extracellular vesicles (EVs) have been identified as playing a role in atherosclerosis. METHODS A group of 37 hypercholesterolemic patients with atherosclerotic cardiovascular diseases (ASCVD) and 9 patients requiring hemodialysis (HD) were selected for the study. RESULTS EVs were comparably reduced by various LA methods (Thermo: 87.66% ± 3.64, DALI: 87.96% ± 4.81, H.E.L.P.: 83.38% ± 11.98; represented as SEM). However, LDL-C (66%; 55%; 75%) and Lp(a) (72%; 67%; 79%) were less effectively reduced by DALI. There was no significant difference in the reduction of EVs when comparing different techniques, such as hemoperfusion (DALI; n = 13), a precipitation (H.E.L.P.; n = 5), and a double filtration procedure (Thermofiltration; n = 19). Additionally, no effect of hemodialysis on EVs reduction was found. CONCLUSIONS The study suggests that EVs can be effectively removed by various LA procedures, and this effect appears to be independent of the specific LA procedure used, as compared to hemodialysis.
Collapse
Affiliation(s)
- Sophie Schröder
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Robert Epple
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | | |
Collapse
|
2
|
Wang FD, Ding Y, Zhou JH, Zhou E, Zhang TT, Fan YQ, He Q, Zhang ZQ, Mao CY, Zhang JF, Zhou J. Gamma-aminobutyric acid enhances miR-21-5p loading into adipose-derived stem cell extracellular vesicles to alleviate myocardial ischemia-reperfusion injury via TXNIP regulation. World J Stem Cells 2024; 16:873-895. [PMID: 39493825 PMCID: PMC11525649 DOI: 10.4252/wjsc.v16.i10.873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/21/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) poses a prevalent challenge in current reperfusion therapies, with an absence of efficacious interventions to address the underlying causes. AIM To investigate whether the extracellular vesicles (EVs) secreted by adipose mesenchymal stem cells (ADSCs) derived from subcutaneous inguinal adipose tissue (IAT) under γ-aminobutyric acid (GABA) induction (GABA-EVsIAT) demonstrate a more pronounced inhibitory effect on mitochondrial oxidative stress and elucidate the underlying mechanisms. METHODS We investigated the potential protective effects of EVs derived from mouse ADSCs pretreated with GABA. We assessed cardiomyocyte injury using terminal deoxynucleotidyl transferase dUTP nick end-labeling and Annexin V/propidium iodide assays. The integrity of cardiomyocyte mitochondria morphology was assessed using electron microscopy across various intervention backgrounds. To explore the functional RNA diversity between EVsIAT and GABA-EVsIAT, we employed microRNA (miR) sequencing. Through a dual-luciferase reporter assay, we confirmed the molecular mechanism by which EVs mediate thioredoxin-interacting protein (TXNIP). Western blotting and immunofluorescence were conducted to determine how TXNIP is involved in mediation of oxidative stress and mitochondrial dysfunction. RESULTS Our study demonstrates that, under the influence of GABA, ADSCs exhibit an increased capacity to encapsulate a higher abundance of miR-21-5p within EVs. Consequently, this leads to a more pronounced inhibitory effect on mitochondrial oxidative stress compared to EVs from ADSCs without GABA intervention, ultimately resulting in myocardial protection. On a molecular mechanism level, EVs regulate the expression of TXNIP and mitigating excessive oxidative stress in mitochondria during MIRI process to rescue cardiomyocytes. CONCLUSION Administration of GABA leads to the specific loading of miR-21-5p into EVs by ADSCs, thereby regulating the expression of TXNIP. The EVs derived from ADSCs treated with GABA effectively ameliorates mitochondrial oxidative stress and mitigates cardiomyocytes damage in the pathological process of MIRI.
Collapse
Affiliation(s)
- Feng-Dan Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yi Ding
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Hong Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - En Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tian-Tian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu-Qi Fan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qing He
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zong-Qi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Cheng-Yu Mao
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jun-Feng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jing Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
3
|
Gu X, He L, Zhang J, Xu H, Shen H, Huang R, Li Z. Recent Advances in Wash-Free Detection Methods of Extracellular Vesicles: A Mini Review. ACS Sens 2024. [PMID: 39446112 DOI: 10.1021/acssensors.4c00315] [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: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are emerging biomarkers in liquid biopsy that have gained increasing attention in disease diagnosis and prognosis monitoring. Most reported detection methods require the isolation of EVs from complex body liquids, often involving multiple washing steps to remove excess reagents and eliminate background interference. Nonetheless, these methods not only cause the loss of EVs but also result in poor repeatability and prolonged detection duration. The focus on wash-free detection methods is increasing due to the specific ability to avoid the removal of surplus reagents and, in some cases, even the isolation and purification of EVs. Viewing from different methodological perspectives, this review summarizes the recent advances in wash-free detection of EVs, containing aggregation induction, proximity sensing, allosteric probes, phase separation, Roman spectroscopy, field-effect transistor and microcantilever. The pros and cons of each detection strategy are impartially evaluated and this review concludes the prospects for future developments in this field.
Collapse
Affiliation(s)
- Xinrui Gu
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Lei He
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Jinsong Zhang
- Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Hongpan Xu
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Han Shen
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, South Puzhu Road 30, Nanjing, Jiangsu Province 211816, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| |
Collapse
|
4
|
Stone C, Sabe SA, Harris DD, Broadwin M, Kant RJ, Kanuparthy M, Abid MR, Sellke FW. Metformin Preconditioning Augments Cardiac Perfusion and Performance in a Large Animal Model of Chronic Coronary Artery Disease. Ann Surg 2024; 280:547-556. [PMID: 39041226 DOI: 10.1097/sla.0000000000006437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
OBJECTIVE To test the efficacy of metformin (MET) during the induction of coronary ischemia on myocardial performance in a large animal model of coronary artery disease (CAD) and metabolic syndrome (MS), with or without concomitant extracellular vesicular (EV) therapy. BACKGROUND Although surgical and endovascular revascularization are durably efficacious for many patients with CAD, up to one-third are poor candidates for standard therapies. For these patients, many of whom have comorbid MS, adjunctive strategies are needed. EV therapy has shown promise in this context, but its efficacy is attenuated by MS. We investigated whether MET pretreatment could ameliorate therapeutic decrements associated with MS. METHODS Yorkshire swine (n = 29) were provided a high-fat diet to induce MS, whereupon an ameroid constrictor was placed to induce CAD. Animals were initiated on 1000 mg oral MET or placebo; all then underwent repeat thoracotomy for intramyocardial injection of EVs or saline. Swine were maintained for 5 weeks before the acquisition of functional and perfusion data immediately before terminal myocardial harvest. Immunoblotting and immunofluorescence were performed on the most ischemic tissue from all groups. RESULTS Regardless of EV administration, animals that received MET exhibited significantly improved ejection fraction, cardiac index, and contractility at rest and during rapid myocardial pacing, improved perfusion to the most ischemic myocardial region at rest and during pacing, and markedly reduced apoptosis. CONCLUSIONS MET administration reduced apoptotic cell death, improved perfusion, and augmented both intrinsic and load-dependent myocardial performance in a highly translatable large animal model of chronic myocardial ischemia and MS.
Collapse
Affiliation(s)
- Christopher Stone
- Department of Surgery, Division of Cardiothoracic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Ding Z, Qi F, Liu L, Wang Z, Zhang N, Lyu X, Sun W, Du J, Song H, Hou H, Guo Y, Wang X, Liu ML, Wei W. Circulating extracellular vesicles as novel biomarkers for pulmonary arterial hypertension in patients with systemic lupus erythematosus. Front Immunol 2024; 15:1374100. [PMID: 39364410 PMCID: PMC11446868 DOI: 10.3389/fimmu.2024.1374100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/28/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction Pulmonary arterial hypertension (PAH) is a serious complication of systemic lupus erythematosus (SLE) with increased mortality. A prothrombotic state may contribute to pathogenesis of SLE-PAH. Extracellular vesicles (EVs) are known to be associated with thrombosis. Here, we investigated circulating EVs and their associations with SLE-PAH. Methods Eighteen SLE-PAH patients, 36 SLE-non-PAH patients, and 36 healthy controls (HCs) were enrolled. Flow cytometry was used to analyze circulating EVs from leukocytes (LEVs), red blood cells (REVs), platelets (PEVs), endothelial cells (EEVs), and Annexin V+ EVs with membrane phosphatidylserine (PS) exposure. Results Plasma levels of all EV subgroups were elevated in SLE patients with or without PAH compared to HCs. Furthermore, plasma Annexin V+ EVs, LEVs, PEVs, REVs, EEVs, and Annexin V+ REVs were significantly elevated in SLE-PAH patients compared to SLE-non-PAH patients. Additionally, PAH patients with moderate/high SLE showed a significant increase in LEVs, PEVs, REVs, Annexin V+ EVs, and Annexin V+ REVs compared to SLE-non-PAH patients. However, PAH patients with inactive/mild SLE only exhibited elevations in Annexin V+ EVs, REVs, and Annexin V+ REVs. In the SLE-PAH patients, EEVs were positively correlated with pulmonary arterial systolic pressure, while PEVs and EEVs were positively correlated with right ventricular diameter. Moreover, the receiver operating characteristic curve indicated that Annexin V+ EVs, LEVs, PEVs, REVs, EEVs and Annexin V+ REVs could predict the presence of PAH in SLE patients. Importantly, multivariate logistic regression analysis showed that circulating levels of LEVs or REVs, anti-nRNP antibody, and serositis were independent risk factors for PAH in SLE patients. Discussion Findings reveal that specific subgroups of circulating EVs contribute to the hypercoagulation state and the severity of SLE-PAH. Higher plasma levels of LEVs or REVs may serve as biomarkers for SLE-PAH.
Collapse
Affiliation(s)
- Zhe Ding
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Fumin Qi
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Li Liu
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhouming Wang
- Department of Cardiovascular, Tianjin Medical University General Hospital, Tianjin, China
| | - Na Zhang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Xing Lyu
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Wenwen Sun
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Jun Du
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Haoming Song
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hou Hou
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Ying Guo
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Xiaomei Wang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Ming-Lin Liu
- Corporal Michael J. Crescenz Veterans Affairs Medical Center (VAMC), Philadelphia, PA, United States
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| |
Collapse
|
6
|
Moreno A, Alarcón-Zapata P, Guzmán-Gútierrez E, Radojkovic C, Contreras H, Nova-Lampeti E, A Zúñiga F, Rodriguez-Alvárez L, Escudero C, Lagos P, Aguayo C. Changes in the Release of Endothelial Extracellular Vesicles CD144+, CCR6+, and CXCR3+ in Individuals with Acute Myocardial Infarction. Biomedicines 2024; 12:2119. [PMID: 39335632 PMCID: PMC11430588 DOI: 10.3390/biomedicines12092119] [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: 06/18/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Acute myocardial infarction (AMI) results from vulnerable plaque rupture, causing ischemic cardiomyocyte necrosis and intense inflammation. Paradoxically, this inflammation releases factors that aid heart repair. Recent findings suggest a role for extracellular vesicles (EVs) in intercellular communication during post-AMI cardiac repair. However, EVs' tissue origin and chemokine profile in the blood of patients with AMI remains unclear. This study characterized the tissue origin and chemokine receptor profile of EVs in the coronary and peripheral blood of patients with AMI. The results reveal that vesicles isolated from coronary and peripheral blood plasma are enriched in tetraspanin (CD9) and express CD81+, CD90+, and CD144+. The vesicle size ranged between 145 and 162 nm, with the control group exhibiting smaller vesicles (D10) than the AMI group. Furthermore, all vesicles expressed CCR6 and CXCR3, whereas a small percentage expressed CCR4. In addition, a decrease in CXCR3 and CCR6 expression was observed in coronary and peripheral AMI blood vesicles compared with the control; however, no difference was found between AMI coronary and AMI peripheral blood vesicles. In conclusion, our study demonstrates, for the first time, changes in the number of extracellular vesicles expressing CD144+, CXCR3, and CCR6 in the peripheral circulation of patients with AMI. Extracellular vesicles present in the circulation of patients with AMI hold excellent promise as a potential diagnostic tool.
Collapse
Affiliation(s)
- Alexa Moreno
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Clinical Laboratory Program, Faculty of Health Sciences, State University of Southern Manabí, Jipijapa 130402, Ecuador
| | - Pedro Alarcón-Zapata
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Enrique Guzmán-Gútierrez
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudia Radojkovic
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Héctor Contreras
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Estefanía Nova-Lampeti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Felipe A Zúñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Llerenty Rodriguez-Alvárez
- Department of Animal Science, Faculty of Veterinary Sciences, University of Concepcion, Chillán 3780000, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bio-Bio, Chillán 3780000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| | - Paola Lagos
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| |
Collapse
|
7
|
Evanoff NG, Dengel DR, Stockelman KA, Fandl H, DeSouza NM, Greiner JJ, Dufresne SR, Kotlyar M, Garcia VP. Circulating extracellular microvesicles associated with electronic cigarette use increase endothelial cell inflammation and reduce nitric oxide production. Exp Physiol 2024; 109:1593-1603. [PMID: 39092897 PMCID: PMC11363099 DOI: 10.1113/ep091715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
The purpose of this study was to determine the effect of circulating microvesicles isolated from chronic electronic (e-)cigarette users on cultured human umbilical vein endothelial cell (HUVEC) expression of nuclear factor-κB (NF-κB), cellular cytokine release, phosphorylation of endothelial nitric oxide synthase (eNOS) and NO production. The HUVECs were treated with microvesicles isolated via flow cytometry from nine non-tobacco users (five male and four female; 22 ± 2 years of age) and 10 e-cigarette users (six male and four female; 22 ± 2 years of age). Microvesicles from e-cigarette users induced significantly greater release of interleukin-6 (183.4 ± 23.6 vs. 150.6 ± 15.4 pg/mL; P = 0.002) and interleukin-8 (160.0 ± 31.6 vs. 129.4 ± 11.2 pg/mL; P = 0.01), in addition to expression of p-NF-κB p65 (Ser536) (18.8 ± 3.4 vs. 15.6 ± 1.5 a.u.; P = 0.02) from HUVECs compared with microvesicles from non-tobacco users. Nuclear factor-κB p65 was not significantly different between microvesicles from the non-tobacco users and from the e-cigarette users (87.6 ± 8.7 vs. 90.4 ± 24.6 a.u.; P = 0.701). Neither total eNOS (71.4 ± 21.8 vs. 80.4 ± 24.5 a.u.; P = 0.413) nor p-eNOS (Thr495) (229.2 ± 26.5 vs. 222.1 ± 22.7 a.u.; P = 0.542) was significantly different between microvesicle-treated HUVECs from non-tobacco users and e-cigarette users. However, p-eNOS (Ser1177) (28.9 ± 6.2 vs. 45.8 ± 9.0 a.u.; P < 0.001) expression was significantly lower from e-cigarette users compared with non-tobacco users. Nitric oxide production was significantly lower (8.2 ± 0.6 vs. 9.7 ± 0.9 μmol/L; P = 0.001) in HUVECs treated with microvesicles from e-cigarette users compared with microvesicles from non-tobacco users. This study demonstrated increased NF-κB activation and inflammatory cytokine production, in addition to diminished eNOS activity and NO production resulting from e-cigarette use. HIGHLIGHTS: What is the central question of this study? Circulating microvesicles contribute to cardiovascular health and disease via their effects on the vascular endothelium. The impact of electronic (e-)cigarette use on circulating microvesicle phenotype is not well understood. What is the main finding and its importance? Circulating microvesicles from e-cigarette users increase endothelial cell inflammation and impair endothelial nitric oxide production. Endothelial inflammation and diminished nitric oxide bioavailability are central factors underlying endothelial dysfunction and, in turn, cardiovascular disease risk. Deleterious changes in the functional phenotype of circulating microvesicles might contribute to the reported adverse effects of e-cigarette use on cardiovascular health.
Collapse
Affiliation(s)
- Nicholas G. Evanoff
- School of KinesiologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Center for Pediatric Obesity MedicineUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Donald R. Dengel
- School of KinesiologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Center for Pediatric Obesity MedicineUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Kelly A. Stockelman
- Department of Integrative PhysiologyUniversity of ColoradoBoulderColoradoUSA
| | - Hannah Fandl
- Department of Integrative PhysiologyUniversity of ColoradoBoulderColoradoUSA
| | - Noah M. DeSouza
- Department of Integrative PhysiologyUniversity of ColoradoBoulderColoradoUSA
| | - Jared J. Greiner
- Department of Integrative PhysiologyUniversity of ColoradoBoulderColoradoUSA
| | - Sheena R. Dufresne
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michael Kotlyar
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Vinicius P. Garcia
- Department of Integrative PhysiologyUniversity of ColoradoBoulderColoradoUSA
| |
Collapse
|
8
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [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: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
He Y, Zhang Q, Pan L, Yang H, Liu T, Bei J, Peter K, Hu H. Platelets in Vascular Calcification: A Comprehensive Review of Platelet-Derived Extracellular Vesicles, Protein Interactions, Platelet Function Indices, and their Impact on Cellular Crosstalk. Semin Thromb Hemost 2024. [PMID: 39191407 DOI: 10.1055/s-0044-1789023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Vascular calcification (VC) commonly accompanies the development of atherosclerosis, defined by the accumulation of calcium in the arterial wall, potentially leading to stroke and myocardial infarction. Severe and unevenly distributed calcification poses challenges for interventional procedures, elevating the risks of vascular dissection, acute vascular occlusion, restenosis, and other major adverse cardiovascular events. Platelets promote the development of atherosclerosis by secreting various inflammatory mediators, regulating cell migration, aggregation, adhesion, and initiating and expanding inflammatory responses. There is emerging evidence that platelets play a direct role in VC; however, this novel concept has not yet been critically assessed. This review describes the intricate mechanisms by which platelets promote VC, focusing on three key aspects and the potential opportunities for their therapeutic targeting: extracellular vesicles, platelet-regulatory proteins, and indices related to platelet function.
Collapse
Affiliation(s)
- Yi He
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiongyue Zhang
- Department of Nephrology, Daping Hospital, Army Medical Center, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lina Pan
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Yang
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao Liu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junjie Bei
- Department of Cardiology, Guangxi Zhuang Autonomous Region Corps Hospital of People's Armed Police, Nanning, China
| | - Karlheinz Peter
- Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
10
|
Vachon L, Jean G, Milasan A, Babran S, Lacroix E, Guadarrama Bello D, Villeneuve L, Rak J, Nanci A, Mihalache-Avram T, Tardif JC, Finnerty V, Ruiz M, Boilard E, Tessier N, Martel C. Platelet extracellular vesicles preserve lymphatic endothelial cell integrity and enhance lymphatic vessel function. Commun Biol 2024; 7:975. [PMID: 39128945 PMCID: PMC11317532 DOI: 10.1038/s42003-024-06675-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/02/2024] [Indexed: 08/13/2024] Open
Abstract
Lymphatic vessels are essential for preventing the accumulation of harmful components within peripheral tissues, including the artery wall. Various endogenous mechanisms maintain adequate lymphatic function throughout life, with platelets being essential for preserving lymphatic vessel integrity. However, since lymph lacks platelets, their impact on the lymphatic system has long been viewed as restricted to areas where lymphatics intersect with blood vessels. Nevertheless, platelets can also exert long range effects through the release of extracellular vesicles (EVs) upon activation. We observed that platelet EVs (PEVs) are present in lymph, a compartment to which they could transfer regulatory effects of platelets. Here, we report that PEVs in lymph exhibit a distinct signature enabling them to interact with lymphatic endothelial cells (LECs). In vitro experiments show that the internalization of PEVs by LECs maintains their functional integrity. Treatment with PEVs improves lymphatic contraction capacity in atherosclerosis-prone mice. We suggest that boosting lymphatic pumping with exogenous PEVs offers a novel therapeutic approach for chronic inflammatory diseases characterized by defective lymphatics.
Collapse
Affiliation(s)
- Laurent Vachon
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Gabriel Jean
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Sara Babran
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Elizabeth Lacroix
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | | | - Janusz Rak
- McGill University and Research, Institute of the McGill University Health Centre, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Antonio Nanci
- Department of Stomatology, Faculty of Dental Medicine, Université de Montréal, Montreal, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | | | - Jean-Claude Tardif
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | - Matthieu Ruiz
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Metabolomics platform, Montreal, Canada
| | - Eric Boilard
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.
- Montreal Heart Institute, Montreal, Canada.
| |
Collapse
|
11
|
Cheung SWY, Chamley LW, Barrett CJ, Lau SYS. Extracellular vesicles and their effect on vascular haemodynamics: a systematic review. Hypertens Res 2024; 47:1588-1606. [PMID: 38600279 PMCID: PMC11150158 DOI: 10.1038/s41440-024-01659-x] [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: 07/12/2023] [Revised: 02/03/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Extracellular vesicles (EVs) are released from all cell types studied to date and act as intercellular communicators containing proteins, nucleic acids and lipid cargos. They have been shown to be involved in maintaining homoeostasis as well as playing a role in the development of pathology including hypertension and cardiovascular disease. It is estimated that there is 109-1010 circulating EVs/mL in the plasma of healthy individuals derived from various sources. While the effect of EVs on vascular haemodynamic parameters will be dependent on the details of the model studied, we systematically searched and summarized current literature to find patterns in how exogenously injected EVs affected vascular haemodynamics. Under homoeostatic conditions, evidence from wire and pressure myography data demonstrate that injecting isolated EVs derived from cell types found in blood and blood vessels resulted in the impairment of vasodilation in blood vessels ex vivo. Impaired vasodilation was also observed in rodents receiving intravenous injections of human plasma EVs from cardiovascular diseases including valvular heart disease, acute coronary syndrome, myocardial infarction and end stage renal disease. When EVs were derived from models of metabolic syndromes, such as diabetes, these EVs enhanced vasoconstriction responses in blood vessels ex vivo. There were fewer publications that assessed the effect of EVs in anaesthetised or conscious animals to confirm whether effects on the vasculature observed in ex vivo studies translated into alterations in vascular haemodynamics in vivo. In the available conscious animal studies, the in vivo data did not always align with the ex vivo data. This highlights the importance of in vivo work to determine the effects of EVs on the integrative vascular haemodynamics.
Collapse
Affiliation(s)
- Sharon W Y Cheung
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Hub for Extracellular Vesicle Investigations, The University of Auckland, Auckland, New Zealand
| | - Carolyn J Barrett
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Sien Yee S Lau
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
12
|
Jiang Z, Yu J, Zhou H, Feng J, Xu Z, Wan M, Zhang W, He Y, Jia C, Shao S, Guo H, Liu B. Research hotspots and emerging trends of mesenchymal stem cells in cardiovascular diseases: a bibliometric-based visual analysis. Front Cardiovasc Med 2024; 11:1394453. [PMID: 38873270 PMCID: PMC11169657 DOI: 10.3389/fcvm.2024.1394453] [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: 03/01/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) have important research value and broad application prospects in cardiovascular diseases (CVDs). However, few bibliometric analyses on MSCs in cardiovascular diseases are available. This study aims to provide a thorough review of the cooperation and influence of countries, institutions, authors, and journals in the field of MSCs in cardiovascular diseases, with the provision of discoveries in the latest progress, evolution paths, frontier research hotspots, and future research trends in the regarding field. Methods The articles related to MSCs in cardiovascular diseases were retrieved from the Web of Science. The bibliometric study was performed by CiteSpace and VOSviewer, and the knowledge map was generated based on data obtained from retrieved articles. Results In our study, a total of 4,852 publications launched before August 31, 2023 were accessed through the Web of Science Core Collection (WoSCC) database via our searching strategy. Significant fluctuations in global publications were observed in the field of MSCs in CVDs. China emerged as the nation with the largest number of publications, yet a shortage of high-quality articles was noted. The interplay among countries, institutions, journals and authors is visually represented in the enclosed figures. Importantly, current research trends and hotspots are elucidated. Cluster analysis on references has highlighted the considerable interest in exosomes, extracellular vesicles, and microvesicles. Besides, keywords analysis revealed a strong emphasis on myocardial infarction, therapy, and transplantation. Treatment methods-related keywords were prominent, while keywords associated with extracellular vesicles gathered significant attention from the long-term perspective. Conclusion MSCs in CVDs have become a topic of active research interest, showcasing its latent value and potential. By summarizing the latest progress, identifying the research hotspots, and discussing the future trends in the advancement of MSCs in CVDs, we aim to offer valuable insights for considering research prospects.
Collapse
Affiliation(s)
- Zhihang Jiang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Yu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houle Zhou
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zehui Xu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Melisandre Wan
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing He
- Department of Preventive Medicine, College of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyao Jia
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Shuijin Shao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haidong Guo
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Páramo JA, Cenarro A, Civeira F, Roncal C. Extracellular vesicles in atherosclerosis: Current and forthcoming impact? CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024:S0214-9168(24)00037-8. [PMID: 38714381 DOI: 10.1016/j.arteri.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 05/09/2024]
Abstract
Atherosclerosis is the main pathogenic substrate for cardiovascular diseases (CVDs). Initially categorized as a passive cholesterol storage disease, nowadays, it is considered an active process, identifying inflammation among the key players for its initiation and progression. Despite these advances, patients with CVDs are still at high risk of thrombotic events and death, urging to deepen into the molecular mechanisms underlying atherogenesis, and to identify novel diagnosis and prognosis biomarkers for their stratification. In this context, extracellular vesicles (EVs) have been postulated as an alternative in search of novel biomarkers in atherosclerotic diseases, as well as to investigate the crosstalk between the cells participating in the processes leading to arterial remodelling. EVs are nanosized lipidic particles released by most cell types in physiological and pathological conditions, that enclose lipids, proteins, and nucleic acids from parental cells reflecting their activation status. First considered cellular waste disposal systems, at present, EVs have been recognized as active effectors in a myriad of cellular processes, and as potential diagnosis and prognosis biomarkers also in CVDs. This review summarizes the role of EVs as potential biomarkers of CVDs, and their involvement into the processes leading to atherosclerosis.
Collapse
Affiliation(s)
- José A Páramo
- Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain; Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERCV, ISCIII, Madrid, Spain
| | - Ana Cenarro
- CIBERCV, ISCIII, Madrid, Spain; Hospital Universitario Miguel Servet, Zaragoza, Spain; Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
| | - Fernando Civeira
- CIBERCV, ISCIII, Madrid, Spain; Hospital Universitario Miguel Servet, Zaragoza, Spain; Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
| | - Carmen Roncal
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERCV, ISCIII, Madrid, Spain.
| |
Collapse
|
14
|
González-King H, Rodrigues PG, Albery T, Tangruksa B, Gurrapu R, Silva AM, Musa G, Kardasz D, Liu K, Kull B, Åvall K, Rydén-Markinhuhta K, Incitti T, Sharma N, Graneli C, Valadi H, Petkevicius K, Carracedo M, Tejedor S, Ivanova A, Heydarkhan-Hagvall S, Menasché P, Synnergren J, Dekker N, Wang QD, Jennbacken K. Head-to-head comparison of relevant cell sources of small extracellular vesicles for cardiac repair: Superiority of embryonic stem cells. J Extracell Vesicles 2024; 13:e12445. [PMID: 38711334 DOI: 10.1002/jev2.12445] [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: 11/15/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI.
Collapse
Affiliation(s)
- Hernán González-King
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Patricia G Rodrigues
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Tamsin Albery
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Benyapa Tangruksa
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ramya Gurrapu
- AstraZeneca India Private Limited, Neville Tower 11th Floor, Ramanujan IT SEZ, Rajv Gandhi Salai (OMR), Taramani, Chennai, Tamil Nadu, India
| | - Andreia M Silva
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Anjarium Biosciences AG, Schlieren, Switzerland
| | - Gentian Musa
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Dominika Kardasz
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Kai Liu
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Pharmaceutical Sciences, Advanced Drug Delivery, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Bengt Kull
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Karin Åvall
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Katarina Rydén-Markinhuhta
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Tania Incitti
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Nitin Sharma
- AstraZeneca India Private Limited, Neville Tower 11th Floor, Ramanujan IT SEZ, Rajv Gandhi Salai (OMR), Taramani, Chennai, Tamil Nadu, India
| | - Cecilia Graneli
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kasparas Petkevicius
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Miguel Carracedo
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sandra Tejedor
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Alena Ivanova
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sepideh Heydarkhan-Hagvall
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Chief Medical Office, Global Patient Safety, AstraZeneca, Mölndal, Sweden
| | - Phillipe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Université de Paris, PARCC, INSERM, Paris, France
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Qing-Dong Wang
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Karin Jennbacken
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
15
|
Osorio LA, Lozano M, Soto P, Moreno-Hidalgo V, Arévalo-Gil A, Ramírez-Balaguera A, Hevia D, Cifuentes J, Hidalgo Y, Alcayaga-Miranda F, Pasten C, Morales D, Varela D, Urquidi C, Iturriaga A, Rivera-Palma A, Larrea-Gómez R, Irarrázabal CE. Levels of Small Extracellular Vesicles Containing hERG-1 and Hsp47 as Potential Biomarkers for Cardiovascular Diseases. Int J Mol Sci 2024; 25:4913. [PMID: 38732154 PMCID: PMC11084293 DOI: 10.3390/ijms25094913] [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: 02/12/2024] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 05/13/2024] Open
Abstract
The diagnosis of cardiovascular disease (CVD) is still limited. Therefore, this study demonstrates the presence of human ether-a-go-go-related gene 1 (hERG1) and heat shock protein 47 (Hsp47) on the surface of small extracellular vesicles (sEVs) in human peripheral blood and their association with CVD. In this research, 20 individuals with heart failure and 26 participants subjected to cardiac stress tests were enrolled. The associations between hERG1 and/or Hsp47 in sEVs and CVD were established using Western blot, flow cytometry, electron microscopy, ELISA, and nanoparticle tracking analysis. The results show that hERG1 and Hsp47 were present in sEV membranes, extravesicularly exposing the sequences 430AFLLKETEEGPPATE445 for hERG1 and 169ALQSINEWAAQTT- DGKLPEVTKDVERTD196 for Hsp47. In addition, upon exposure to hypoxia, rat primary cardiomyocytes released sEVs into the media, and human cardiomyocytes in culture also released sEVs containing hERG1 (EV-hERG1) and/or Hsp47 (EV-Hsp47). Moreover, the levels of sEVs increased in the blood when cardiac ischemia was induced during the stress test, as well as the concentrations of EV-hERG1 and EV-Hsp47. Additionally, the plasma levels of EV-hERG1 and EV-Hsp47 decreased in patients with decompensated heart failure (DHF). Our data provide the first evidence that hERG1 and Hsp47 are present in the membranes of sEVs derived from the human cardiomyocyte cell line, and also in those isolated from human peripheral blood. Total sEVs, EV-hERG1, and EV-Hsp47 may be explored as biomarkers for heart diseases such as heart failure and cardiac ischemia.
Collapse
Affiliation(s)
- Luis A. Osorio
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Mauricio Lozano
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Paola Soto
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Viviana Moreno-Hidalgo
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angely Arévalo-Gil
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angie Ramírez-Balaguera
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Daniel Hevia
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Jorge Cifuentes
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Yessia Hidalgo
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Consuelo Pasten
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| | - Danna Morales
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Varela
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Cinthya Urquidi
- Department of Epidemiology and Health Studies, Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Andrés Iturriaga
- Departamento de Matemática y Ciencia de la Computación, Facultad de Ciencia, Universidad de Santiago de Chile, Santiago 9170020, Chile
| | | | | | - Carlos E. Irarrázabal
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| |
Collapse
|
16
|
Li J, Lu S, Chen F, Zhu H. Unveiling the hidden role of extracellular vesicles in brain metastases: a comprehensive review. Front Immunol 2024; 15:1388574. [PMID: 38726015 PMCID: PMC11079170 DOI: 10.3389/fimmu.2024.1388574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Background Extracellular vesicles (EVs) are small, transparent vesicles that can be found in various biological fluids and are derived from the amplification of cell membranes. Recent studies have increasingly demonstrated that EVs play a crucial regulatory role in tumorigenesis and development, including the progression of metastatic tumors in distant organs. Brain metastases (BMs) are highly prevalent in patients with lung cancer, breast cancer, and melanoma, and patients often experience serious complications and are often associated with a poor prognosis. The immune microenvironment of brain metastases was different from that of the primary tumor. Nevertheless, the existing review on the role and therapeutic potential of EVs in immune microenvironment of BMs is relatively limited. Main body This review provides a comprehensive analysis of the published research literature, summarizing the vital role of EVs in BMs. Studies have demonstrated that EVs participate in the regulation of the BMs immune microenvironment, exemplified by their ability to modify the permeability of the blood-brain barrier, change immune cell infiltration, and activate associated cells for promoting tumor cell survival and proliferation. Furthermore, EVs have the potential to serve as biomarkers for disease surveillance and prediction of BMs. Conclusion Overall, EVs play a key role in the regulation of the immune microenvironment of brain metastasis and are expected to make advances in immunotherapy and disease diagnosis. Future studies will help reveal the specific mechanisms of EVs in brain metastases and use them as new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
17
|
Yao C, Zhang H, Wang C. Recent advances in therapeutic engineered extracellular vesicles. NANOSCALE 2024; 16:7825-7840. [PMID: 38533676 DOI: 10.1039/d3nr05470e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Extracellular vesicles (EVs) are natural particles secreted by living cells, which hold significant potential for various therapeutic applications. Native EVs have specific components and structures, allowing them to cross biological barriers, and circulate in vivo for a long time. Native EVs have also been bioengineered to enhance their therapeutic efficacy and targeting affinity. Recently, the therapeutic potential of surface-engineered EVs has been explored in the treatment of tumors, autoimmune diseases, infections and other diseases by ongoing research and clinical trials. In this review, we will introduce the modified methods of engineered EVs, summarize the application of engineered EVs in preclinical and clinical trials, and discuss the opportunities and challenges for the clinical translation of surface-engineered EVs.
Collapse
Affiliation(s)
- Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
18
|
Manzo OL, Nour J, Sasset L, Marino A, Rubinelli L, Palikhe S, Smimmo M, Hu Y, Bucci MR, Borczuk A, Elemento O, Freed JK, Norata GD, Di Lorenzo A. Rewiring Endothelial Sphingolipid Metabolism to Favor S1P Over Ceramide Protects From Coronary Atherosclerosis. Circ Res 2024; 134:990-1005. [PMID: 38456287 PMCID: PMC11009055 DOI: 10.1161/circresaha.123.323826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence correlated changes in bioactive sphingolipids, particularly S1P (sphingosine-1-phosphate) and ceramides, with coronary artery diseases. Furthermore, specific plasma ceramide species can predict major cardiovascular events. Dysfunction of the endothelium lining lesion-prone areas plays a pivotal role in atherosclerosis. Yet, how sphingolipid metabolism and signaling change and contribute to endothelial dysfunction and atherosclerosis remain poorly understood. METHODS We used an established model of coronary atherosclerosis in mice, combined with sphingolipidomics, RNA-sequencing, flow cytometry, and immunostaining to investigate the contribution of sphingolipid metabolism and signaling to endothelial cell (EC) activation and dysfunction. RESULTS We demonstrated that hemodynamic stress induced an early metabolic rewiring towards endothelial sphingolipid de novo biosynthesis, favoring S1P signaling over ceramides as a protective response. This finding is a paradigm shift from the current belief that ceramide accrual contributes to endothelial dysfunction. The enzyme SPT (serine palmitoyltransferase) commences de novo biosynthesis of sphingolipids and is inhibited by NOGO-B (reticulon-4B), an ER membrane protein. Here, we showed that NOGO-B is upregulated by hemodynamic stress in myocardial EC of ApoE-/- mice and is expressed in the endothelium lining coronary lesions in mice and humans. We demonstrated that mice lacking NOGO-B specifically in EC (Nogo-A/BECKOApoE-/-) were resistant to coronary atherosclerosis development and progression, and mortality. Fibrous cap thickness was significantly increased in Nogo-A/BECKOApoE-/- mice and correlated with reduced necrotic core and macrophage infiltration. Mechanistically, the deletion of NOGO-B in EC sustained the rewiring of sphingolipid metabolism towards S1P, imparting an atheroprotective endothelial transcriptional signature. CONCLUSIONS These data demonstrated that hemodynamic stress induced a protective rewiring of sphingolipid metabolism, favoring S1P over ceramide. NOGO-B deletion sustained the rewiring of sphingolipid metabolism toward S1P protecting EC from activation under hemodynamic stress and refraining coronary atherosclerosis. These findings also set forth the foundation for sphingolipid-based therapeutics to limit atheroprogression.
Collapse
Affiliation(s)
- Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jasmine Nour
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Sailesh Palikhe
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Yang Hu
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Alain Borczuk
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Julie K. Freed
- Department of Anesthesiology, Medical College of Wisconsin Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Rd. Milwaukee, WI 53226, USA
| | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
19
|
Guo J, Cui B, Zheng J, Yu C, Zheng X, Yi L, Zhang S, Wang K. Platelet-derived microparticles and their cargos: The past, present and future. Asian J Pharm Sci 2024; 19:100907. [PMID: 38623487 PMCID: PMC11016590 DOI: 10.1016/j.ajps.2024.100907] [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: 07/05/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 04/17/2024] Open
Abstract
All eukaryotic cells can secrete extracellular vesicles, which have a double-membrane structure and are important players in the intercellular communication involved in a variety of important biological processes. Platelets form platelet-derived microparticles (PMPs) in response to activation, injury, or apoptosis. This review introduces the origin, pathway, and biological functions of PMPs and their importance in physiological and pathological processes. In addition, we review the potential applications of PMPs in cancer, vascular homeostasis, thrombosis, inflammation, neural regeneration, biomarkers, and drug carriers to achieve targeted drug delivery. In addition, we comprehensively report on the origin, biological functions, and applications of PMPs. The clinical transformation, high heterogeneity, future development direction, and limitations of the current research on PMPs are also discussed in depth. Evidence has revealed that PMPs play an important role in cell-cell communication, providing clues for the development of PMPs as carriers for relevant cell-targeted drugs. The development history and prospects of PMPs and their cargos are explored in this guidebook.
Collapse
Affiliation(s)
- Jingwen Guo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Bufeng Cui
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Jie Zheng
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chang Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xuran Zheng
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Lixin Yi
- School of Pharmacy, China Medical University, Shenyang 110122, China
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Simeng Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Keke Wang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
20
|
Ma Y, Gu T, He S, He S, Jiang Z. Development of stem cell therapy for atherosclerosis. Mol Cell Biochem 2024; 479:779-791. [PMID: 37178375 DOI: 10.1007/s11010-023-04762-8] [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: 04/10/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Cardiovascular disease (CVD) has a high incidence and low cure rate worldwide, and atherosclerosis (AS) is the main factor inducing cardiovascular disease, of which lipid deposition in the vessel wall is the main marker of AS. Currently, although statins can be used to lower lipids and low-density lipoprotein (LDL) in AS, the cure rate for AS remains low. Therefore, there is an urgent need to develop new therapeutic approaches, and stem cells are now widely studied, while stem cells are a class of cell types that always maintain the ability to differentiate and can differentiate to form other cells and tissues, and stem cell transplantation techniques have shown efficacy in the treatment of other diseases. With the establishment of cellular therapies and continued research in stem cell technology, stem cells are also being used to address the problem of AS. In this paper, we focus on recent research advances in stem cell therapy for AS and briefly summarize the relevant factors that induce the formation of AS. We mainly discuss the efficacy and application prospects of mesenchymal stem cells (MSCs) for the treatment of AS, in addition to the partial role and potential of exosomes in the treatment of AS. Further, provide new ideas for the clinical application of stem cells.
Collapse
Affiliation(s)
- Yun Ma
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhe Gu
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Siqi He
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Shuya He
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhisheng Jiang
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
21
|
Bharti N, Rai MK, Singh S, Agarwal V, Prasad N, Pandey R, Agrawal V. Prognostic significance of circulating microparticles in IgA nephropathy. Int Urol Nephrol 2024; 56:1071-1081. [PMID: 37615844 DOI: 10.1007/s11255-023-03743-6] [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: 04/26/2022] [Accepted: 08/10/2023] [Indexed: 08/25/2023]
Abstract
PURPOSE Endothelial injury, involved in the pathogenesis of renal fibrosis, can generate microparticles (MPs). These are 0.1-1 µm membrane-bound vesicles shed from the damaged or activated cell surfaces. We analyzed the presence of circulating MPs and EnMPs in IgAN and correlated with markers of endothelial injury and disease activity. METHODS The study included 30 IgAN (mean age 31.5 ± 9 years), 25 healthy controls and Lupus nephritis (n = 10) as disease controls. Circulating MPs were quantitated by Flow cytometry and EnMPs were analyzed using anti-CD31-FITC and anti-CD146-PE antibodies. Their levels were correlated with serum von Willebrand Factor, histological Oxford MEST-C score and renal outcome. A prospective validation group of 20 patients of biopsy-proven IgA nephropathy was also included. RESULTS IgAN had significantly higher levels of MPs, EnMPs and vWF compared to controls. On multivariate analysis, plasma levels of total MPs, EnMPs and serum vWF correlated significantly with the presence of hypertension and E1 on histology. E1 and high MPs (> 130 counts/µl) were associated with shorter time to doubling of serum creatinine. MPs cutoff level of 130 counts/µl had a sensitivity of 75%, specificity of 93.3% and diagnostic accuracy of 89.5% for E1 in the validation cohort. CONCLUSION Circulating MPs and EnMPs in IgAN correlate with E1 on histology and have a potential as non-invasive biomarkers to predict disease activity and renal outcome.
Collapse
Affiliation(s)
- Niharika Bharti
- Departments of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Mohit Kumar Rai
- Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Snigdha Singh
- Departments of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Vikas Agarwal
- Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Narayan Prasad
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Rakesh Pandey
- Departments of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Vinita Agrawal
- Departments of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India.
| |
Collapse
|
22
|
Marlęga-Linert J, Gąsecka A, van der Pol E, Kuchta A, Filipiak KJ, Fijałkowski M, Gruchała M, Nieuwland R, Mickiewicz A. Lipoprotein apheresis affects the concentration of extracellular vesicles in patients with elevated lipoprotein (a). Sci Rep 2024; 14:2762. [PMID: 38307884 PMCID: PMC10837138 DOI: 10.1038/s41598-024-51782-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
Lipoprotein apheresis (LA) is a therapeutic option for hyperlipoproteinemia(a) (hyper-Lp(a)) and atherosclerotic cardiovascular disease (ASCVD). LA improves blood rheology, reduces oxidative stress parameters and improves endothelial function. The underlying molecular mechanisms of LA beneficial effects are unknown, but it has been suggested that LA exhibits multiple activities beyond simply removing lipoproteins. We hypothesized that LA removes not only lipoproteins, but also extracellular vesicles (EVs). To test this hypothesis, we performed a prospective study in 22 patients undergoing LA for hyper-Lp(a) and ASCVD. Different EVs subtypes were measured before and directly after LA, and after 7 days. We used calibrated flow cytometry to detect total particle concentration (diameter > ~ 100 nm), total lipoproteins concentration (diameter > 200 nm, RI > 1.51), total EV concentration (diameter > 200 nm, RI < 1.41), concentrations of EVs derived from erythrocytes (CD235a+; diameter > 200 nm, RI < 1.41), leukocytes (CD45+; diameter > 200 nm, RI < 1.41) and platelets (CD61+, PEVs; diameter > 200 nm, RI < 1.41). LA reduced the concentrations of all investigated EVs subtypes and lipoproteins. Lp(a) concentration was lowered by 64.5% [(58% - 71%); p < 0.001]. Plasma concentrations of EVs > 200 nm in diameter derived from platelets (CD61 +), leukocytes (CD45+) and erythrocytes (CD235a+) decreased after single LA procedure by 42.7% [(12.8-54.7); p = 0.005], 42.6% [(29.7-54.1); p = 0.030] and 26.7% [(1.0-62.7); p = 0.018], respectively, compared to baseline. All EV subtypes returned to the baseline concentrations in blood plasma after 7 days. To conclude, LA removes not only Lp(a), but also cell-derived EVs, which may contribute to LA beneficial effects.
Collapse
Affiliation(s)
- Joanna Marlęga-Linert
- First Chair and Department of Cardiology, Medical University of Gdansk, Gdańsk, Poland
| | - Aleksandra Gąsecka
- 1St Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Amsterdam Vesicle Center and Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Edwin van der Pol
- Amsterdam Vesicle Center and Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centres, Amsterdam, The Netherlands
- Biomedical Engineering and Physics, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Krzysztof J Filipiak
- Institute of Clinical Sciences, Maria Skłodowska-Curie Medical Academy in Warsaw, Warsaw, Poland
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Marcin Fijałkowski
- First Chair and Department of Cardiology, Medical University of Gdansk, Gdańsk, Poland
| | - Marcin Gruchała
- First Chair and Department of Cardiology, Medical University of Gdansk, Gdańsk, Poland
| | - Rienk Nieuwland
- Amsterdam Vesicle Center and Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Agnieszka Mickiewicz
- First Chair and Department of Cardiology, Medical University of Gdansk, Gdańsk, Poland.
| |
Collapse
|
23
|
Ashoub MH, Salavatipour MS, Kasgari FH, Valandani HM, Khalilabadi RM. Extracellular microvesicles: biologic properties, biogenesis, and applications in leukemia. Mol Cell Biochem 2024; 479:419-430. [PMID: 37084166 DOI: 10.1007/s11010-023-04734-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
Microvesicles are cellular membrane vesicles of which size is limited to 30-1000 nm. Almost all cells release them in response to activation signals and apoptosis. Their ability for intercellular communication and enhancement of potential for information exchange (between them) has attracted much interest. Their content is affected by the content of the mother cell, which can help identify their origin. Furthermore, these particles can change the physiology of the target cells by transferring a set of molecules to them and changing the epigenetics of the cells by transferring DNA and RNA. These changes can be induced in cells close to the mother and distant cells. Significant activities of these microvesicles are known both in physiological and pathologic conditions. In this regard, we have reviewed these small particle elements, their contents, and the way of synthesis. Finally, we discussed their current known roles to reveal more potential applications in leukemia.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Samareh Salavatipour
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Hoseinpour Kasgari
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hajar Mardani Valandani
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
24
|
Guo Y, Wei R, Deng J, Guo W. Research progress in the management of vascular disease with cannabidiol: a review. J Cardiothorac Surg 2024; 19:6. [PMID: 38172934 PMCID: PMC10765825 DOI: 10.1186/s13019-023-02476-y] [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: 05/25/2023] [Accepted: 12/25/2023] [Indexed: 01/05/2024] Open
Abstract
The morbidity and mortality rates associated with vascular disease (VD) have been gradually increasing. Currently, the most common treatment for VD is surgery, with the progress in drug therapy remaining slow. Cannabidiol (CBD) is a natural extract of Cannabis sativa L. with sedative, analgesic, and nonaddictive properties. CBD binds to 56 cardiovascular-related receptors and exerts extensive regulatory effects on the cardiovascular system, making it a potential pharmacological agent for the management of VD. However, most CBD studies have focused on neurological and cardiac diseases, and research on the management of VD with CBD is still rare. In this review, we summarize the currently available data on CBD in the management of VD, addressing four aspects: the major molecular targets of CBD in VD management, pharmacokinetic properties, therapeutic effects of CBD on common VDs, and side effects. The findings indicate that CBD has anti-anxiety, anti-oxidation, and anti-inflammatory properties and can inhibit abnormal proliferation and apoptosis of vascular smooth muscle and endothelial cells; these effects suggest CBD as a therapeutic agent for atherosclerosis, stress-induced hypertension, diabetes-related vasculopathy, ischemia-reperfusion injury, and vascular damage caused by smoking and alcohol abuse. This study provides a theoretical basis for further research on CBD in the management of VD.
Collapse
Affiliation(s)
- Yilong Guo
- Medical School of Chinese PLA, Beijing, 100037, China
- Department of Vascular and Endovascular Surgery, The First Medical Centre of PLA General Hospital, 28#, Fuxing Road, Beijing, 100037, China
| | - Ren Wei
- Department of Vascular and Endovascular Surgery, The First Medical Centre of PLA General Hospital, 28#, Fuxing Road, Beijing, 100037, China
| | - Jianqing Deng
- Senior Department of Cardiology, The Six Medical Centre of PLA General Hospital, Beijing, 100037, China
| | - Wei Guo
- Medical School of Chinese PLA, Beijing, 100037, China.
- Department of Vascular and Endovascular Surgery, The First Medical Centre of PLA General Hospital, 28#, Fuxing Road, Beijing, 100037, China.
| |
Collapse
|
25
|
Ma J, Wang W, Zhang W, Xu D, Ding J, Wang F, Peng X, Wang D, Li Y. The recent advances in cell delivery approaches, biochemical and engineering procedures of cell therapy applied to coronary heart disease. Biomed Pharmacother 2023; 169:115870. [PMID: 37952359 DOI: 10.1016/j.biopha.2023.115870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
Cell therapy is an important topic in the field of regeneration medicine that is gaining attention within the scientific community. However, its potential for treatment in coronary heart disease (CHD) has yet to be established. Several various strategies, types of cells, routes of distribution, and supporting procedures have been tried and refined to trigger heart rejuvenation in CHD. However, only a few of them result in a real considerable promise for clinical usage. In this review, we give an update on techniques and clinical studies of cell treatment as used to cure CHD that are now ongoing or have been completed in the previous five years. We also highlight the emerging efficacy of stem cell treatment for CHD. We specifically examine and comment on current breakthroughs in cell treatment applied to CHD, including the most effective types of cells, transport modalities, engineering, and biochemical approaches used in this context. We believe the current review will be helpful for the researcher to distill this information and design future studies to overcome the challenges faced by this revolutionary approach for CHD.
Collapse
Affiliation(s)
- Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 13000, China
| | - Wenhai Wang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Wenbin Zhang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Dexin Xu
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Jian Ding
- Department of Electrodiagnosis, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Fang Wang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Xia Peng
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Dahai Wang
- Department of Rehabilitation, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Yanwei Li
- Department of General Practice and Family Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
26
|
Mebarek S, Buchet R, Pikula S, Strzelecka-Kiliszek A, Brizuela L, Corti G, Collacchi F, Anghieri G, Magrini A, Ciancaglini P, Millan JL, Davies O, Bottini M. Do Media Extracellular Vesicles and Extracellular Vesicles Bound to the Extracellular Matrix Represent Distinct Types of Vesicles? Biomolecules 2023; 14:42. [PMID: 38254642 PMCID: PMC10813234 DOI: 10.3390/biom14010042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Mineralization-competent cells, including hypertrophic chondrocytes, mature osteoblasts, and osteogenic-differentiated smooth muscle cells secrete media extracellular vesicles (media vesicles) and extracellular vesicles bound to the extracellular matrix (matrix vesicles). Media vesicles are purified directly from the extracellular medium. On the other hand, matrix vesicles are purified after discarding the extracellular medium and subjecting the cells embedded in the extracellular matrix or bone or cartilage tissues to an enzymatic treatment. Several pieces of experimental evidence indicated that matrix vesicles and media vesicles isolated from the same types of mineralizing cells have distinct lipid and protein composition as well as functions. These findings support the view that matrix vesicles and media vesicles released by mineralizing cells have different functions in mineralized tissues due to their location, which is anchored to the extracellular matrix versus free-floating.
Collapse
Affiliation(s)
- Saida Mebarek
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Rene Buchet
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (S.P.); (A.S.-K.)
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (S.P.); (A.S.-K.)
| | - Leyre Brizuela
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Giada Corti
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
| | - Federica Collacchi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
| | - Genevieve Anghieri
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE113TU, UK; (G.A.); (O.D.)
| | - Andrea Magrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil;
| | - Jose Luis Millan
- Sanford Children’s Health Research Center, Sanford Burnham Prebys, La Jolla, CA 92037, USA;
| | - Owen Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE113TU, UK; (G.A.); (O.D.)
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
- Sanford Children’s Health Research Center, Sanford Burnham Prebys, La Jolla, CA 92037, USA;
| |
Collapse
|
27
|
Olejarz W, Sadowski K, Radoszkiewicz K. Extracellular Vesicles in Atherosclerosis: State of the Art. Int J Mol Sci 2023; 25:388. [PMID: 38203558 PMCID: PMC10779125 DOI: 10.3390/ijms25010388] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/17/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipid accumulation in the arteries, leading to narrowing and thrombosis that causes mortality. Emerging evidence has confirmed that atherosclerosis affects younger people and is involved in the majority of deaths worldwide. EVs are associated with critical steps in atherosclerosis, cholesterol metabolism, immune response, endothelial dysfunction, vascular inflammation, and remodeling. Endothelial cell-derived EVs can interact with platelets and monocytes, thereby influencing endothelial dysfunction, atherosclerotic plaque destabilization, and the formation of thrombus. EVs are potential diagnostic and prognostic biomarkers in atherosclerosis (AS) and cardiovascular disease (CVD). Importantly, EVs derived from stem/progenitor cells are essential mediators of cardiogenesis and cardioprotection and may be used in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
28
|
Zhang X, Sun S, Ren G, Liu W, Chen H. Advances in Intercellular Communication Mediated by Exosomal ncRNAs in Cardiovascular Disease. Int J Mol Sci 2023; 24:16197. [PMID: 38003385 PMCID: PMC10671547 DOI: 10.3390/ijms242216197] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases are a leading cause of worldwide mortality, and exosomes have recently gained attention as key mediators of intercellular communication in these diseases. Exosomes are double-layered lipid vesicles that can carry biomolecules such as miRNAs, lncRNAs, and circRNAs, and the content of exosomes is dependent on the cell they originated from. They can be involved in the pathophysiological processes of cardiovascular diseases and hold potential as diagnostic and monitoring tools. Exosomes mediate intercellular communication, stimulate or inhibit the activity of target cells, and affect myocardial hypertrophy, injury and infarction, ventricular remodeling, angiogenesis, and atherosclerosis. Exosomes can be released from various types of cells, including endothelial cells, smooth muscle cells, cardiomyocytes, fibroblasts, platelets, adipocytes, immune cells, and stem cells. In this review, we highlight the communication between different cell-derived exosomes and cardiovascular cells, with a focus on the roles of RNAs. This provides new insights for further exploring targeted therapies in the clinical management of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China;
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (S.S.); (G.R.)
| | - Shengjie Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (S.S.); (G.R.)
| | - Gang Ren
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (S.S.); (G.R.)
| | - Wujun Liu
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China;
| | - Hong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China;
| |
Collapse
|
29
|
Schmidt C, Weißmüller S, Heinz CC. Multifaceted Tissue-Protective Functions of Polyvalent Immunoglobulin Preparations in Severe Infections-Interactions with Neutrophils, Complement, and Coagulation Pathways. Biomedicines 2023; 11:3022. [PMID: 38002022 PMCID: PMC10669904 DOI: 10.3390/biomedicines11113022] [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: 09/26/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Severe infections induce immune defense mechanisms and initial tissue damage, which produce an inflammatory neutrophil response. Upon dysregulation of these responses, inflammation, further tissue damage, and systemic spread of the pathogen may occur. Subsequent vascular inflammation and activation of coagulation processes may cause microvascular obstruction at sites distal to the primary site of infection. Low immunoglobulin (Ig) M and IgG levels have been detected in patients with severe infections like sCAP and sepsis, associated with increased severity and mortality. Based on Ig's modes of action, supplementation with polyvalent intravenous Ig preparations (standard IVIg or IgM/IgA-enriched Ig preparations) has long been discussed as a treatment option for severe infections. A prerequisite seems to be the timely administration of Ig preparations before excessive tissue damage has occurred and coagulopathy has developed. This review focuses on nonclinical and clinical studies that evaluated tissue-protective activities resulting from interactions of Igs with neutrophils, complement, and the coagulation system. The data indicate that coagulopathy, organ failure, and even death of patients can possibly be prevented by the timely combined interactions of (natural) IgM, IgA, and IgG with neutrophils and complement.
Collapse
Affiliation(s)
- Carolin Schmidt
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| | | | - Corina C Heinz
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| |
Collapse
|
30
|
Abdolalian M, Zarif MN, Javan M. The role of extracellular vesicles on the occurrence of clinical complications in β-thalassemia. Exp Hematol 2023; 127:28-39. [PMID: 37652128 DOI: 10.1016/j.exphem.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/20/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Thalassemia is the most common monogenic disorder of red blood cells (RBCs) caused by defects in the synthesis of globin chains. Thalassemia phenotypes have a wide spectrum of clinical manifestations and vary from severe anemia requiring regular blood transfusions to clinically asymptomatic states. Ineffective erythropoiesis and toxicity caused by iron overload are major factors responsible for various complications in thalassemia patients, especially patients with β-thalassemia major (β-TM). Common complications in patients with thalassemia include iron overload, thrombosis, cardiac morbidity, vascular dysfunction, inflammation, and organ dysfunction. Extracellular vesicles (EVs) are small membrane vesicles released from various cells' plasma membranes due to activation and apoptosis. Based on studies, EVs play a role in various processes, including clot formation, vascular damage, and proinflammatory processes. In recent years, they have also been studied as biomarkers in the diagnosis and prognosis of diseases. Considering the high concentration of EVs in thalassemia and their role in cellular processes, this study reviews the role of EVs in the common complications of patients with β-thalassemia for the first time.
Collapse
Affiliation(s)
- Mehrnaz Abdolalian
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran; Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Mahin Nikogouftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mohammadreza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran; Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
31
|
Slijkhuis N, Towers M, Mirzaian M, Korteland SA, Heijs B, van Gaalen K, Nieuwenhuizen I, Nigg A, van der Heiden K, de Rijke YB, van der Lugt A, Sijbrands EJG, Claude E, van Soest G. Identifying lipid traces of atherogenic mechanisms in human carotid plaque. Atherosclerosis 2023; 385:117340. [PMID: 37913561 DOI: 10.1016/j.atherosclerosis.2023.117340] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND AND AIMS Lipids play an important role in atherosclerotic plaque development and are interesting candidate predictive biomarkers. However, the link between circulating lipids, accumulating lipids in the vessel wall, and plaque destabilization processes in humans remains largely unknown. This study aims to provide new insights into the role of lipids in atherosclerosis using lipidomics and mass spectrometry imaging to investigate lipid signatures in advanced human carotid plaque and plasma samples. METHODS We used lipidomics and desorption electrospray ionization mass spectrometry imaging (DESI-MSI) to investigate lipid signatures of advanced human carotid plaque and plasma obtained from patients who underwent carotid endarterectomy (n = 14 out of 17 whose plaque samples were analyzed by DESI-MSI). Multivariate data analysis and unsupervised clustering were applied to identify lipids that were the most discriminative species between different patterns in plaque and plasma. These patterns were interpreted by quantitative comparison with conventional histology. RESULTS Lipidomics detected more than 300 lipid species in plasma and plaque, with markedly different relative abundances. DESI-MSI visualized the spatial distribution of 611 lipid-related m/z features in plaques, of which 330 m/z features could be assigned based on exact mass, comparison to the lipidomic data, and high mass resolution MSI. Matching spatial lipid patterns to histological areas of interest revealed several molecular species that were colocalized with pertinent disease processes in plaque including specific sphingomyelin and ceramide species with calcification, phospholipids and free fatty acids with inflammation, and triacylglycerols and phosphatidylinositols with fibrin-rich areas. CONCLUSIONS By comparing lipid species in plaque and plasma, we identified those circulating species that were also prominently present in plaque. Quantitative comparison of lipid spectral patterns with histology revealed the presence of specific lipid species in destabilized plaque areas, corroborating previous in vitro and animal studies.
Collapse
Affiliation(s)
- Nuria Slijkhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Mark Towers
- Waters Corporation, Wilmslow, United Kingdom
| | - Mina Mirzaian
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Suze-Anne Korteland
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim van Gaalen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ingeborg Nieuwenhuizen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Alex Nigg
- Optical Imaging Center, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Kim van der Heiden
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Gijs van Soest
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
32
|
Wang C, Liang Q, He S, Zhu J, Lin X, Lin G, Wu D, Zhang W, Wang Z. Role of inflammation and immunity in vascular calcification: a bibliometric and visual analysis, 2000-2022. Front Cardiovasc Med 2023; 10:1258230. [PMID: 37965089 PMCID: PMC10642504 DOI: 10.3389/fcvm.2023.1258230] [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: 07/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Background In recent years, a great deal of research has been done on vascular calcification (VC), and inflammation and immunity have been displayed to play important roles in the mechanism of VC. However, to date, no comprehensive or systematic bibliometric analyses have been conducted on this topic. Methods Articles and reviews on the roles of inflammation and immunity in VC were obtained from the Web of Science Core Collection on August 5, 2022. Four scientometric software packages-HistCite, CiteSpace, VOSviewer, and R-bibliometrix-were used for the bibliometric and knowledge mapping analyses. Results The obtained 1,868 papers were published in 627 academic journals by 9,595 authors of 2,217 institutions from 69 countries. The annual number of publications showed a clear growth trend. The USA and China were the most productive countries. Karolinska Institutet, Harvard University, and the University of Washington were the most active institutions. Stenvinkel P published the most articles, whereas Demer LL received the most citations. Atherosclerosis published the most papers, while Circulation was the most highly cited journal. The largest cluster among the 22 clusters, based on the analysis of co-citations, was osteo-/chondrogenic transdifferentiation. "Vascular calcification," "inflammation," "chronic kidney disease," and "expression" were the main keywords in the field. The keyword "extracellular vesicle" attracted great attention in recent years with the strongest citation burst. Conclusions Osteo-/chondrogenic transdifferentiation is the primary research topic in this field. Extracellular vesicles are expected to become a new research focus for exploring the inflammatory and immune mechanisms of VC.
Collapse
Affiliation(s)
- Chen Wang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jie Zhu
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiafei Lin
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guanwen Lin
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenqi Zhang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
33
|
Zhang S, Yang Y, Lv X, Liu W, Zhu S, Wang Y, Xu H. Unraveling the Intricate Roles of Exosomes in Cardiovascular Diseases: A Comprehensive Review of Physiological Significance and Pathological Implications. Int J Mol Sci 2023; 24:15677. [PMID: 37958661 PMCID: PMC10650316 DOI: 10.3390/ijms242115677] [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: 09/13/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Exosomes, as potent intercellular communication tools, have garnered significant attention due to their unique cargo-carrying capabilities, which enable them to influence diverse physiological and pathological functions. Extensive research has illuminated the biogenesis, secretion, and functions of exosomes. These vesicles are secreted by cells in different states, exerting either protective or harmful biological functions. Emerging evidence highlights their role in cardiovascular disease (CVD) by mediating comprehensive interactions among diverse cell types. This review delves into the significant impacts of exosomes on CVD under stress and disease conditions, including coronary artery disease (CAD), myocardial infarction, heart failure, and other cardiomyopathies. Focusing on the cellular signaling and mechanisms, we explore how exosomes mediate multifaceted interactions, particularly contributing to endothelial dysfunction, oxidative stress, and apoptosis in CVD pathogenesis. Additionally, exosomes show great promise as biomarkers, reflecting differential expressions of NcRNAs (miRNAs, lncRNAs, and circRNAs), and as therapeutic carriers for targeted CVD treatment. However, the specific regulatory mechanisms governing exosomes in CVD remain incomplete, necessitating further exploration of their characteristics and roles in various CVD-related contexts. This comprehensive review aims to provide novel insights into the biological implications of exosomes in CVD and offer innovative perspectives on the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| | - Hongfei Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| |
Collapse
|
34
|
Xiong Z, An Q, Chen L, Xiang Y, Li L, Zheng Y. Cell or cell derivative-laden hydrogels for myocardial infarction therapy: from the perspective of cell types. J Mater Chem B 2023; 11:9867-9888. [PMID: 37751281 DOI: 10.1039/d3tb01411h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Myocardial infarction (MI) is a global cardiovascular disease with high mortality and morbidity. To treat acute MI, various therapeutic approaches have been developed, including cells, extracellular vesicles, and biomimetic nanoparticles. However, the clinical application of these therapies is limited due to low cell viability, inadequate targetability, and rapid elimination from cardiac sites. Injectable hydrogels, with their three-dimensional porous structure, can maintain the biomechanical stabilization of hearts and the transplantation activity of cells. However, they cannot regenerate cardiomyocytes or repair broken hearts. A better understanding of the collaborative relationship between hydrogel delivery systems and cell or cell-inspired therapy will facilitate advancing innovative therapeutic strategies against MI. Following that, from the perspective of cell types, MI progression and recent studies on using hydrogel to deliver cell or cell-derived preparations for MI treatment are discussed. Finally, current challenges and future prospects of cell or cell derivative-laden hydrogels for MI therapy are proposed.
Collapse
Affiliation(s)
- Ziqing Xiong
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yucheng Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China.
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Huang D, Kidd JM, Zou Y, Wu X, Gehr TWB, Li PL, Li G. Regulation of NLRP3 Inflammasome Activation and Inflammatory Exosome Release in Podocytes by Acid Sphingomyelinase During Obesity. Inflammation 2023; 46:2037-2054. [PMID: 37477734 PMCID: PMC10777441 DOI: 10.1007/s10753-023-01861-y] [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: 11/22/2022] [Revised: 05/30/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023]
Abstract
The activation of nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome has been reported to importantly contribute to glomerular inflammation and injury under different pathological conditions such as obesity. However, the mechanism mediating NLRP3 inflammasome activation in podocytes and subsequent glomerular injury remains poorly understood. Given that the ceramide signaling pathway has been reported to be implicated in obesity-related glomerulopathy (ORG), the present study was designed to test whether the ceramide-producing enzyme, acid sphingomyelinase (ASM), determines NLRP3 inflammasome activation and inflammatory exosome release in podocytes leading to glomerular inflammation and injury during ORG. In Smpd1trg/Podocre mice, podocyte-specific overexpression of Smpd1 gene which encodes ASM significantly exaggerated high-fat diet (HFD)-induced NLRP3 inflammasome activation in podocytes and immune cell infiltration in glomeruli compared to WT/WT mice. Smpd1 gene deletion, however, blocked these pathological changes induced by HFD in Smpd1-/- mice. Accompanied with NLRP3 inflammasome activation and glomerular inflammation, urinary excretion of exosomes containing podocyte marker and NLRP3 inflammasome products (IL-1β and IL-18) in Smpd1trg/Podocre mice on the HFD was much higher than that in WT/WT mice. In contrast, Smpd1-/- mice on the HDF had significantly lower urinary exosome excretion than WT/WT mice. Correspondingly, HFD-induced podocyte injury, glomerular sclerosis, and proteinuria were more severe in Smpd1trg/Podocre mice, but milder in Smpd1-/- mice compared to WT/WT mice. Using podocytes isolated from these mice, we demonstrated that visfatin, a prototype pro-inflammatory adipokine, induced NLRP3 inflammasome activation and enrichment of multivesicular bodies (MVBs) containing IL-1β in podocytes, which was much stronger in podocytes from Smpd1trg/Podocre mice, but weaker in those from Smpd1-/- mice than WT/WT podocytes. By quantitative analysis of exosomes, it was found that upon visfatin stimulation, podocytes from Smpd1trg/Podocre mice released much more exosomes containing NLRP3 inflammasome products, but podocytes from Smpd1-/- mice released much less exosomes compared to WT/WT podocytes. Super-resolution microscopy demonstrated that visfatin inhibited lysosome-MVB interaction in podocytes, indicating impaired MVB degradation by lysosome. The inhibition of lysosome-MVB interaction by visfatin was amplified by Smpd1 gene overexpression but attenuated by Smpd1 gene deletion. Taken together, our results suggest that ASM in podocytes is a crucial regulator of NLRP3 inflammasome activation and inflammatory exosome release that instigate glomerular inflammation and injury during obesity.
Collapse
Affiliation(s)
- Dandan Huang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason M Kidd
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yao Zou
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiaoyuan Wu
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd W B Gehr
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
36
|
Vilella-Figuerola A, Cordero A, Mirabet S, Muñoz-García N, Suades R, Padró T, Badimon L. Platelet-Released Extracellular Vesicle Characteristics Differ in Chronic and in Acute Heart Disease. Thromb Haemost 2023; 123:892-903. [PMID: 37075787 DOI: 10.1055/s-0043-57017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), shed in response to cell activation, stress, or injury, are increased in the blood of patients with cardiovascular disease. EVs are characterized by expressing parental-cell antigens, allowing the determination of their cellular origin. Platelet-derived EVs (pEVs) are the most abundant in blood. Although not universally given, EVs generally express phosphatidylserine (PS) in their membrane. OBJECTIVES To investigate pEVs in chronic and acute conditions, such as chronic heart failure (CHF) and first-onset acute coronary syndrome (ACS), in patients treated as per guidelines. METHODS EVs in CHF patients (n = 119), ACS patients (n = 58), their respective controls (non-CHF [n = 21] and non-ACS [n = 24], respectively), and a reference control group (n = 31) were characterized and quantified by flow cytometry, using monoclonal antibodies against platelet antigens, and annexin V (AV) to determine PS exposure. RESULTS CHF patients had higher EVs-PS- numbers, while ACS had predominantly EVs-PS+. In contrast to ACS, CHF patients had significantly reduced numbers of pEVs carrying PECAM and αIIb-integrin epitopes (CD31+/AV+, CD41a+/AV+, and CD31+/CD41a+/AV+), while no differences were observed in P-selectin-rich pEVs (CD62P+/AV+) compared with controls. Additionally, background etiology of CHF (ischemic vs. nonischemic) or ACS type (ST-elevation myocardial infarction [STEMI] vs. non-STEMI [NSTEMI]) did not affect pEV levels. CONCLUSION PS exposure in EV and pEV-release differ between CHF and ACS patients, with tentatively different functional capacities beyond coagulation to inflammation and cross-talk with other cell types.
Collapse
Affiliation(s)
- Alba Vilella-Figuerola
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Alberto Cordero
- Cardiology Department, Hospital Universitario de San Juan, Alicante, Spain
- Unidad de Investigación en Cardiología, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), València, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Sònia Mirabet
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Heart Failure Group, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Natàlia Muñoz-García
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Rosa Suades
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- UAB-Chair Cardiovascular Research, Barcelona, Spain
| |
Collapse
|
37
|
Suades R, Vilella-Figuerola A, Padró T, Mirabet S, Badimon L. Red Blood Cells and Endothelium Derived Circulating Extracellular Vesicles in Health and Chronic Heart Failure: A Focus on Phosphatidylserine Dynamics in Vesiculation. Int J Mol Sci 2023; 24:11824. [PMID: 37511585 PMCID: PMC10380787 DOI: 10.3390/ijms241411824] [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: 06/27/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Circulating extracellular microvesicles (cEVs) are characterised by presenting surface antigens of parental cells. Since their biogenesis involves the translocation of phosphatidylserine (PS) from the inner to the outer leaflet of the plasma membrane, exposed PS has been considered as a recognition hallmark of cEVs. However, not all cEVs externalise PS. In this study, we have phenotypically and quantitatively characterised cEVs by flow cytometry, paying special attention to the proportions of PS in chronic heart failure patients (cHF; n = 119) and a reference non-HF group (n = 21). PS--cEVs were predominantly found in both groups. Parental markers showed differential pattern depending on the PS exposure. Endothelium-derived and connexin 43-rich cEVs were mainly PS--cEVs and significantly increased in cHF. On the contrary, platelet-derived cEVs were mostly PS+ and were increased in the non-HF group. We observed similar levels of PS+- and PS--cEVs in non-HF subjects when analysing immune cell-derived Evs, but there was a subset-specific difference in cHF patients. Indeed, those cEVs carrying CD45+, CD29+, CD11b+, and CD15+ were mainly PS+-cEVs, while those carrying CD14+, CD3+, and CD56+ were mainly PS--cEVs. In conclusion, endothelial and red blood cells are stressed in cHF patients, as detected by a high shedding of cEVs. Despite PS+-cEVs and PS--cEVs representing two distinct cEV populations, their release and potential function as both biomarkers and shuttles for cell communication seem unrelated to their PS content.
Collapse
Affiliation(s)
- Rosa Suades
- Cardiovascular Program ICCC, Research Institute of Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08049 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Alba Vilella-Figuerola
- Cardiovascular Program ICCC, Research Institute of Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08049 Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Research Institute of Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08049 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sonia Mirabet
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Cardiology Department, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute of Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08049 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| |
Collapse
|
38
|
Maalouf KE, Vaine CA, Frederick DM, Yoshinaga A, Obuchi W, Mahjoum S, Nieland L, Al Ali J, Bragg DC, Breakefield XO, Breyne K. Tracking human neurologic disease status in mouse brain/plasma using reporter-tagged, EV-associated biomarkers. Mol Ther 2023; 31:2206-2219. [PMID: 37198883 PMCID: PMC10362415 DOI: 10.1016/j.ymthe.2023.05.011] [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: 11/02/2022] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023] Open
Abstract
X-linked dystonia-parkinsonism (XDP) is a neurodegenerative disease caused by a retrotransposon insertion in intron 32 of the TAF1 gene. This insertion causes mis-splicing of intron 32 (TAF1-32i) and reduced TAF1 levels. TAF1-32i transcript is unique to XDP patient cells and can be detected in their extracellular vesicles (EVs). We engrafted patient and control iPSC-derived neural progenitor cells (hNPCs) into the striatum of mice. To track TAF1-32i transcript spread by EVs, we transduced the brain-implanted hNPCs with a lentiviral construct called ENoMi, which consists of a re-engineered tetraspanin scaffold tagged with bioluminescent and fluorescent reporter proteins under an EF-1α promoter. Alongside this improved detection in ENoMi-hNPCs-derived EVs, their surface allows specific immunocapture purification, thereby facilitating TAF1-32i analysis. Using this ENoMi-labeling method, TAF1-32i was demonstrated in EVs released from XDP hNPCs implanted in mouse brains. Post-implantation of ENoMi-XDP hNPCs, TAF1-32i transcript was retrieved in EVs isolated from mouse brain and blood, and levels increased over time in plasma. We compared and combined our EV isolation technique to analyze XDP-derived TAF1-32i with other techniques, including size exclusion chromatography and Exodisc. Overall, our study demonstrates the successful engraftment of XDP patient-derived hNPCs in mice as a tool for monitoring disease markers with EVs.
Collapse
Affiliation(s)
- Katia E Maalouf
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Christine A Vaine
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Dawn M Frederick
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Akiko Yoshinaga
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wataru Obuchi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shadi Mahjoum
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lisa Nieland
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jamal Al Ali
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - D Cristopher Bragg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Xandra O Breakefield
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA; Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA.
| | - Koen Breyne
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
39
|
Asaro RJ, Profumo E, Buttari B, Cabrales P. The Double-Edged Sword of Erythrocytes in Health and Disease via Their Adhesiveness. Int J Mol Sci 2023; 24:10382. [PMID: 37373527 DOI: 10.3390/ijms241210382] [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: 05/25/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Their widespread presence throughout the vasculature, coupled with their reactivity, and thereby to their potential to release reactive oxidative species, or to utilize their anti-oxidative capacities, has promoted much discussion of the role(s) of red blood cells (RBCs) in the progression of health or, alternatively, a wide range of disease states. Moreover, these role(s) have been linked to the development of adhesiveness and, in fact, thereby to the essential pathway to their eventual clearance, e.g., by macrophages in the spleen. These disparate roles coupled with the mechanisms involved are reviewed and given. Following an analysis, novel perspectives are provided; these perspectives can lead to novel assays for identifying the potential for RBC adhesiveness as suggested herein. We describe this paradigm, that involves RBC adhesiveness, hemolysis, and ghost formation, with examples including, inter alia, the progression of atherosclerosis and the suppression of tumor growth along with other disease states.
Collapse
Affiliation(s)
- Robert J Asaro
- Department of Structural Engineering, University of California, La Jolla, CA 92093-0085, USA
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Pedro Cabrales
- Department of Bioengineering, University of California, La Jolla, CA 92093-0085, USA
| |
Collapse
|
40
|
Petroni D, Fabbri C, Babboni S, Menichetti L, Basta G, Del Turco S. Extracellular Vesicles and Intercellular Communication: Challenges for In Vivo Molecular Imaging and Tracking. Pharmaceutics 2023; 15:1639. [PMID: 37376087 PMCID: PMC10301899 DOI: 10.3390/pharmaceutics15061639] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous class of cell-derived membrane vesicles released by various cell types that serve as mediators of intercellular signaling. When released into circulation, EVs may convey their cargo and serve as intermediaries for intracellular communication, reaching nearby cells and possibly also distant organs. In cardiovascular biology, EVs released by activated or apoptotic endothelial cells (EC-EVs) disseminate biological information at short and long distances, contributing to the development and progression of cardiovascular disease and related disorders. The significance of EC-EVs as mediators of cell-cell communication has advanced, but a thorough knowledge of the role that intercommunication plays in healthy and vascular disease is still lacking. Most data on EVs derive from in vitro studies, but there are still little reliable data available on biodistribution and specific homing EVs in vivo tissues. Molecular imaging techniques for EVs are crucial to monitoring in vivo biodistribution and the homing of EVs and their communication networks both in basal and pathological circumstances. This narrative review provides an overview of EC-EVs, trying to highlight their role as messengers of cell-cell interaction in vascular homeostasis and disease, and describes emerging applications of various imaging modalities for EVs visualization in vivo.
Collapse
Affiliation(s)
- Debora Petroni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Costanza Fabbri
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Serena Babboni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Luca Menichetti
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Giuseppina Basta
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Serena Del Turco
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
41
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [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: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
42
|
Pillai SS, Pereira DG, Zhang J, Huang W, Beg MA, Knaack DA, de Souza Goncalves B, Sahoo D, Silverstein RL, Shapiro JI, Sodhi K, Chen Y. Contribution of adipocyte Na/K-ATPase α1/CD36 signaling induced exosome secretion in response to oxidized LDL. Front Cardiovasc Med 2023; 10:1046495. [PMID: 37180782 PMCID: PMC10174328 DOI: 10.3389/fcvm.2023.1046495] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Adipose tissue constantly secretes adipokines and extracellular vesicles including exosomes to crosstalk with distinct tissues and organs for whole-body homeostasis. However, dysfunctional adipose tissue under chronic inflammatory conditions such as obesity, atherosclerosis, and diabetes shows pro-inflammatory phenotypes accompanied by oxidative stress and abnormal secretion. Nevertheless, molecular mechanisms of how adipocytes are stimulated to secrete exosomes under those conditions remain poorly understood. Methods Mouse and human in vitro cell culture models were used for performing various cellular and molecular studies on adipocytes and macrophages. Statistical analysis was performed using Student's t-test (two-tailed, unpaired, and equal variance) for comparisons between two groups or ANOVA followed by Bonferroni's multiple comparison test for comparison among more than two groups. Results and discussion In this work, we report that CD36, a scavenger receptor for oxidized LDL, formed a signaling complex with another membrane signal transducer Na/K-ATPase in adipocytes. The atherogenic oxidized LDL induced a pro-inflammatory response in in vitro differentiated mouse and human adipocytes and also stimulated the cells to secrete more exosomes. This was largely blocked by either CD36 knockdown using siRNA or pNaKtide, a peptide inhibitor of Na/K-ATPase signaling. These results showed a critical role of the CD36/Na/K-ATPase signaling complex in oxidized LDL-induced adipocyte exosome secretion. Moreover, by co-incubation of adipocyte-derived exosomes with macrophages, we demonstrated that oxidized LDL-induced adipocyte-derived exosomes promoted pro-atherogenic phenotypes in macrophages, including CD36 upregulation, IL-6 secretion, metabolic switch to glycolysis, and mitochondrial ROS production. Altogether, we show here a novel mechanism through which adipocytes increase exosome secretion in response to oxidized LDL and that the secreted exosomes can crosstalk with macrophages, which may contribute to atherogenesis.
Collapse
Affiliation(s)
- Sneha S. Pillai
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Duane G. Pereira
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Jue Zhang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Wenxin Huang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Mirza Ahmar Beg
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Darcy A. Knaack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bruno de Souza Goncalves
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joseph I. Shapiro
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Komal Sodhi
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
43
|
Nawaz M, Heydarkhan‐Hagvall S, Tangruksa B, González‐King Garibotti H, Jing Y, Maugeri M, Kohl F, Hultin L, Reyahi A, Camponeschi A, Kull B, Christoffersson J, Grimsholm O, Jennbacken K, Sundqvist M, Wiseman J, Bidar AW, Lindfors L, Synnergren J, Valadi H. Lipid Nanoparticles Deliver the Therapeutic VEGFA mRNA In Vitro and In Vivo and Transform Extracellular Vesicles for Their Functional Extensions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206187. [PMID: 36806740 PMCID: PMC10131815 DOI: 10.1002/advs.202206187] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/19/2022] [Indexed: 05/19/2023]
Abstract
Lipid nanoparticles (LNPs) are currently used to transport functional mRNAs, such as COVID-19 mRNA vaccines. The delivery of angiogenic molecules, such as therapeutic VEGF-A mRNA, to ischemic tissues for producing new blood vessels is an emerging strategy for the treatment of cardiovascular diseases. Here, the authors deliver VEGF-A mRNA via LNPs and study stoichiometric quantification of their uptake kinetics and how the transport of exogenous LNP-mRNAs between cells is functionally extended by cells' own vehicles called extracellular vesicles (EVs). The results show that cellular uptake of LNPs and their mRNA molecules occurs quickly, and that the translation of exogenously delivered mRNA begins immediately. Following the VEGF-A mRNA delivery to cells via LNPs, a fraction of internalized VEGF-A mRNA is secreted via EVs. The overexpressed VEGF-A mRNA is detected in EVs secreted from three different cell types. Additionally, RNA-Seq analysis reveals that as cells' response to LNP-VEGF-A mRNA treatment, several overexpressed proangiogenic transcripts are packaged into EVs. EVs are further deployed to deliver VEGF-A mRNA in vitro and in vivo. Upon equal amount of VEGF-A mRNA delivery via three EV types or LNPs in vitro, EVs from cardiac progenitor cells are the most efficient in promoting angiogenesis per amount of VEGF-A protein produced. Intravenous administration of luciferase mRNA shows that EVs could distribute translatable mRNA to different organs with the highest amounts of luciferase detected in the liver. Direct injections of VEGF-A mRNA (via EVs or LNPs) into mice heart result in locally produced VEGF-A protein without spillover to liver and circulation. In addition, EVs from cardiac progenitor cells cause minimal production of inflammatory cytokines in cardiac tissue compared with all other treatment types. Collectively, the data demonstrate that LNPs transform EVs as functional extensions to distribute therapeutic mRNA between cells, where EVs deliver this mRNA differently than LNPs.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
| | - Sepideh Heydarkhan‐Hagvall
- BioPharmaceuticals R&DEarly CardiovascularRenal and Metabolism (CVRM)Bioscience CardiovascularAstraZenecaGothenburgMölndal43183Sweden
- Systems Biology Research CenterSchool of BioscienceUniversity of SkövdeSkövdeSE‐54128Sweden
| | - Benyapa Tangruksa
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
- Systems Biology Research CenterSchool of BioscienceUniversity of SkövdeSkövdeSE‐54128Sweden
| | - Hernán González‐King Garibotti
- BioPharmaceuticals R&DEarly CardiovascularRenal and Metabolism (CVRM)Bioscience CardiovascularAstraZenecaGothenburgMölndal43183Sweden
| | - Yujia Jing
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&DAstraZenecaGothenburgMölndal43183Sweden
| | - Marco Maugeri
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
- Safety InnovationsClinical Pharmacology and Safety SciencesR&D AstraZenecaGothenburgMölndal43183Sweden
| | - Franziska Kohl
- BioPharmaceuticals R&DDiscovery SciencesTranslational GenomicsAstraZenecaGothenburgMölndal43183Sweden
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteSolnaStockholm17177Sweden
| | - Leif Hultin
- BioPharmaceuticals R&DClinical Pharmacology and Safety ScienceImaging and Data AnalyticsAstraZenecaGothenburgMölndal43183Sweden
| | - Azadeh Reyahi
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
| | - Bengt Kull
- BioPharmaceuticals R&DEarly CardiovascularRenal and Metabolism (CVRM)Bioscience CardiovascularAstraZenecaGothenburgMölndal43183Sweden
| | - Jonas Christoffersson
- BioPharmaceuticals R&DEarly CardiovascularRenal and Metabolism (CVRM)Bioscience CardiovascularAstraZenecaGothenburgMölndal43183Sweden
- Systems Biology Research CenterSchool of BioscienceUniversity of SkövdeSkövdeSE‐54128Sweden
| | - Ola Grimsholm
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
- Institute of Pathophysiology and Allergy ResearchMedical University of ViennaVienna1090Austria
| | - Karin Jennbacken
- BioPharmaceuticals R&DEarly CardiovascularRenal and Metabolism (CVRM)Bioscience CardiovascularAstraZenecaGothenburgMölndal43183Sweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
| | - John Wiseman
- BioPharmaceuticals R&DDiscovery SciencesTranslational GenomicsAstraZenecaGothenburgMölndal43183Sweden
| | - Abdel Wahad Bidar
- BioPharmaceuticals R&DDiscovery SciencesTranslational GenomicsAstraZenecaGothenburgMölndal43183Sweden
| | - Lennart Lindfors
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&DAstraZenecaGothenburgMölndal43183Sweden
| | - Jane Synnergren
- Systems Biology Research CenterSchool of BioscienceUniversity of SkövdeSkövdeSE‐54128Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburg41345Sweden
| | - Hadi Valadi
- Department of Rheumatology and Inflammation ResearchInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburg41346Sweden
| |
Collapse
|
44
|
Mabrouk M, Guessous F, Naya A, Merhi Y, Zaid Y. The Pathophysiological Role of Platelet-Derived Extracellular Vesicles. Semin Thromb Hemost 2023; 49:279-283. [PMID: 36174608 DOI: 10.1055/s-0042-1756705] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Platelets are very abundant in the blood, where they play a role in hemostasis, inflammation, and immunity. When activated, platelets undergo a conformational change that allows the release of numerous effector molecules as well as the production of extracellular vesicles, which are circulating submicron vesicles (10 to 1,000 nm in diameter) released into the extracellular space. Extracellular vesicles are formed by the budding of platelet and they carry some of its contents, including nucleic acids, surface proteins, and organelles. While platelets cannot cross tissue barriers, platelet-derived extracellular vesicles can enter the lymph, bone marrow, and synovial fluid. This allows the transfer of diverse contents carried by these platelet-derived vesicles to cell recipients and organs inaccessible to platelets where they can perform many functions. This review highlights the importance of these platelet-derived extracellular vesicles under different physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Meryem Mabrouk
- Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.,Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| | - Fadila Guessous
- Research of Center, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Abdallah Naya
- Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| | - Yahye Merhi
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Center, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.,Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| |
Collapse
|
45
|
Davidson SM, Boulanger CM, Aikawa E, Badimon L, Barile L, Binder CJ, Brisson A, Buzas E, Emanueli C, Jansen F, Katsur M, Lacroix R, Lim SK, Mackman N, Mayr M, Menasché P, Nieuwland R, Sahoo S, Takov K, Thum T, Vader P, Wauben MHM, Witwer K, Sluijter JPG. Methods for the identification and characterization of extracellular vesicles in cardiovascular studies: from exosomes to microvesicles. Cardiovasc Res 2023; 119:45-63. [PMID: 35325061 PMCID: PMC10233250 DOI: 10.1093/cvr/cvac031] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized vesicles with a lipid bilayer that are released from cells of the cardiovascular system, and are considered important mediators of intercellular and extracellular communications. Two types of EVs of particular interest are exosomes and microvesicles, which have been identified in all tissue and body fluids and carry a variety of molecules including RNAs, proteins, and lipids. EVs have potential for use in the diagnosis and prognosis of cardiovascular diseases and as new therapeutic agents, particularly in the setting of myocardial infarction and heart failure. Despite their promise, technical challenges related to their small size make it challenging to accurately identify and characterize them, and to study EV-mediated processes. Here, we aim to provide the reader with an overview of the techniques and technologies available for the separation and characterization of EVs from different sources. Methods for determining the protein, RNA, and lipid content of EVs are discussed. The aim of this document is to provide guidance on critical methodological issues and highlight key points for consideration for the investigation of EVs in cardiovascular studies.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, WC1E 6HX London, UK
| | - Chantal M Boulanger
- Université Paris Cité, Paris-Cardiovascular Research Center, INSERM, Paris, France
| | - Elena Aikawa
- Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lina Badimon
- Cardiovascular Science Program-ICCC, IR-Hospital de la Santa Creu i Santa Pau-IIBSantPau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale and Faculty of Biomedical Sciences, Università Svizzera italiana, 6900 Lugano, Switzerland
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Alain Brisson
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Edit Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, HCEMM-SU and ELKH-SE Immune Proteogenomics Extracellular Vesicle Research Group, Budapest, Hungary
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Felix Jansen
- Department of Internal Medicine II, Heart Center, University Hospital Bonn, Bonn, Germany
| | - Miroslava Katsur
- The Hatter Cardiovascular Institute, University College London, WC1E 6HX London, UK
| | - Romaric Lacroix
- Aix Marseille University, INSERM 1263, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
- Department of Haematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Sai Kiang Lim
- Institute of Medical Biology and Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France
- Laboratory of Experimental Cardiology, Department of Cardiology, UMC Utrecht Regenerative Medicine Center and Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rienk Nieuwland
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaloyan Takov
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Pieter Vader
- Université Paris Cité, Paris-Cardiovascular Research Center, INSERM, Paris, France
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marca H M Wauben
- Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 2, Utrecht, The Netherlands
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, Department of Cardiology, UMC Utrecht Regenerative Medicine Center and Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
46
|
Ning Y, Huang P, Chen G, Xiong Y, Gong Z, Wu C, Xu J, Jiang W, Li X, Tang R, Zhang L, Hu M, Xu J, Xu J, Qian H, Jin C, Yang Y. Atorvastatin-pretreated mesenchymal stem cell-derived extracellular vesicles promote cardiac repair after myocardial infarction via shifting macrophage polarization by targeting microRNA-139-3p/Stat1 pathway. BMC Med 2023; 21:96. [PMID: 36927608 PMCID: PMC10022054 DOI: 10.1186/s12916-023-02778-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/10/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (MSCs) pretreated with atorvastatin (ATV) (MSCATV-EV) have a superior cardiac repair effect on acute myocardial infarction (AMI). The mechanisms, however, have not been fully elucidated. This study aims to explore whether inflammation alleviation of infarct region via macrophage polarization plays a key role in the efficacy of MSCATV-EV. METHODS MSCATV-EV or MSC-EV were intramyocardially injected 30 min after coronary ligation in AMI rats. Macrophage infiltration and polarization (day 3), cardiac function (days 0, 3, 7, 28), and infarct size (day 28) were measured. EV small RNA sequencing and bioinformatics analysis were conducted for differentially expressed miRNAs between MSCATV-EV and MSC-EV. Macrophages were isolated from rat bone marrow for molecular mechanism analysis. miRNA mimics or inhibitors were transfected into EVs or macrophages to analyze its effects on macrophage polarization and cardiac repair in vitro and in vivo. RESULTS MSCATV-EV significantly reduced the amount of CD68+ total macrophages and increased CD206+ M2 macrophages of infarct zone on day 3 after AMI compared with MSC-EV group (P < 0.01-0.0001). On day 28, MSCATV-EV much more significantly improved the cardiac function than MSC-EV with the infarct size markedly reduced (P < 0.05-0.0001). In vitro, MSCATV-EV also significantly reduced the protein and mRNA expressions of M1 markers but increased those of M2 markers in lipopolysaccharide-treated macrophages (P < 0.05-0.0001). EV miR-139-3p was identified as a potential cardiac repair factor mediating macrophage polarization. Knockdown of miR-139-3p in MSCATV-EV significantly attenuated while overexpression of it in MSC-EV enhanced the effect on promoting M2 polarization by suppressing downstream signal transducer and activator of transcription 1 (Stat1). Furthermore, MSCATV-EV loaded with miR-139-3p inhibitors decreased while MSC-EV loaded with miR-139-3p mimics increased the expressions of M2 markers and cardioprotective efficacy. CONCLUSIONS We uncovered a novel mechanism that MSCATV-EV remarkably facilitate cardiac repair in AMI by promoting macrophage polarization via miR-139-3p/Stat1 pathway, which has the great potential for clinical translation.
Collapse
Affiliation(s)
- Yu Ning
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Peisen Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guihao Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Yuyan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Zhaoting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Chunxiao Wu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Junyan Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Wenyang Jiang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Xiaosong Li
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Ruijie Tang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Lili Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Mengjin Hu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Jun Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Haiyan Qian
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
47
|
Brown PA, Brown PD. Extracellular vesicles and atherosclerotic peripheral arterial disease. Cardiovasc Pathol 2023; 63:107510. [PMID: 36460259 DOI: 10.1016/j.carpath.2022.107510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Atherogenesis involves a complex multifactorial process including chronic inflammation that requires the participation of several cell types and molecules. In addition to their role in vascular homeostasis, extracellular vesicles also appear to play an important role in atherogenesis, including monocyte transmigration and foam cell formation, SMC proliferation and migration, leukocyte transmigration, and thrombosis. Peripheral arterial disease, a major form of peripheral vascular disease, is characterized by structural or functional impairment of peripheral arterial supply, often secondary to atherosclerosis. Elevated levels of extracellular vesicles have been demonstrated in patients with peripheral arterial disease and implicated in the development of atherosclerosis within peripheral vascular beds. However, extracellular vesicles also appear capable of delivering cargo with atheroprotective effects. This capability has been exploited in vesicles engineered to carry content capable of neovascularization, suggesting potential for therapeutic angiogenesis. This dual capacity holds substantial promise for diagnosis and therapy, including possibly limb- and life-saving options for peripheral arterial disease management.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - Paul D Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica
| |
Collapse
|
48
|
Tinè M, Padrin Y, Bonato M, Semenzato U, Bazzan E, Conti M, Saetta M, Turato G, Baraldo S. Extracellular Vesicles (EVs) as Crucial Mediators of Cell-Cell Interaction in Asthma. Int J Mol Sci 2023; 24:ijms24054645. [PMID: 36902079 PMCID: PMC10003413 DOI: 10.3390/ijms24054645] [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: 02/08/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Asthma is the most common chronic respiratory disorder worldwide and accounts for a huge health and economic burden. Its incidence is rapidly increasing but, in parallel, novel personalized approaches have emerged. Indeed, the improved knowledge of cells and molecules mediating asthma pathogenesis has led to the development of targeted therapies that significantly increased our ability to treat asthma patients, especially in severe stages of disease. In such complex scenarios, extracellular vesicles (EVs i.e., anucleated particles transporting nucleic acids, cytokines, and lipids) have gained the spotlight, being considered key sensors and mediators of the mechanisms controlling cell-to-cell interplay. We will herein first revise the existing evidence, mainly by mechanistic studies in vitro and in animal models, that EV content and release is strongly influenced by the specific triggers of asthma. Current studies indicate that EVs are released by potentially all cell subtypes in the asthmatic airways, particularly by bronchial epithelial cells (with different cargoes in the apical and basolateral side) and inflammatory cells. Such studies largely suggest a pro-inflammatory and pro-remodelling role of EVs, whereas a minority of reports indicate protective effects, particularly by mesenchymal cells. The co-existence of several confounding factors-including technical pitfalls and host and environmental confounders-is still a major challenge in human studies. Technical standardization in isolating EVs from different body fluids and careful selection of patients will provide the basis for obtaining reliable results and extend their application as effective biomarkers in asthma.
Collapse
Affiliation(s)
- Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Ylenia Padrin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
- Pulmonology Unit, Ospedale Cà Foncello, Azienda Unità Locale Socio-Sanitaria 2 Marca Trevigiana, 31100 Treviso, Italy
| | - Umberto Semenzato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy
- Correspondence:
| |
Collapse
|
49
|
Circulating microvesicles correlate with radiation proctitis complication after radiotherapy. Sci Rep 2023; 13:2033. [PMID: 36739457 PMCID: PMC9899237 DOI: 10.1038/s41598-022-21726-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/30/2022] [Indexed: 02/05/2023] Open
Abstract
In a large retrospective study, we assessed the putative use of circulating microvesicles (MVs), as innovative biomarkers of radiation toxicity in a cohort of 208 patients with prostate adenocarcinoma overexposed to radiation. The level of platelet (P)-, monocyte (M)- and endothelial (E)-derived MVs were assessed by flow cytometry. Rectal bleeding toxicity scores were collected at the time of blood sampling and during the routine follow-up and were tested for association with MVs using a multivariate logistic regression. MVs dosimetric correlation was investigated using dose volume histograms information available for a subset of 36 patients. The number of PMVs was significantly increased in patients with highest toxicity grades compared to lower grades. Risk prediction analysis revealed that increased numbers of PMVs, and an increased amount of MMVs relative to EMVs, were associated with worst rectal bleeding grade compared to the time of blood sampling. Moreover, a significant correlation was found between PMV and MMV numbers, with the range of doses up to the median exposure (40 Gy) of bladder/rectum and anterior rectal wall, respectively. MVs could be considered as new biomarkers to improve the identification of patients with high toxicity grade and may be instrumental for the prognosis of radiation therapy complications.
Collapse
|
50
|
Mahdi A, Wodaje T, Kövamees O, Tengbom J, Zhao A, Jiao T, Henricsson M, Yang J, Zhou Z, Nieminen AI, Levin M, Collado A, Brinck J, Pernow J. The red blood cell as a mediator of endothelial dysfunction in patients with familial hypercholesterolemia and dyslipidemia. J Intern Med 2023; 293:228-245. [PMID: 36324273 PMCID: PMC10092865 DOI: 10.1111/joim.13580] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Patients with familial hypercholesterolemia (FH) display high levels of low-density lipoprotein cholesterol (LDL-c), endothelial dysfunction, and increased risk of premature atherosclerosis. We have previously shown that red blood cells (RBCs) from patients with type 2 diabetes induce endothelial dysfunction through increased arginase 1 and reactive oxygen species (ROS). OBJECTIVE To test the hypothesis that RBCs from patients with FH (FH-RBCs) and elevated LDL-c induce endothelial dysfunction. METHODS AND RESULTS FH-RBCs and LDL-c >5.0 mM induced endothelial dysfunction following 18-h incubation with isolated aortic rings from healthy rats compared to FH-RBCs and LDL-c <2.5 mM or RBCs from healthy subjects (H-RBCs). Inhibition of vascular but not RBC arginase attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. Furthermore, arginase 1 but not arginase 2 was elevated in the vasculature of aortic segments after incubation with FH-RBCs and LDL-c >5.0 mM. A superoxide scavenger, present throughout the 18-h incubation, attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. ROS production was elevated in these RBCs in comparison with H-RBCs. Scavenging of vascular ROS through various antioxidants also attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. This was corroborated by an increase in the lipid peroxidation product 4-hydroxynonenal. Lipidomic analysis of RBC lysates did not reveal any significant changes across the groups. CONCLUSION FH-RBCs induce endothelial dysfunction dependent on LDL-c levels via arginase 1 and ROS-dependent mechanisms.
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tigist Wodaje
- Division of Cardiology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Tengbom
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tong Jiao
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anni I Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Malin Levin
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Aida Collado
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Brinck
- Division of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|