1
|
Kovácsházi C, Hambalkó S, Sayour NV, Gergely TG, Brenner GB, Pelyhe C, Kapui D, Weber BY, Hültenschmidt AL, Pállinger É, Buzás EI, Zolcsák Á, Kiss B, Bozó T, Csányi C, Kósa N, Kellermayer M, Farkas R, Karvaly GB, Wynne K, Matallanas D, Ferdinandy P, Giricz Z. Effect of hypercholesterolemia on circulating and cardiomyocyte-derived extracellular vesicles. Sci Rep 2024; 14:12016. [PMID: 38797778 PMCID: PMC11128454 DOI: 10.1038/s41598-024-62689-6] [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: 09/07/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Hypercholesterolemia (HC) induces, propagates and exacerbates cardiovascular diseases via various mechanisms that are yet not properly understood. Extracellular vesicles (EVs) are involved in the pathomechanism of these diseases. To understand how circulating or cardiac-derived EVs could affect myocardial functions, we analyzed the metabolomic profile of circulating EVs, and we performed an in-depth analysis of cardiomyocyte (CM)-derived EVs in HC. Circulating EVs were isolated with Vezics technology from male Wistar rats fed with high-cholesterol or control chow. AC16 human CMs were treated with Remembrane HC supplement and EVs were isolated from cell culture supernatant. The biophysical properties and the protein composition of CM EVs were analyzed. THP1-ASC-GFP cells were treated with CM EVs, and monocyte activation was measured. HC diet reduced the amount of certain phosphatidylcholines in circulating EVs, independently of their plasma level. HC treatment significantly increased EV secretion of CMs and greatly modified CM EV proteome, enriching several proteins involved in tissue remodeling. Regardless of the treatment, CM EVs did not induce the activation of THP1 monocytes. In conclusion, HC strongly affects the metabolome of circulating EVs and dysregulates CM EVs, which might contribute to HC-induced cardiac derangements.
Collapse
Affiliation(s)
- Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Gábor B Brenner
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dóra Kapui
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bennet Y Weber
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | | | - Éva Pállinger
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ádám Zolcsák
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Bálint Kiss
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Csilla Csányi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Nikolett Kósa
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Róbert Farkas
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Gellért B Karvaly
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Kieran Wynne
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Pharmahungary Group, Szeged, Hungary.
| |
Collapse
|
2
|
Bozbas E, Zhou R, Soyama S, Allen-Redpath K, Mitchell JL, Fisk HL, Calder PC, Jones C, Gibbins JM, Fischer R, Hester S, Yaqoob P. Dietary n-3 polyunsaturated fatty acids alter the number, fatty acid profile and coagulatory activity of circulating and platelet-derived extracellular vesicles: a randomized, controlled crossover trial. Am J Clin Nutr 2024; 119:1175-1186. [PMID: 38484976 PMCID: PMC11130656 DOI: 10.1016/j.ajcnut.2024.03.008] [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: 12/01/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are proposed to play a role in the development of cardiovascular diseases (CVDs) and are considered emerging markers of CVDs. n-3 PUFAs are abundant in oily fish and fish oil and are reported to reduce CVD risk, but there has been little research to date examining the effects of n-3 PUFAs on the generation and function of EVs. OBJECTIVES We aimed to investigate the effects of fish oil supplementation on the number, generation, and function of EVs in subjects with moderate risk of CVDs. METHODS A total of 40 participants with moderate risk of CVDs were supplemented with capsules containing either fish oil (1.9 g/d n-3 PUFAs) or control oil (high-oleic safflower oil) for 12 wk in a randomized, double-blind, placebo-controlled crossover intervention study. The effects of fish oil supplementation on conventional CVD and thrombogenic risk markers were measured, along with the number and fatty acid composition of circulating and platelet-derived EVs (PDEVs). PDEV proteome profiles were evaluated, and their impact on coagulation was assessed using assays including fibrin clot formation, thrombin generation, fibrinolysis, and ex vivo thrombus formation. RESULTS n-3 PUFAs decreased the numbers of circulating EVs by 27%, doubled their n-3 PUFA content, and reduced their capacity to support thrombin generation by >20% in subjects at moderate risk of CVDs. EVs derived from n-3 PUFA-enriched platelets in vitro also resulted in lower thrombin generation, but did not alter thrombus formation in a whole blood ex vivo assay. CONCLUSIONS Dietary n-3 PUFAs alter the number, composition, and function of EVs, reducing their coagulatory activity. This study provides clear evidence that EVs support thrombin generation and that this EV-dependent thrombin generation is reduced by n-3 PUFAs, which has implications for prevention and treatment of thrombosis. CLINICAL TRIAL REGISTRY This trial was registered at clinicaltrials.gov as NCT03203512.
Collapse
Affiliation(s)
- Esra Bozbas
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Ruihan Zhou
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Shin Soyama
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Keith Allen-Redpath
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Joanne L Mitchell
- Institute for Cardiovascular and Metabolic Research and School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Helena L Fisk
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Chris Jones
- Institute for Cardiovascular and Metabolic Research and School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research and School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Svenja Hester
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Parveen Yaqoob
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Darabi M, Lhomme M, Ponnaiah M, Pučić-Baković M, Guillas I, Frisdal E, Bittar R, Croyal M, Matheron-Duriez L, Poupel L, Bonnefont-Rousselot D, Frere C, Varret M, Krempf M, Cariou B, Lauc G, Guerin M, Carrie A, Bruckert E, Giral P, Le Goff W, Kontush A. Integrated omics approach for the identification of HDL structure-function relationships in PCSK9-related familial hypercholesterolemia. J Clin Lipidol 2023; 17:643-658. [PMID: 37550151 DOI: 10.1016/j.jacl.2023.07.003] [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: 02/24/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND The role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in dyslipidemia may go beyond its immediate effects on low-density lipoprotein receptor (LDL-R) activity. OBJECTIVE This study aimed to assess PCSK9-derived alterations of high-density lipoprotein (HDL) physiology, which bear a potential to contribute to cardiovascular risk profile. METHODS HDL was isolated from 33 patients with familial autosomal dominant hypercholesterolemia (FH), including those carrying PCSK9 gain-of-function (GOF) genetic variants (FH-PCSK9, n = 11), together with two groups of dyslipidemic patients employed as controls and carrying genetic variants in the LDL-R not treated (ntFH-LDLR, n = 11) and treated (tFH-LDLR, n = 11) with statins, and 11 normolipidemic controls. Biological evaluations paralleled by proteomic, lipidomic and glycomic analyses were applied to characterize functional and compositional properties of HDL. RESULTS Multiple deficiencies in the HDL function were identified in the FH-PCSK9 group relative to dyslipidemic FH-LDLR patients and normolipidemic controls, which involved reduced antioxidative, antiapoptotic, anti-thrombotic and anti-inflammatory activities. By contrast, cellular cholesterol efflux capacity of HDL was unchanged. In addition, multiple alterations of the proteomic, lipidomic and glycomic composition of HDL were found in the FH-PCSK9 group. Remarkably, HDLs from FH-PCSK9 patients were systematically enriched in several lysophospholipids as well as in A2G2S2 (GP13) glycan and apolipoprotein A-IV. Based on network analysis of functional and compositional data, a novel mosaic structure-function model of HDL biology involving FH was developed. CONCLUSION Several metrics of anti-atherogenic HDL functionality are altered in FH-PCSK9 patients paralleled by distinct compositional alterations. These data provide a first-ever overview of the impact of GOF PCSK9 genetic variants on structure-function relationships in HDL.
Collapse
Affiliation(s)
- Maryam Darabi
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France; LPS-BioSciences (Current affiliation of Dr Darabi), Université de Paris-Saclay, Orsay, France
| | - Marie Lhomme
- ICAN Analytics (Dr Lhomme), Lipidomics Core, Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), Paris, France
| | - Maharajah Ponnaiah
- ICAN I/O (Dr Ponnaiah), Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), Paris, France
| | - Maja Pučić-Baković
- Genos Glycoscience Research Laboratory (Drs Pučić-Baković and Lauc), Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Isabelle Guillas
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Eric Frisdal
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Randa Bittar
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France; Department of Metabolic Biochemistry (Drs Bittar and Bonnefont-Rousselot), Pitié-Salpêtrière-Charles Foix Hospital, AP-HP, Paris, France
| | - Mikaël Croyal
- Université de Nantes (Drs Cariou et Croyal), CHU Nantes, CNRS, INSERM, l'Institut du Thorax, F-44000 Nantes, France; Université de Nantes (Dr Croyal), CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, F-44000 Nantes, France; CRNH-Ouest Mass Spectrometry Core Facility (Drs Croyal and Krempf), F-44000 Nantes, France
| | - Lucrèce Matheron-Duriez
- Platform MS3U (Dr Matheron), Institut de Biologie Paris Seine FR 3631, Sorbonne Université, Paris, France
| | - Lucie Poupel
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Dominique Bonnefont-Rousselot
- Department of Metabolic Biochemistry (Drs Bittar and Bonnefont-Rousselot), Pitié-Salpêtrière-Charles Foix Hospital, AP-HP, Paris, France; Université de Paris (Dr Bonnefont-Rousselot), CNRS, INSERM, UTCBS, F-75006 Paris, France
| | - Corinne Frere
- Department of Haematology (Dr Frere), Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Mathilde Varret
- Paris University and Sorbonne Paris Nord University (Dr Varret), National Institute for Health and Medical Research (INSERM, LVTS), F-75018 Paris, France
| | - Michel Krempf
- CRNH-Ouest Mass Spectrometry Core Facility (Drs Croyal and Krempf), F-44000 Nantes, France; Clinique Bretéché (Dr Krempf), Groupe Elsan, Nantes, France
| | - Bertrand Cariou
- Université de Nantes (Drs Cariou et Croyal), CHU Nantes, CNRS, INSERM, l'Institut du Thorax, F-44000 Nantes, France
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory (Drs Pučić-Baković and Lauc), Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Maryse Guerin
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Alain Carrie
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Eric Bruckert
- Endocrinologie Métabolisme et Prévention Cardiovasculaire (Drs Bruckert and Giral), Institut E3M et IHU Cardiométabolique (ICAN), Hôpital Pitié Salpêtrière, Paris, France
| | - Philippe Giral
- Endocrinologie Métabolisme et Prévention Cardiovasculaire (Drs Bruckert and Giral), Institut E3M et IHU Cardiométabolique (ICAN), Hôpital Pitié Salpêtrière, Paris, France
| | - Wilfried Le Goff
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Anatol Kontush
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France.
| |
Collapse
|
6
|
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
|
7
|
Santoyo JM, Noguera JA, Avilés F, Hernández-Caselles T, de Paco-Matallana C, Delgado JL, Cuevas S, Llinás MT, Hernández I. Pravastatin reduces plasma levels of extracellular vesicles in pregnancies at high risk of term preeclampsia. Front Pharmacol 2023; 14:1166123. [PMID: 37426825 PMCID: PMC10323224 DOI: 10.3389/fphar.2023.1166123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: Elevated plasma levels of extracellular vesicles have been associated with impaired placentation, angiogenesis imbalance, intravascular inflammation, and endothelial dysfunction in women with preeclampsia, thus suggesting that circulating vesicles may be a good therapeutic target for the treatment of the disease. Recently, statins have been considered a potential treatment for the prevention of preeclampsia because of their pleiotropic effects, including the improvement of endothelial dysfunction and inhibition of inflammatory responses. However, the effects of these drugs on circulating vesicles concentration in women at risk of preeclampsia have not been established. Herein, we aimed to assess the effects of pravastatin on circulating extracellular vesicle generation in women at high risk of term preeclampsia. Methods: In a sample of 68 singleton pregnant women participating in the multicenter, double-blind, placebo-controlled STATIN trial (Nº EducraCT 2016-005206-19 ISRCTN), 35 women received a placebo and 33 women received a 20 mg/day dose of pravastatin for approximately 3 weeks (from 35 to 37 weeks of gestation until delivery). Large extracellular vesicles were characterized and quantified by flow cytometry using annexin V and cell-specific antibodies directed against platelet, endothelial, leukocyte, and syncytiotrophoblast cell surface markers. Results: In women who received the placebo, a significant increase in the plasma levels of large extracellular vesicles from platelets (34%, p < 0.01), leukocytes (33%, p < 0.01), monocytes (60%, p < 0.01), endothelial cells (40%, p < 0.05), and syncytiotrophoblast cells (22%, p < 0.05) were observed. However, treatment with pravastatin significantly reduced the plasma levels of large extracellular vesicles from platelets (42%, p < 0.001), leukocytes (25%, p < 0.001), monocytes (61%, p < 0.001), endothelial cells (69%, p < 0.001), activated endothelial cells (55%, p < 0.001), and syncytiotrophoblast cells (44%, p < 0.001). Discussion: These results indicate that pravastatin reduces the levels of activated cell-derived membrane vesicles from the maternal vasculature, blood, and placental syncytiotrophoblast of women at high risk of term preeclampsia, suggesting that this statin may be beneficial in reducing endothelial dysfunction and pro-inflammatory and pro-coagulatory state characteristics of the disease.
Collapse
Affiliation(s)
- Jean Michell Santoyo
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - José Antonio Noguera
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Francisco Avilés
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Trinidad Hernández-Caselles
- Department of Biochemistry and Molecular Biology “B” and Immunology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Catalina de Paco-Matallana
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Juan Luis Delgado
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Santiago Cuevas
- Molecular Inflammation Group, Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - M. Teresa Llinás
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Isabel Hernández
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| |
Collapse
|
8
|
Han Z, Liu Q, Li H, Zhang M, You L, Lin Y, Wang K, Gou Q, Wang Z, Zhou S, Cai Y, Yuan L, Chen H. The role of monocytes in thrombotic diseases: a review. Front Cardiovasc Med 2023; 10:1113827. [PMID: 37332592 PMCID: PMC10272466 DOI: 10.3389/fcvm.2023.1113827] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Cardiovascular and cerebrovascular diseases are the number one killer threatening people's life and health, among which cardiovascular thrombotic events are the most common. As the cause of particularly serious cardiovascular events, thrombosis can trigger fatal crises such as acute coronary syndrome (myocardial infarction and unstable angina), cerebral infarction and so on. Circulating monocytes are an important part of innate immunity. Their main physiological functions are phagocytosis, removal of injured and senescent cells and their debris, and development into macrophages and dendritic cells. At the same time, they also participate in the pathophysiological processes of pro-coagulation and anticoagulation. According to recent studies, monocytes have been found to play a significant role in thrombosis and thrombotic diseases of the immune system. In this manuscript, we review the relationship between monocyte subsets and cardiovascular thrombotic events and analyze the role of monocytes in arterial thrombosis and their involvement in intravenous thrombolysis. Finally, we summarize the mechanism and therapeutic regimen of monocyte and thrombosis in hypertension, antiphospholipid syndrome, atherosclerosis, rheumatic heart disease, lower extremity deep venous thrombosis, and diabetic nephropathy.
Collapse
Affiliation(s)
- Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongpeng Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meiqi Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luling You
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiaoyin Gou
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhanzhan Wang
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Shuwei Zhou
- Department of Radiology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - YiJin Cai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Yuan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haoran Chen
- Science and Education Department, Chengdu Xinhua Hospital, Chengdu, China
| |
Collapse
|
9
|
Rossi-Herring G, Belmonte T, Rivas-Urbina A, Benítez S, Rotllan N, Crespo J, Llorente-Cortés V, Sánchez-Quesada JL, de Gonzalo-Calvo D. Circulating lipoprotein-carried miRNome analysis reveals novel VLDL-enriched microRNAs that strongly correlate with the HDL-microRNA profile. Biomed Pharmacother 2023; 162:114623. [PMID: 37023624 DOI: 10.1016/j.biopha.2023.114623] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Lipoproteins have been described as microRNAs (miRNAs) carriers. Unfortunately, the bibliography on this topic is scarce and shows a high variability between independent investigations. In addition, the miRNA profiles of the LDL and VLDL fractions have not been completely elucidated. Here, we profiled the human circulating lipoprotein-carried miRNome. Lipoprotein fractions (VLDL, LDL and HDL) were isolated from the serum of healthy subjects by ultracentrifugation and purified by size-exclusion chromatography. A panel of 179 miRNAs commonly expressed in circulation was evaluated in the lipoprotein fractions using quantitative real-time PCR (qPCR) assays. A total of 14, 4 and 24 miRNAs were stably detected in the VLDL, LDL and HDL fractions, respectively. VLDL- and HDL-miRNA signatures were highly correlated (rho 0.814), and miR-16-5p, miR-142-3p, miR-223-3p and miR-451a were among the top 5 expressed miRNAs in both fractions. miR-125a-5p, miR-335-3p and miR-1260a, were detected in all lipoprotein fractions. miR-107 and miR-221-3p were uniquely detected in the VLDL fraction. HDL showed the larger number of specifically detected miRNAs (n = 13). Enrichment in specific miRNA families and genomic clusters was observed for HDL-miRNAs. Two sequence motifs were also detected for this group of miRNAs. Functional enrichment analysis including the miRNA signatures from each lipoprotein fraction suggested a potential role in mechanistic pathways previously associated with cardiovascular disease: fibrosis, senescence, inflammation, immune response, angiogenesis, and cardiomyopathy. Collectively, our results not only support the role of lipoproteins as circulating miRNA carriers but also describe for the first time the role of VLDL as a miRNA transporter.
Collapse
|
10
|
Increased Plasma Concentrations of Extracellular Vesicles Are Associated with Pro-Inflammatory and Pro-Thrombotic Characteristics of Left and Right Ventricle Mechanical Support Devices. J Cardiovasc Dev Dis 2023; 10:jcdd10010021. [PMID: 36661916 PMCID: PMC9866833 DOI: 10.3390/jcdd10010021] [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: 11/21/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023] Open
Abstract
Mechanical circulatory support (MCS) allows for functional left and right heart ventricle replacement. MCS induces a systemic inflammatory reaction and prothrombotic state leading to an increased risk of thrombus formation. The extracellular vesicles (EVs) are nanoparticles released from active/injured cells characterized by prothrombotic properties. Simple inflammatory parameters from whole blood count analysis have established a clinical role in everyday practice to describe immune-inflammatory activation. We hypothesized that increased plasma concentrations of EVs might be associated with the proinflammatory and pro-thrombotic characteristics of left ventricle assist device (LVAD) and right ventricle assist device (RVAD) devices. We presented a pilot study showing the concentration of peripheral blood serum, right and left ventricle mechanical assist device extracellular concentration in relation to thrombotic complication in patients treated with a biventricular pulsatile assist device (BIVAD). The observation was based on 12 replacements of pulsatile pumps during 175 days of observation. The proinflammatory characteristics of LVAD were noted. The proinflammatory and procoagulant activation by RVAD was observed. The results may provide possible explanations for the worse results of right-sided mechanical supports observed in clinical practice.
Collapse
|
11
|
Badimon L, Padro T, Arderiu G, Vilahur G, Borrell-Pages M, Suades R. Extracellular vesicles in atherothrombosis: From biomarkers and precision medicine to therapeutic targets. Immunol Rev 2022; 312:6-19. [PMID: 35996799 DOI: 10.1111/imr.13127] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of global mortality. Extracellular vesicles (EVs) are small phospholipid vesicles that convey molecular bioactive cargoes and play essential roles in intercellular communication and, hence, a multifaceted role in health and disease. The present review offers a glimpse into the current state and up-to-date concepts on EV field. It also covers their association with several cardiovascular risk factors and ischemic conditions, being subclinical atherosclerosis of utmost relevance for prevention. Interestingly, we show that EVs hold promise as prognostic and diagnostic as well as predictive markers of ASCVD in the precision medicine era. We then report on the role of EVs in atherothrombosis, disentangling the mechanisms involved in the initiation, progression, and complication of atherosclerosis and showing their direct effect in the context of arterial thrombosis. Finally, their potential use for therapeutic intervention is highlighted.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Peng M, Sun R, Hong Y, Wang J, Xie Y, Zhang X, Li J, Guo H, Xu P, Li Y, Wang X, Wan T, Zhao Y, Huang F, Wang Y, Ye R, Liu Q, Liu G, Liu X, Xu G. Extracellular vesicles carrying proinflammatory factors may spread atherosclerosis to remote locations. Cell Mol Life Sci 2022; 79:430. [PMID: 35851433 PMCID: PMC11071964 DOI: 10.1007/s00018-022-04464-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
Most cells involved in atherosclerosis release extracellular vesicles (EVs), which can carry bioactive substances to downstream tissues via circulation. We hypothesized that EVs derived from atherosclerotic plaques could promote atherogenesis in remote locations, a mechanism that mimics the blood metastasis of cancer. Ldlr gene knockout (Ldlr KO) rats were fed on a high cholesterol diet and underwent partial carotid ligation to induce local atherosclerosis. EVs were separated from carotid artery tissues and downstream blood of carotid ligation by centrifugation. MiRNA sequencing and qPCR were then performed to detect miRNA differences in EVs from rats with and without induced carotid atherosclerosis. Biochemical analyses demonstrated that EVs derived from atherosclerosis could increase the expression of ICAM-1, VCAM-1, and E-selectin in endothelial cells in vitro. EVs derived from atherosclerosis contained a higher level of miR-23a-3p than those derived from controls. MiR-23a-3p could promote endothelial inflammation by targeting Dusp5 and maintaining ERK1/2 phosphorylation in vitro. Inhibiting EV release could attenuate atherogenesis and reduce macrophage infiltration in vivo. Intravenously administrating atherosclerotic plaque-derived EVs could induce intimal inflammation, arterial wall thickening and lumen narrowing in the carotids of Ldlr KO rats, while simultaneous injection of miR-23a-3p antagomir could reverse this reaction. The results suggested that EVs may transfer atherosclerosis to remote locations by carrying proinflammatory factors, particularly miR-23a-3p.
Collapse
Affiliation(s)
- Mengna Peng
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Rui Sun
- Department of Neurology, Shanghai Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Jia Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Yi Xie
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiaohao Zhang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Juanji Li
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Hongquan Guo
- Department of Neurology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China
| | - Pengfei Xu
- Division of Life Sciences and Medicine, Stroke Center & Department of Neurology, Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Yunzi Li
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiaoke Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Ting Wan
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Ying Zhao
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Feihong Huang
- Department of Neurology, Guilin People's Hospital, Guilin, 541002, Guangxi, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191, China
- Institute of Cardiovascular Sciences, School of Basic Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Ruidong Ye
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Qian Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191, China
- Institute of Cardiovascular Sciences, School of Basic Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Xinfeng Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China.
- Division of Life Sciences and Medicine, Stroke Center & Department of Neurology, Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230036, Anhui, China.
| | - Gelin Xu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
13
|
Georgatzakou HT, Fortis SP, Papageorgiou EG, Antonelou MH, Kriebardis AG. Blood Cell-Derived Microvesicles in Hematological Diseases and beyond. Biomolecules 2022; 12:803. [PMID: 35740926 PMCID: PMC9220817 DOI: 10.3390/biom12060803] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Microvesicles or ectosomes represent a major type of extracellular vesicles that are formed by outward budding of the plasma membrane. Typically, they are bigger than exosomes but smaller than apoptotic vesicles, although they may overlap with both in size and content. Their release by cells is a means to dispose redundant, damaged, or dangerous material; to repair membrane lesions; and, primarily, to mediate intercellular communication. By participating in these vital activities, microvesicles may impact a wide array of cell processes and, consequently, changes in their concentration or components have been associated with several pathologies. Of note, microvesicles released by leukocytes, red blood cells, and platelets, which constitute the vast majority of plasma microvesicles, change under a plethora of diseases affecting not only the hematological, but also the nervous, cardiovascular, and urinary systems, among others. In fact, there is evidence that microvesicles released by blood cells are significant contributors towards pathophysiological states, having inflammatory and/or coagulation and/or immunomodulatory arms, by either promoting or inhibiting the relative disease phenotypes. Consequently, even though microvesicles are typically considered to have adverse links with disease prognosis, progression, or outcomes, not infrequently, they exert protective roles in the affected cells. Based on these functional relations, microvesicles might represent promising disease biomarkers with diagnostic, monitoring, and therapeutic applications, equally to the more thoroughly studied exosomes. In the current review, we provide a summary of the features of microvesicles released by blood cells and their potential implication in hematological and non-hematological diseases.
Collapse
Affiliation(s)
- Hara T. Georgatzakou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (H.T.G.); (S.P.F.); (E.G.P.)
| | - Sotirios P. Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (H.T.G.); (S.P.F.); (E.G.P.)
| | - Effie G. Papageorgiou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (H.T.G.); (S.P.F.); (E.G.P.)
| | - Marianna H. Antonelou
- Department of Biology, Section of Cell Biology and Biophysics, National & Kapodistrian University of Athens (NKUA), 15784 Athens, Greece
| | - Anastasios G. Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (H.T.G.); (S.P.F.); (E.G.P.)
| |
Collapse
|
14
|
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis. Cells 2022; 11:cells11111845. [PMID: 35681540 PMCID: PMC9180657 DOI: 10.3390/cells11111845] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality all over the world. Extracellular vesicles (EVs), small lipid-bilayer membrane vesicles released by most cellular types, exert pivotal and multifaceted roles in physiology and disease. Emerging evidence emphasizes the importance of EVs in intercellular communication processes with key effects on cell survival, endothelial homeostasis, inflammation, neoangiogenesis, and thrombosis. This review focuses on EVs as effective signaling molecules able to both derail vascular homeostasis and induce vascular dysfunction, inflammation, plaque progression, and thrombus formation as well as drive anti-inflammation, vascular repair, and atheroprotection. We provide a comprehensive and updated summary of the role of EVs in the development or regression of atherosclerotic lesions, highlighting the link between thrombosis and inflammation. Importantly, we also critically describe their potential clinical use as disease biomarkers or therapeutic agents in atherothrombosis.
Collapse
|
15
|
Suades R, Padró T, Vilahur G, Badimon L. Platelet-released extracellular vesicles: the effects of thrombin activation. Cell Mol Life Sci 2022; 79:190. [PMID: 35288766 PMCID: PMC8920058 DOI: 10.1007/s00018-022-04222-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022]
Abstract
Platelets exert fundamental roles in thrombosis, inflammation, and angiogenesis, contributing to different pathologies from cardiovascular diseases to cancer. We previously reported that platelets release extracellular vesicles (pEVs) which contribute to thrombus formation. However, pEV composition remains poorly defined. Indeed, pEV quality and type, rather than quantity, may be relevant in intravascular cross-talk with either circulating or vascular cells. We aimed to define the phenotypic characteristics of pEVs released spontaneously and those induced by thrombin activation to better understand their role in disease dissemination. pEVs obtained from washed platelets from healthy donor blood were characterized by flow cytometry. pEVs from thrombin-activated platelets (T-pEVs) showed higher levels of P-selectin and active form of glycoprotein IIb/IIIa than baseline non-activated platelets (B-pEVs). Following mass spectrometry-based differential proteomic analysis, significant changes in the abundance of proteins secreted in T-pEVs compared to B-pEVs were found. These differential proteins were involved in coagulation, adhesion, cytoskeleton, signal transduction, metabolism, and vesicle-mediated transport. Interestingly, release of proteins relevant for cell adhesion, intrinsic pathway coagulation, and platelet activation signalling was significantly modified by thrombin stimulation. A novel pEV-associated protein (protocadherin-α4) was found to be significantly reduced in T-pEVs showing a shift towards increased expression in the membranes of activated platelets. In summary, platelet activation induced by thrombin triggers the shedding of pEVs with a complex proteomic pattern rich in procoagulant and proadhesive proteins. Crosstalk with other vascular and blood cells in a paracrine regulatory mode could extend the prothrombotic signalling as well as promote proteostasic changes in other cellular types.
Collapse
Affiliation(s)
- Rosa Suades
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain.
- Cardiovascular Research Chair, UAB, Barcelona, Spain.
| |
Collapse
|
16
|
Dow R, Ridger V. Neutrophil microvesicles and their role in disease. Int J Biochem Cell Biol 2021; 141:106097. [PMID: 34655813 DOI: 10.1016/j.biocel.2021.106097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022]
Abstract
Microvesicles are formed through shedding from the plasma membrane, a process shared by almost all human cells. Microvesicles are highly abundant and have been detected in blood, urine, cerebrospinal fluid, and saliva. They contain a library of cargo derived from their parental cell during formation, including proteases, micro-RNAs and lipids and delivery of this parental cell-derived cargo to other cells can alter target cell function and drive disease. Cell specific molecules on the surface of microvesicles, obtained during microvesicle formation, allows their parental cell to be identified and populations of microvesicles to be investigated for roles in the pathogenesis of various diseases. For instance, recent work by our group has identified a role for neutrophil microvesicles in atherosclerosis. Microvesicle profiles could in future be associated with certain diseases and act as a biomarker to allow for earlier diagnosis. This short review will discuss some of the processes central to all microvesicles before focusing on neutrophil microvesicles, their potential role in cardiovascular disease and the mechanisms that may underpin this.
Collapse
Affiliation(s)
- Reece Dow
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Victoria Ridger
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
| |
Collapse
|
17
|
Garcia-Arguinzonis M, Diaz-Riera E, Peña E, Escate R, Juan-Babot O, Mata P, Badimon L, Padro T. Alternative C3 Complement System: Lipids and Atherosclerosis. Int J Mol Sci 2021; 22:ijms22105122. [PMID: 34066088 PMCID: PMC8151937 DOI: 10.3390/ijms22105122] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Familial hypercholesterolemia (FH) is increasingly associated with inflammation, a phenotype that persists despite treatment with lipid lowering therapies. The alternative C3 complement system (C3), as a key inflammatory mediator, seems to be involved in the atherosclerotic process; however, the relationship between C3 and lipids during plaque progression remains unknown. The aim of the study was to investigate by a systems biology approach the role of C3 in relation to lipoprotein levels during atherosclerosis (AT) progression and to gain a better understanding on the effects of C3 products on the phenotype and function of human lipid-loaded vascular smooth muscle cells (VSMCs). By mass spectrometry and differential proteomics, we found the extracellular matrix (ECM) of human aortas to be enriched in active components of the C3 complement system, with a significantly different proteomic signature in AT segments. Thus, C3 products were more abundant in AT-ECM than in macroscopically normal segments. Furthermore, circulating C3 levels were significantly elevated in FH patients with subclinical coronary AT, evidenced by computed tomographic angiography. However, no correlation was identified between circulating C3 levels and the increase in plaque burden, indicating a local regulation of the C3 in AT arteries. In cell culture studies of human VSMCs, we evidenced the expression of C3, C3aR (anaphylatoxin receptor) and the integrin αMβ2 receptor for C3b/iC3b (RT-PCR and Western blot). C3mRNA was up-regulated in lipid-loaded human VSMCs, and C3 protein significantly increased in cell culture supernatants, indicating that the C3 products in the AT-ECM have a local vessel-wall niche. Interestingly, C3a and iC3b (C3 active fragments) have functional effects on VSMCs, significantly reversing the inhibition of VSMC migration induced by aggregated LDL and stimulating cell spreading, organization of F-actin stress fibers and attachment during the adhesion of lipid-loaded human VSMCs. This study, by using a systems biology approach, identified molecular processes involving the C3 complement system in vascular remodeling and in the progression of advanced human atherosclerotic lesions.
Collapse
MESH Headings
- Adult
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Case-Control Studies
- Cell Adhesion
- Cells, Cultured
- Complement C3/metabolism
- Female
- Humans
- Hyperlipoproteinemia Type II/immunology
- Hyperlipoproteinemia Type II/metabolism
- Hyperlipoproteinemia Type II/pathology
- Male
- Middle Aged
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proteome/analysis
- Proteome/metabolism
- Vascular Remodeling
- Wound Healing
- Young Adult
Collapse
Affiliation(s)
- Maisa Garcia-Arguinzonis
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Elisa Diaz-Riera
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Esther Peña
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Escate
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol Juan-Babot
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, 28010 Madrid, Spain;
| | - Lina Badimon
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-935-565-886; Fax: +34-935-565-559
| |
Collapse
|
18
|
Circulating Extracellular Vesicles As Biomarkers and Drug Delivery Vehicles in Cardiovascular Diseases. Biomolecules 2021; 11:biom11030388. [PMID: 33808038 PMCID: PMC8001426 DOI: 10.3390/biom11030388] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are composed of a lipid bilayer containing transmembrane and soluble proteins. Subtypes of EVs include ectosomes (microparticles/microvesicles), exosomes, and apoptotic bodies that can be released by various tissues into biological fluids. EV cargo can modulate physiological and pathological processes in recipient cells through near- and long-distance intercellular communication. Recent studies have shown that origin, amount, and internal cargos (nucleic acids, proteins, and lipids) of EVs are variable under different pathological conditions, including cardiovascular diseases (CVD). The early detection and management of CVD reduce premature morbidity and mortality. Circulating EVs have attracted great interest as a potential biomarker for diagnostics and follow-up of CVD. This review highlights the role of circulating EVs as biomarkers for diagnosis, prognosis, and therapeutic follow-up of CVD, and also for drug delivery. Despite the great potential of EVs as a tool to study the pathophysiology of CVD, further studies are needed to increase the spectrum of EV-associated applications.
Collapse
|
19
|
Konkoth A, Saraswat R, Dubrou C, Sabatier F, Leroyer AS, Lacroix R, Duchez AC, Dignat-George F. Multifaceted role of extracellular vesicles in atherosclerosis. Atherosclerosis 2020; 319:121-131. [PMID: 33261815 DOI: 10.1016/j.atherosclerosis.2020.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/13/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are small vesicles released by the majority of cells in response to cell activation or death stimuli. They are grouped as small EVs or exosomes, large EVs such as microvesicles (MVs) and apoptotic bodies, resulting from distinct mechanisms of generation. EVs are released into the extracellular space, in most human biological fluids and tissues, including atherosclerotic plaques. They transport complex cargo of bioactive molecules, including proteins, lipids and genetic material and are therefore involved in pathophysiological pathways of cell-cell communication. Indeed, EVs are involved in several processes such as inflammation, coagulation, vascular dysfunction, angiogenesis and senescence, contributing to the initiation and progression of atherothrombotic diseases. Consequently, they behave as a determinant of atherosclerotic plaque vulnerability leading to major cardiovascular disorders. Over the last decade, the field of EVs research has grown, highlighting their involvement in atherosclerosis. However, limitations in both detection methodologies and standardisation have hindered implementation of EVs in the clinical settings. This review summarizes the effect of EVs in atherosclerosis development, progression and severity, with specific attention devoted to their ambivalent roles in senescence and hemostasis. This review will also highlight the role of MVs as multifaceted messengers, able to promote or to attenuate atherosclerosis progression. Finally, we will discuss the main technical challenges and prerequisites of standardization for driving EVs to the clinics and delineate their relevance as emergent biomarkers and innovative therapeutic approaches in atherosclerosis.
Collapse
Affiliation(s)
- Akhil Konkoth
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Ronald Saraswat
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Cléa Dubrou
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Florence Sabatier
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | | | - Romaric Lacroix
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | | | - Francoise Dignat-George
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| |
Collapse
|
20
|
Badimon L, Suades R, Vilella-Figuerola A, Crespo J, Vilahur G, Escate R, Padro T, Chiva-Blanch G. Liquid Biopsies: Microvesicles in Cardiovascular Disease. Antioxid Redox Signal 2020; 33:645-662. [PMID: 31696726 DOI: 10.1089/ars.2019.7922] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Circulating microvesicles (cMV) are small (0.1-1 μm) phospholipid-rich blebs released by almost all cell types, and their release increases with cell activation and injury, thus reflecting the state of the cell from which they are originated. Microvesicles (MV) are found in the bloodstream, and they affect the phenotype of recipient cells, after local or systemic circulation, by intercellular transfer of their molecular content. Recent Advances: Several studies suggest the use of cell-specific MV subpopulations as predictive biomarkers for cardiovascular diseases (CVDs) at different stages and degrees of severity. In this review, we describe the state of the art of cMV as noninvasive surrogate biomarkers of vascular injury and dysfunction correlated with poor clinical outcomes in CVD. Critical Issues: Despite the growing body of evidence supporting the importance of cMV as hallmarks of CVD and their utility as biomarkers of CVD, the specific roles of each phenotype of cMV in CVD burden and prognosis still remain to be elucidated and validated in large cohorts. In addition, the development of standardized and reproducible techniques is required to be used as biomarkers for disease progression in the clinical setting. Future Directions: A multipanel approach with specific cMV phenotypes, added to current biomarkers and scores, will undoubtedly provide unique prognostic information to stratify patients for appropriate therapy on the basis of their risk of atherothrombotic disease and will open a new research area as therapeutic targets for CVD. MV will add to the implementation of precision medicine by helping the cellular and molecular characterization of CVD patients.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,Cardiology Unit, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Alba Vilella-Figuerola
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rafael Escate
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| |
Collapse
|
21
|
Arauna D, Chiva-Blanch G, Padró T, Fuentes E, Palomo I, Badimon L. Frail older adults show a distinct plasma microvesicle profile suggesting a prothrombotic and proinflammatory phenotype. J Cell Physiol 2020; 236:2099-2108. [PMID: 32749745 DOI: 10.1002/jcp.29996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
In a global context of advanced aging, geriatric diseases such as frailty syndrome face challenges in the search for biomarkers and preventive strategies. Frailty has been associated with atherothrombotic pathologies. Circulating microvesicles (cMVs), phospholipid-rich vesicles with a size of 0.1-1.0 μm, have been shown to participate in atherothrombosis onset and progression. We have hypothesized that cMVs from platelets, and vascular and immune cells, are increased in frail older adults. To verify this, a prevalent-case control study was designed with 28 frail older and 27 nonfrail older adults older than 64 years. Frailty was defined by Fried's phenotype. Total cMVs, annexin V positive (AV+)-cMVs, and annexin V negative (AV- )-cMVs derived from blood and vascular cells were measured by flow cytometry. In the analysis of total cMVs, the frail group presented higher levels of CD14+ /CD142+ (p = .042), CD41a+ /CD142+ (p = .041), and CD56+ (p = .025), CD14+ cMVs (p = .043), and CD16+ /CD14+ (p = .019) cMVs levels. Within the phosphatidylserine-exposing cMVs (AV+ ), the frail group showed higher CD14+ /AV+ (p = .044), CD9+ /AV+ (p = .031), P2RY12+ /AV+ (p = .028), and CD235a+ /AV+ (p = .043) cMVs concentrations. Finally, within AV- cMVs, the frail group showed higher CD142+ /CD41a+ /AV- cMVs concentrations originated from platelets (p = .027), CD56+ /AV- originated from natural killer cells (p = .022), and CD34+ /AV- cMVs from hematopoietic stem cells (p = .037). In summary, frail older adults present higher concentrations of platelet-, leukocyte-, and hematopoietic cell-derived cMVs compared to robust age-matched older adults. These cMVs may be involved in the deregulation of the immune system, endothelial damage, and increased risk of thrombosis associated with frailty.
Collapse
Affiliation(s)
- Diego Arauna
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Thrombosis Research Center, Medical Technology, Universidad de Talca, Talca, Chile
| | - Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau; IIB-Sant Pau, Barcelona, Spain.,Department of Endocrinology and Nutrition, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau; IIB-Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Thrombosis Research Center, Medical Technology, Universidad de Talca, Talca, Chile
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Thrombosis Research Center, Medical Technology, Universidad de Talca, Talca, Chile
| | - Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau; IIB-Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
22
|
Escate R, Padró T, Suades R, Camino S, Muñiz O, Diaz-Diaz JL, Sionis A, Mata P, Badimon L. High miR-133a levels in the circulation anticipates presentation of clinical events in familial hypercholesterolaemia patients. Cardiovasc Res 2020; 117:109-122. [PMID: 32061123 DOI: 10.1093/cvr/cvaa039] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 11/17/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Presentation of acute events in patients with atherosclerosis remains unpredictable even after controlling for classical risk factors. MicroRNAs (miRNAs) measured in liquid biopsies could be good candidate biomarkers to improve risk prediction. Here, we hypothesized that miRNAs could predict atherosclerotic plaque progression and clinical event presentation in familial hypercholesterolaemia (FH) patients. METHODS AND RESULTS Circulating miRNAs (plasma, exosomes, and microvesicles) were investigated by TaqMan Array and RT-qPCR assays. Patients with genetic diagnosis of FH and healthy relatives from the SAFEHEART cohort were included. A differential signature of 10 miRNA was obtained by comparing two extreme phenotypes consisting of FH patients suffering a cardiovascular event (CVE) within a 8-year follow-up period (FH-CVE, N = 42) and non-FH hypercholesterolaemic relatives from the same cohort, matched for age and treatment, without CVE during the same period (nFH-nCVE, N = 30). The validation studies included two independent groups of patients with FH background (discovery group, N = 89, validation group N = 196), developing a future CVE (FH-CVE) or not (FH-nCVE) within the same time period of follow-up. Of the 10 miRNAs initially selected, miR-133a was significantly higher in FH-CVE than in FH-nCVE patients. Receiver operating characteristic analysis confirmed miR-133a as the best microRNA for predicting CVE in FH patients (0.76 ± 0.054; P < 0.001). Furthermore, Kaplan-Meier and COX analysis showed that high plasma miR-133a levels associated to the higher risk of presenting a CVE within the next 8 years (hazard ratio 3.89, 95% confidence interval 1.88-8.07; P < 0.001). In silico analysis of curate biological interactions related miR-133a with target genes involved in regulation of the cell-membrane lipid-receptor LRP6 and inflammatory cytokines (CXCL8, IL6, and TNF). These predictions were experimentally proven in human macrophages and endothelial cells transfected with agomiR-133a. CONCLUSION Elevated levels of miR-133a in the circulation anticipate those FH patients that are going to present a clinical CVE within the next 2 years (average). Mechanistically, miR-133a is directly related with lipid- and inflammatory signalling in key cells for atherosclerosis progression.
Collapse
Affiliation(s)
- Rafael Escate
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV) Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Padró
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV) Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Suades
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Sandra Camino
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Ovidio Muñiz
- Servicio de Medicina Interna, Hospital Virgen del Rocío, Sevilla, España, Spain
| | | | - Alessandro Sionis
- Cardiology Department, Acute and Intensive Cardiac Care Unit, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Lina Badimon
- Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV) Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
23
|
Vilahur G, Ben-Aicha S, Diaz-Riera E, Badimon L, Padró T. Phytosterols and Inflammation. Curr Med Chem 2020; 26:6724-6734. [PMID: 29932029 DOI: 10.2174/0929867325666180622151438] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 01/27/2018] [Accepted: 01/31/2018] [Indexed: 02/08/2023]
Abstract
Besides the well-characterized effect of foods and supplements enriched with plant sterols/stanols on serum LDL-C concentrations, evidence is now emerging that phytosterols exert beneficial effects on non-lipid variables such as inflammatory and oxidative stress markers, coagulation parameters and endothelial function. This makes sterols and stanols an attractive alternative for dietary interventions in cardiovascular disease prevention, particularly in populations at low or medium risk. This review aims to summarize the current knowledge derived from experimental studies and human data on the anti-inflammatory effects of phytosterols/stanols and their relevance in promoting atheroprotection and preventing cardiovascular disease. The anti-inflammatory effects induced by plant sterols/stanols have been demonstrated in in vitro studies and in experimental animal models. However, not all the beneficial effects seen at an experimental level have translated into clinical benefit. Indeed, clinical studies that evaluate the association between phytosterols consumption and inflammatory variables (CRP and cytokines) are inconsistent and have not yet provided a solid answer. Plant sterols have been proposed as useful adjuncts to statin therapy to further reduce the risk of cardiovascular disease. However, there is limited available data and more research needs to be done.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute - Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Cardiovascular Program ICCC, Research Institute - Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Elisa Diaz-Riera
- Cardiovascular Program ICCC, Research Institute - Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute - Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Barcelona, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Research Institute - Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
24
|
Bratseth V, Margeirsdottir HD, Chiva-Blanch G, Heier M, Solheim S, Arnesen H, Dahl-Jørgensen K, Seljeflot I. Annexin V + Microvesicles in Children and Adolescents with Type 1 Diabetes: A Prospective Cohort Study. J Diabetes Res 2020; 2020:7216863. [PMID: 32309448 PMCID: PMC7149325 DOI: 10.1155/2020/7216863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Type 1 diabetes is a chronic disease including hyperglycemia and accelerated atherosclerosis, with high risk of micro- and macrovascular complications. Circulating microvesicles (cMVs) are procoagulant cell fragments shed during activation/apoptosis and discussed to be markers of vascular dysfunction and hypercoagulability. Limited knowledge exists on hypercoagulability in young diabetics. We aimed to investigate cMVs over a five-year period in children/adolescents with type 1 diabetes compared with controls and any associations with glycemic control and cardiovascular risk factors. We hypothesized increased shedding of cMVs in type 1 diabetes in response to vascular activation. METHODS The cohort included type 1 diabetics (n = 40) and healthy controls (n = 40), mean age 14 years (range 11) at inclusion, randomly selected from the Norwegian Atherosclerosis and Childhood Diabetes (ACD) study. Citrated plasma was prepared and stored at -80°C until cMV analysis by flow cytometry. RESULTS Comparable levels of Annexin V (AV+) cMVs were observed at inclusion. At five-year follow-up, total AV+ cMVs were significantly lower in subjects with type 1 diabetes compared with controls; however, no significant differences were observed after adjusting for covariates. In the type 1 diabetes group, the total AV+, tissue factor-expressing AV+/CD142+, neutrophil-derived AV+/CD15+ and AV+/CD45+/CD15+, and endothelial-derived AV+/CD309+ and CD309+/CD34+ cMVs were inversely correlated with HbA1c (r = -0.437, r = -0.515, r = -0.575, r = -0.529, r = -0.416, and r = -0.445, respectively; all p ≤ 0.01), however, only at inclusion. No significant correlations with cardiovascular risk factors were observed. CONCLUSIONS Children/adolescents with type 1 diabetes show similar levels of AV+ cMVs as healthy controls and limited associations with glucose control. This indicates that our young diabetics on intensive insulin treatment have preserved vascular homeostasis and absence of procoagulant cMVs.
Collapse
Affiliation(s)
- Vibeke Bratseth
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hanna D. Margeirsdottir
- Pediatric Department, Oslo University Hospital Ullevaal, Oslo, Norway
- Oslo Diabetes Research Centre, Oslo, Norway
| | - Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau-IIB Sant Pau, Sant Antoni Maria Claret, 167, 08025 Barcelona, Spain
- Endocrinology and Nutrition Department Institut d' Investigacions Biomediques August Pi Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
- Centro de Investigacion Biomedica en Red Fisiopatologia de la Obesidad y Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain
| | - Martin Heier
- Pediatric Department, Oslo University Hospital Ullevaal, Oslo, Norway
- Oslo Diabetes Research Centre, Oslo, Norway
| | - Svein Solheim
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Pediatric Department, Oslo University Hospital Ullevaal, Oslo, Norway
- Oslo Diabetes Research Centre, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
25
|
Chiva-Blanch G, Badimon L. Cross-Talk between Lipoproteins and Inflammation: The Role of Microvesicles. J Clin Med 2019; 8:E2059. [PMID: 31771128 PMCID: PMC6947387 DOI: 10.3390/jcm8122059] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022] Open
Abstract
Atherothrombosis is the principal underlying cause of cardiovascular disease (CVD). Microvesicles (MV) are small blebs originated by an outward budding at the cell plasma membranes, which are released in normal conditions. However, MV release is increased in pathophysiologic conditions such as CVD. Low density lipoprotein (LDL) and MV contribute to atherothrombosis onset and progression by promoting inflammation and leukocyte recruitment to injured endothelium, as well as by increasing thrombosis and plaque vulnerability. Moreover, (oxidized)LDL induces MV release and vice-versa, perpetuating endothelium injury leading to CVD progression. Therefore, MV and lipoproteins exhibit common features, which should be considered in the interpretation of their respective roles in the pathophysiology of CVD. Understanding the pathways implicated in this process will aid in developing novel therapeutic approaches against atherothrombosis.
Collapse
Affiliation(s)
- Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau—IIB Sant Pau, Sant Antoni Maria Claret, 167, 08025 Barcelona, Spain;
| | - Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau—IIB Sant Pau, Sant Antoni Maria Claret, 167, 08025 Barcelona, Spain;
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
26
|
Peng M, Liu X, Xu G. Extracellular Vesicles as Messengers in Atherosclerosis. J Cardiovasc Transl Res 2019; 13:121-130. [PMID: 31664614 DOI: 10.1007/s12265-019-09923-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/06/2019] [Indexed: 01/31/2023]
Abstract
Atherosclerosis is a major cause of cardiovascular diseases. Most cells involved in atherosclerosis can shed extracellular vesicles (EVs). Both atherogenic factors, such as hypoxia and oxidative stress, and atheroprotective factors, such as laminar blood flow, can influence the production of EV shedding. EVs can carry protein, DNA, mRNA, and noncoding RNA and act as mediators or messengers for cell-to-cell communications. EVs have been proven to promote or inhibit atherogenesis under particular circumstances. Therefore, EVs might be targeted for preventing or treating atherosclerotic diseases. The level of circulating EVs has been associated with the presence, progressiveness, or severity of atherosclerosis. Therefore, EVs may be utilized as indexes for diagnosing and grading atherosclerosis. Here, we reviewed the progress concerning the involvements of EVs in atherogenesis and atheroprotection. We also discussed the potential applications of EVs in managing atherosclerotic diseases.
Collapse
Affiliation(s)
- Mengna Peng
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
27
|
Haller PM, Stojkovic S, Piackova E, Andric T, Wisgrill L, Spittler A, Wojta J, Huber K, Jäger B. The association of P2Y 12 inhibitors with pro-coagulatory extracellular vesicles and microRNAs in stable coronary artery disease. Platelets 2019; 31:497-504. [PMID: 31389740 DOI: 10.1080/09537104.2019.1648780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EV) act as a cellular communication tool by carrying lipids, proteins and micro RNA (miR) between cells, thereby playing a pivotal role in thromboembolic processes. The effect of P2Y12 inhibitors on pro-coagulatory, phosphatidylserine (PS)-expressing EV has been investigated previously, but only in vitro or during confounding clinical conditions, such as acute coronary syndrome. Hence, we enrolled 62 consecutive patients 12 month after percutaneous coronary intervention and stent implantation and consequent treatment with dual-antiplatelet therapy consisting of low-dose aspirin and P2Y12 inhibitors. Blood for platelet function testing and EV and miR measurements was taken on the last day of P2Y12 inhibitor intake (baseline, on-treatment) and 10, 30 and 180 days thereafter (off-treatment). We did not observe any influence of P2Y12 inhibitors on the levels of PS-EV or EV sub-population from platelets, erythrocytes, monocytes or endothelial cells, respectively. There was no relationship between platelet function and EV levels in plasma. However, the association of miR-21 and miR-150 with platelet EVs was significantly different between on- and off-treatment measurements. Hence, our study suggests no influence of P2Y12 inhibition on the count of EVs in plasma, but on the potential cargo of platelet-derived EV.
Collapse
Affiliation(s)
- Paul M Haller
- 3 Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital , Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna , Vienna, Austria
| | - Edita Piackova
- 3 Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital , Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria
| | - Tijana Andric
- 3 Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital , Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna , Vienna, Austria
| | - Andreas Spittler
- Department of Surgery, Research Laboratories, Medical University of Vienna , Vienna, Austria.,Core Facility Flow Cytometry, Medical University of Vienna , Vienna, Austria
| | - Johann Wojta
- Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria.,Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna , Vienna, Austria.,Core Facility Flow Cytometry, Medical University of Vienna , Vienna, Austria
| | - Kurt Huber
- 3 Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital , Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria.,Faculty of Medicine, Sigmund Freud University , Vienna, Austria
| | - Bernhard Jäger
- 3 Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital , Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research , Vienna, Austria.,Faculty of Medicine, Sigmund Freud University , Vienna, Austria
| |
Collapse
|
28
|
Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019; 115:1266-1285. [PMID: 30918936 DOI: 10.1093/cvr/cvz084] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- University Heart Center, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
- Department of Research and Education, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| |
Collapse
|
29
|
Suades R, Padró T, Crespo J, Sionis A, Alonso R, Mata P, Badimon L. Liquid Biopsy of Extracellular Microvesicles Predicts Future Major Ischemic Events in Genetically Characterized Familial Hypercholesterolemia Patients. Arterioscler Thromb Vasc Biol 2019; 39:1172-1181. [DOI: 10.1161/atvbaha.119.312420] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objective—
Circulating microvesicles (cMVs) exert regulatory roles in atherothrombosis. Patients with familial hypercholesterolemia (FH) that are at high risk for premature cardiovascular events (CVEs) have previously shown high levels of cMVs related to disease severity. However, much remains unknown about their value as markers of CVE. We sought to investigate the prognostic cMV signature for future major CVE presentation in patients with FH.
Approach and Results—
Liquid biopsies from genetically characterized patients with FH from the SAFEHEART (Spanish Familial Hypercholesterolemia Cohort Study)-cohort without clinical manifestation of disease at entry that were going to suffer a CVE within a mean period of 3.3±2.6 years postsampling (CVE, N=92) and from age/cardiovascular risk factor/treatment-matched patients with FH that did not suffer an event within the same time-period (non-CVE, N=48) were investigated. cMVs were phenotyped by flow cytometry to identify activated parental cells. Patients with CVE had higher number of overall procoagulant annexin V
+
-cMVs than non-CVE (
P
<0.05). Pan-leukocyte-derived and neutrophil-derived cMVs, as well as activated platelet-derived cMVs, were significantly higher in patients with CVE. Baseline number of cMVs derived from lymphocytes, neutrophils, and activated platelets were positively associated with mortality at follow-up (
P
<0.05). Patient-risk calculated by classical cardiovascular risk-factor scores did not correlate with cMVs. Inclusion of the cMV signature into the SAFEHEART risk model for patients with FH for the prediction of ischemic events increased the area under the curve from 0.603±0.050 to 0.768±0.042 (
P
<0.005).
Conclusions—
Patients with FH who are going to suffer a CVE within a mean period of 3.3 years, despite being treated according to guidelines, have ongoing innate immune cell and platelet activation. The proposed cMV signature is a prognostic marker for accelerated atherosclerosis and clinical event presentation in patients with FH.
Collapse
Affiliation(s)
- Rosa Suades
- From the Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain (R.S., T.P., J.C., L.B.)
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden (R.S.)
| | - Teresa Padró
- From the Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain (R.S., T.P., J.C., L.B.)
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain (T.P., J.C., A.S., L.B.)
| | - Javier Crespo
- From the Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain (R.S., T.P., J.C., L.B.)
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain (T.P., J.C., A.S., L.B.)
| | - Alessandro Sionis
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain (T.P., J.C., A.S., L.B.)
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain (A.S.)
| | - Rodrigo Alonso
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (R.A., P.M.)
- Department of Nutrition, Clínica Las Condes Santiago, Chile (R.A.)
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (R.A., P.M.)
| | - Lina Badimon
- From the Cardiovascular-Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain (R.S., T.P., J.C., L.B.)
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain (T.P., J.C., A.S., L.B.)
- Cardiovascular Research Chair, UAB, Barcelona, Spain (L.B.)
| |
Collapse
|
30
|
Chiva-Blanch G, Padró T, Alonso R, Crespo J, Perez de Isla L, Mata P, Badimon L. Liquid Biopsy of Extracellular Microvesicles Maps Coronary Calcification and Atherosclerotic Plaque in Asymptomatic Patients With Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2019; 39:945-955. [DOI: 10.1161/atvbaha.118.312414] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Heterozygous familial hypercholesterolemia (FH) is the most common genetic disorder associated with premature atherosclerotic cardiovascular disease. Circulating microvesicles (cMV) are released when cells are activated. We investigated whether cMV could provide information on coronary calcification and atherosclerosis in FH patients.
Approach and Results—
Eighty-two patients (mean of 44±9 years old) with molecular diagnosis of heterozygous FH and asymptomatic cardiovascular disease were investigated. Atherosclerotic plaque characterization was performed by computed tomography angiography, and Agatston coronary calcium score and plaque composition sum were calculated. cMV were quantified by flow cytometry using AV (annexin V) and cell surface-specific antibodies. Of the 82 FH patients, 48 presented atherosclerotic plaque. Patients with atherosclerosis were men and older in a higher percentage than patients without atherosclerotic plaque. FH patients with atherosclerotic plaque showed higher levels of total AV
+
cMV, cMV AV
+
from platelet origin, from granulocytes and neutrophils, and cMV AV
+/−
from endothelial cells than FH-patients without atherosclerotic plaque. Plaque composition sum correlated with platelet- and endothelial-derived cMV, and Agatston coronary calcium score correlated with granulocyte-, platelet-, and endothelial-derived cMV. Receiver operating characteristic curve analyses indicated that the cluster of platelet-, granulocyte-, neutrophil, and endothelial-derived cMV considered together, added significant predictive value to the specific SAFEHEART (Spanish Familial Hypercholesterolaemia Cohort Study) risk equation for plaque presence (area under the curve=0.866, 95% CI, 0.775–0.958;
P
<0.0001,
P
=0.030 for the increment of the area under the curve).
Conclusions—
Endothelial-, granulocyte-, neutrophil- and platelet-derived cMV discriminate and map coronary atherosclerotic plaque and calcification in asymptomatic patients with FH. Liquid biopsy of cMV may be a surrogate biomarker of coronary atherosclerotic plaque burden in FH patients.
Collapse
Affiliation(s)
- Gemma Chiva-Blanch
- From the Cardiovascular Science Institute – ICCC; IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain (G.C.-B., T.P., J.C., L.B.)
| | - Teresa Padró
- From the Cardiovascular Science Institute – ICCC; IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain (G.C.-B., T.P., J.C., L.B.)
- CiberCV, Institute Carlos III, Madrid, Spain (T.P., L.B.)
| | - Rodrigo Alonso
- Nutrition Department, Clínica las Condes, Santiago de Chile, Chile (R.A.)
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (R.A., L.P.d.I., P.M.)
| | - Javier Crespo
- From the Cardiovascular Science Institute – ICCC; IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain (G.C.-B., T.P., J.C., L.B.)
| | - Leopoldo Perez de Isla
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (R.A., L.P.d.I., P.M.)
- Cardiology Department, Hospital Clínico San Carlos, IDISSC, Universidad Complutense, Madrid, Spain (L.P.d.I.)
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (R.A., L.P.d.I., P.M.)
| | - Lina Badimon
- From the Cardiovascular Science Institute – ICCC; IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain (G.C.-B., T.P., J.C., L.B.)
- CiberCV, Institute Carlos III, Madrid, Spain (T.P., L.B.)
| |
Collapse
|
31
|
Mata P, Alonso R, Pérez de Isla L. Atherosclerotic cardiovascular disease risk assessment in familial hypercholesterolemia: does one size fit all? Curr Opin Lipidol 2018; 29:445-452. [PMID: 30382952 DOI: 10.1097/mol.0000000000000553] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Familial hypercholesterolemia is a frequent genetic disease associated with lifelong elevation of LDL-cholesterol and premature atherosclerotic cardiovascular disease (ASCVD). Statins are the cornerstone of treatment. However, with the introduction of novel LDL-cholesterol-lowering therapies, it is necessary to identify familial hypercholesterolemia patients presenting a significantly high residual ASCVD risk. The aim of this review is to provide an update on the recent literature concerning cardiovascular risk stratification including the role of coronary imaging. RECENT FINDINGS Several factors have shown to be independent predictors of ASCVD in familial hypercholesterolemia. These include clinical scores with cardiovascular risk factors, coronary imaging and novel protein biomarkers. However, the recent introduction of the SAFEHEART risk-equation (SAFEHEART-RE) could allow a more accurate ASCVD risk prediction in familial hypercholesterolemia. SUMMARY This article highlights the SAFEHEART-RE as a model to predict incident ASCVD in familial hypercholesterolemia. This equation is a simple and widely applicable tool for use in every clinical setting. Furthermore, coronary atherosclerosis assessed by coronary computed-tomographic angiography (coronary-CTA) is independently associated to the cardiovascular risk estimated according to the SAFEHEART-RE. This equation, as well as coronary-CTA and new biomarkers, could increase individual ASCVD risk stratification and could improve the efficiency and the use of new lipid-lowering therapies in familial hypercholesterolemia.
Collapse
Affiliation(s)
- Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Rodrigo Alonso
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
- Nutrition Department, Clínica las Condes, Santiago de Chile, Chile
| | - Leopoldo Pérez de Isla
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
- Cardiology Department, Hospital Clínico San Carlos, IDISSC, Universidad Complutense, Madrid, Spain
| |
Collapse
|
32
|
Sionis A, Suades R, Sans-Roselló J, Sánchez-Martínez M, Crespo J, Padró T, Cubedo J, Ferrero-Gregori A, Vila-Perales M, Duran-Cambra A, Badimon L. Circulating microparticles are associated with clinical severity of persistent ST-segment elevation myocardial infarction complicated with cardiogenic shock. Int J Cardiol 2018; 258:249-256. [PMID: 29544939 DOI: 10.1016/j.ijcard.2017.10.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/12/2017] [Accepted: 10/12/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cardiogenic shock (CS) is the leading cause of death in patients admitted for acute myocardial infarction (MI). Despite the recent advances in reperfusion and medical treatment mortality remains unacceptably high. Whether cells of the blood compartment in CS-patients are activated and release microparticles (cMPs) that may be both messengers and biomarkers of cell damage is not known. We aimed to investigate the cMP subtypes and parental activated cells of ST-elevation MI (STEMI)-patients complicated by CS and that of non-CS STEMI-patients (non-CS) in order to identify a cMP signature that could aid CS patient's risk stratification. METHODS Clinically-characterized STEMI-patients with and without CS (36/group) were included. Treatment was delivered according to guidelines and included primary percutaneous coronary intervention. cMPs were characterized by triple-labeling flow cytometry using Annexin V and cell surface-specific monoclonal antibodies. RESULTS Increased levels of leukocyte-derived (neutrophil and granulocyte origin) and platelet-derived cMPs were detected in CS compared to non-CS patients. A signature of cMPs derived from platelets, leukocytes, and endothelium discriminated CS-patients (AUC of 0.743±0.059 [95% CI: 0.628-0.859], P<0.0001) and predicted mortality in CS (AUC of 0.869±0.06 [95% CI: 0.750-0.988], P<0.0001). In CS-patients, a higher number of platelet- and monocyte-cMPs and of tissue factor-rich cMPs associated to worse myocardial blush grade and thrombolysis in myocardial infarction flow. CONCLUSIONS cMPs derived from proinflammatory and prothrombotic cells were found to be elevated in CS-patients. In treated as per guidelines CS patients, granulocytes and neutrophils remained activated and actively shed cMPs. These cMPs were biomarkers of adverse prognosis in CS. TRANSLATIONAL ASPECT Increased levels of leukocyte and platelet-derived circulating microparticles (cMPs) are found in cardiogenic shock (CS) patients as compared to non-CS patients. In CS-patients, a higher number of platelet- and monocyte-cMPs and a higher number of tissue factor-rich cMPs were associated to worse myocardial reperfusion. A specific prothrombotic and proinflammatory cMPs signature in cardiogenic shock (CS) patients is a potential discriminator and survival prognostic biomarker for CS, which could aid management and improve clinical outcomes.
Collapse
Affiliation(s)
- A Sionis
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; CiberCV, Institute of Health Carlos III, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - R Suades
- ICCC, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - J Sans-Roselló
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - M Sánchez-Martínez
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - J Crespo
- ICCC, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - T Padró
- CiberCV, Institute of Health Carlos III, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; ICCC, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - J Cubedo
- CiberCV, Institute of Health Carlos III, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; ICCC, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - A Ferrero-Gregori
- Epidemiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - M Vila-Perales
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - A Duran-Cambra
- Acute and Intensive Cardiac Care Unit, Cardiology Department, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - L Badimon
- CiberCV, Institute of Health Carlos III, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; ICCC, Hospital Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Cardiovascular Research Chair, UAB, Barcelona, Spain.
| |
Collapse
|
33
|
Escate R, Mata P, Cepeda JM, Padró T, Badimon L. miR-505-3p controls chemokine receptor up-regulation in macrophages: role in familial hypercholesterolemia. FASEB J 2018; 32:601-612. [PMID: 29457550 DOI: 10.1096/fj.201700476rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Familial hypercholesterolemia (FH) conveys a high risk of premature atherosclerosis as a result of lifelong exposure to high LDL cholesterol levels that are not fully reduced by standard-of-care lipid-lowering treatment. Inflammatory mediators have played a role in the progression of atherosclerotic lesions. Here, we investigated whether innate immunity cells in patients with FH have a specific proinflammatory phenotype that is distinct from that of cells in normal participants. To this end, miR-505-3p-a microRNA related to chronic inflammation-and its target genes were investigated in monocyte-derived macrophages (MACs) of patients with FH (FH-MACs) and non-FH controls (co-MACs). On the basis of the profiler PCR array analysis of agomiR-505-3p-transfected MACs, we identified the chemokine receptors, CCR3, CCR4, and CXCR1, as genes that are regulated by miR-505-3p via the transcription factor, RUNX1. miR-505-3p was significantly down-regulated, whereas CCR3, CCR4, CXCR, and RUNX1 were increased in FH-MAC compared with co-MAC, with the increase being more evident in the proinflammatory M1-like FH-MAC. Chemokine receptor levels were unrelated to LDL plasma levels at entry, but correlated with age in patients with FH, not in controls. In summary, we demonstrate for first time to our knowledge that MACs from FH-MACs have an inflammatory phenotype that is characterized by the up-regulation of CCR3, CCR4, and CXCR1 under the control of miR-505-3p. These results suggest a chronic inflammatory condition in FH innate immunity cells that is not reverted by standard lipid-lowering treatment.-Escate, R., Mata, P., Cepeda, J. M., Padró, T., Badimon, L. miR-505-3p controls chemokine receptor up-regulation in macrophages: role in familial hypercholesterolemia.
Collapse
Affiliation(s)
- Rafael Escate
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Jose Maria Cepeda
- Department of Internal Medicine, Hospital Vega Baja, Orihuela, Spain
| | - Teresa Padró
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Chen Y, Li G, Liu ML. Microvesicles as Emerging Biomarkers and Therapeutic Targets in Cardiometabolic Diseases. GENOMICS PROTEOMICS & BIOINFORMATICS 2018; 16:50-62. [PMID: 29462670 PMCID: PMC6000161 DOI: 10.1016/j.gpb.2017.03.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/03/2017] [Accepted: 03/23/2017] [Indexed: 12/20/2022]
Abstract
Microvesicles (MVs, also known as microparticles) are small vesicles that originate from plasma membrane of almost all eukaryotic cells during apoptosis or activation. MVs can serve as extracellular vehicles to transport bioactive molecules from their parental cells to recipient target cells, thereby serving as novel mediators for intercellular communication. Importantly, more and more evidence indicates that MVs could play important roles in early pathogenesis and subsequent progression of cardiovascular and metabolic diseases. Elevated plasma concentrations of MVs, originating from red blood cells, leukocytes, platelets, or other organs and tissues, have been reported in various cardiometabolic diseases. Circulating MVs could serve as potential biomarkers for disease diagnosis or therapeutic monitoring. In this review, we summarized recently-published studies in the field and discussed the role of MVs in the pathogenesis of cardiometabolic diseases. The emerging values of MVs that serve as biomarker for non-invasive diagnosis and prognosis, as well as their roles as novel therapeutic targets in cardiometabolic diseases, were also described.
Collapse
Affiliation(s)
- Yan Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Ming-Lin Liu
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19140, USA; Philadelphia VA Medical Center, Philadelphia, PA 19140, USA.
| |
Collapse
|
35
|
Ganjali S, Momtazi-Borojeni AA, Banach M, Kovanen PT, Gotto AM, Sahebkar A. HDL functionality in familial hypercholesterolemia: effects of treatment modalities and pharmacological interventions. Drug Discov Today 2018; 23:171-180. [DOI: 10.1016/j.drudis.2017.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/02/2017] [Accepted: 09/25/2017] [Indexed: 01/14/2023]
|
36
|
Badimon L, Suades R, Arderiu G, Peña E, Chiva-Blanch G, Padró T. Microvesicles in Atherosclerosis and Angiogenesis: From Bench to Bedside and Reverse. Front Cardiovasc Med 2017; 4:77. [PMID: 29326946 PMCID: PMC5741657 DOI: 10.3389/fcvm.2017.00077] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/22/2017] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis (AT) is a progressive chronic disease involving lipid accumulation, fibrosis, and inflammation in medium and large-sized arteries, and it is the main cause of cardiovascular disease (CVD). AT is caused by dyslipidemia and mediated by both innate and adaptive immune responses. Despite lipid-lowering drugs have shown to decrease the risk of cardiovascular events (CVEs), there is a significant burden of AT-related morbidity and mortality. Identification of subjects at increased risk for CVE as well as discovery of novel therapeutic targets for improved treatment strategies are still unmet clinical needs in CVD. Microvesicles (MVs), small extracellular plasma membrane particles shed by activated and apoptotic cells have been widely linked to the development of CVD. MVs from vascular and resident cells by facilitating exchange of biological information between neighboring cells serve as cellular effectors in the bloodstream and play a key role in all stages of disease progression. This article reviews the current knowledge on the role of MVs in AT and CVD. Attention is focused on novel aspects of MV-mediated regulatory mechanisms from endothelial dysfunction, vascular wall inflammation, oxidative stress, and apoptosis to coagulation and thrombosis in the progression and development of atherothrombosis. MV contribution to vascular remodeling is also discussed, with a particular emphasis on the effect of MVs on the crosstalk between endothelial cells and smooth muscle cells, and their role regulating the active process of AT-driven angiogenesis and neovascularization. This review also highlights the latest findings and main challenges on the potential prognostic, diagnostic, and therapeutic value of cell-derived MVs in CVD. In summary, MVs have emerged as new regulators of biological functions in atherothrombosis and might be instrumental in cardiovascular precision medicine; however, significant efforts are still needed to translate into clinics the latest findings on MV regulation and function.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Arderiu
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| |
Collapse
|
37
|
Li M, Zhang X, Yu J, Wu X, Sun L. Monocyte-Derived Procoagulant Microvesicles Induced by High Glucose Can Be Attenuated by the Antioxidant N-Acetyl-L-Cysteine, Partly Through the P38/MAPK Pathway. Metab Syndr Relat Disord 2017; 15:521-526. [DOI: 10.1089/met.2017.0089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Mingzhen Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Department of Diabetes and Gout, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiao Zhang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Department of Diabetes and Gout, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jing Yu
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Department of Diabetes and Gout, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xin Wu
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Department of Diabetes and Gout, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Lirong Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Department of Diabetes and Gout, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
38
|
Suades R, Padró T, Alonso R, Mata P, Badimon L. High levels of TSP1+/CD142+ platelet-derived microparticles characterise young patients with high cardiovascular risk and subclinical atherosclerosis. Thromb Haemost 2017; 114:1310-21. [DOI: 10.1160/th15-04-0325] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/05/2015] [Indexed: 12/12/2022]
Abstract
SummaryCirculating microparticles (cMPs) play important roles in cellular crosstalk and are messengers of cell activation. We have previously reported that platelet-released microparticles (pMPs) stimulate thrombosis and that lipid-lowering treatment as per guidelines in patients with familial hypercholesterolaemia (FH) is not sufficiently effective in reducing pro-inflammatory cell activation and, consequently, CD45+/CD3+-lymphocyte-derived cMP shedding. FH patients, due to life-long vascular exposure to high LDL-cholesterol levels, are at high cardiovascular risk (HCVR) and develop premature coronary artery disease. Our objectives were to investigate a) whether patients with HCVR have cMPs with a prothrombotic phenotype, and b) whether patients with magnetic resonance imaging (MRI) evidence of lipid-rich atherosclerotic lesions have a specific cMP profile regarding prothrombotic protein cargos. cMPs were isolated from HCVR-patients and from age/gender/treatment-matched control patients. cMP phenotype was characterised by triple-labelling flow cytometry. HCVR--patients have higher numbers of pMPs derived from activated platelets as well as of tissue factor-rich microparticles (TF+-cMPs) than controls (P< 0.0001). TF+-cMPs showed procoagulant activity, which associate with atherosclerotic plaque burden, indicating that TF in the cMPs is functional. In HCVR-patients, overall TF+-cMPs (monocyte-derived [CD142+/CD14+] and platelet-derived [CD142+/TSP1+]) and activated pMPs directly correlate with MRI-detected lipid-rich atherosclerotic plaques while inversely correlate with MRI-detected calcified plaques. C-statistics analysis showed that prothrombotic cMPs add significant prognostic value to a risk factor model for the prediction of lipid-rich plaques. In conclusion, the activation status of blood cells in HCVR-patients differed markedly from controls as shown by higher circulating levels of prothrombotic and TF+-cMPs. Prothrombotic cMP numbers identify subclinical atherosclerotic plaque burden.
Collapse
|
39
|
Lv JX, Kong Q, Ma X. Current advances in circulating inflammatory biomarkers in atherosclerosis and related cardio-cerebrovascular diseases. Chronic Dis Transl Med 2017; 3:207-212. [PMID: 29354803 PMCID: PMC5747494 DOI: 10.1016/j.cdtm.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis (AS) is a systemic chronic disease affecting both the coronary and cerebral arteries. Inflammation plays a key role in the initiation and progression of AS, and numerous inflammatory factors have been proposed as potential biomarkers. This article reviews recent research in studies on major circulating inflammatory biomarkers to identify surrogates that may reflect processes associated with AS development and the risk of AS-related vascular events, such as Von Willebrand factor, lectin-like oxidized low-density-lipoprotein receptor-1, soluble urokinase plasminogen activator receptor, regulated upon activation, normal T-cell expressed and secreted, and microparticles, which may provide new perspectives for clinical AS evaluation and risk stratification.
Collapse
Affiliation(s)
- Jun-Xuan Lv
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Qi Kong
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Xin Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| |
Collapse
|
40
|
Peña E, Arderiu G, Badimon L. Protein disulphide-isomerase A2 regulated intracellular tissue factor mobilisation in migrating human vascular smooth muscle cells. Thromb Haemost 2017; 113:891-902. [DOI: 10.1160/th14-09-0776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/01/2014] [Indexed: 11/05/2022]
Abstract
SummaryProtein-disulphide isomerase family (PDI) are an ER-stress protein that controls TF-procoagulant activity but its role in HVSMC migration and coronary artery disease remains to be elucidated. We aimed to investigate whether in human coronary smooth muscle cells (HVSMC) the ER-stress protein-disulphide isomerase family A member 2 (PDIA2) regulates tissue factor (TF) polarisation during migration and atherosclerotic remodeling. PDIA2 and TF were analysed by confocal microscopy, silenced by small interfering RNAs (siRNA) and their function analysed by transwell and migration assays in vitro and in vivo. PDIA2and TF co-localise in the front edge of motile HVSMC. Silencing PDIA2, as well as silencing TF, reduces migration. PDIA2 silenced cells show increased TF-rich microparticle shedding. In vivo cell-loaded plug implants in nude mice of PDIA2 silenced HVSMC together with microvascular endothelial cells showed a significant impairment in mature microvessel formation. PDIA2 and TF are found in remodelled atherosclerotic plaques but not in healthy coronaries. In conclusion, we demonstrate that TF is chaperoned by PDIA2 to the HVSMC membrane and to the cell migratory front. Absence of PDIA2 impairs TF intracellular trafficking to its membrane docking favoring its uncontrolled release in microparticles. TF-regulated HVSMC migration and microvessel formation is under the control of the ER-protein PDIA2.
Collapse
|
41
|
Escate R, Mata P, Cepeda JM, Padreó T, Badimon L. miR-505-3p controls chemokine receptor up-regulation in macrophages: role in familial hypercholesterolemia. FASEB J 2017; 32:601-612. [PMID: 32172543 DOI: 10.1096/fj.201700476r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/11/2017] [Indexed: 12/16/2022]
Abstract
Familial hypercholesterolemia (FH) conveys a high risk of premature atherosclerosis as a result of lifelong exposure to high LDL cholesterol levels that are not fully reduced by standard-of-care lipid-lowering treatment. Inflammatory mediators have played a role in the progression of atherosclerotic lesions. Here, we investigated whether innate immunity cells in patients with FH have a specific proinflammatory phenotype that is distinct from that of cells in normal participants. To this end, miR-505-3p-a microRNA related to chronic inflammation-and its target genes were investigated in monocyte-derived macrophages (MACs) of patients with FH (FH-MACs) and non-FH controls (co-MACs). On the basis of the profiler PCR array analysis of agomiR-505-3p-transfected MACs, we identified the chemokine receptors, CCR3, CCR4, and CXCR1, as genes that are regulated by miR-505-3p via the transcription factor, RUNX1. miR-505-3p was significantly down-regulated, whereas CCR3, CCR4, CXCR, and RUNX1 were increased in FH-MAC compared with co-MAC, with the increase being more evident in the proinflammatory M1-like FH-MAC. Chemokine receptor levels were unrelated to LDL plasma levels at entry, but correlated with age in patients with FH, not in controls. In summary, we demonstrate for first time to our knowledge that MACs from FH-MACs have an inflammatory phenotype that is characterized by the up-regulation of CCR3, CCR4, and CXCR1 under the control of miR-505-3p. These results suggest a chronic inflammatory condition in FH innate immunity cells that is not reverted by standard lipid-lowering treatment.-Escate, R., Mata, P., Cepeda, J.M., Padró, T., Badimon, L. miR-505-3p controls chemokine receptor up-regulation in macrophages: role in familial hypercholesterolemia. FASEB J. 32, 601-612 (2018). www.fasebj.org.
Collapse
Affiliation(s)
- Rafael Escate
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain
| | - Pedro Mata
- Fundacion Hipercolesterolemia Familiar, Madrid, Spain
| | - Jose Maria Cepeda
- Department of Internal Medicine, Hospital Vega Baja, Orihuela, Spain
| | - Teresa Padreó
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Catalan Institute of Cardiovascular Sciences (ICCC), Sant Pau Biomedical Research Institute (IIB-Sant Pau) Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital de Sant Pau, Barcelona, Spain.,Universitat Autonoma de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Giannella A, Radu CM, Franco L, Campello E, Simioni P, Avogaro A, de Kreutzenberg SV, Ceolotto G. Circulating levels and characterization of microparticles in patients with different degrees of glucose tolerance. Cardiovasc Diabetol 2017; 16:118. [PMID: 28927403 PMCID: PMC5606070 DOI: 10.1186/s12933-017-0600-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/09/2017] [Indexed: 12/12/2022] Open
Abstract
Background Microparticles (MPs) are vesicular structures shed from endothelial or circulating blood cells, after activation or apoptosis, and can be considered markers of vascular damage. We aimed to determine the levels of circulating MPs, their content of miRNA-126-3p and 5p, and their relationship with early endothelial activation/damage, in patients with different degree of glucose tolerance. Methods CD62E+, CD62P+, CD142+, CD45+ circulating MPs, their apoptotic (AnnexinV+) fractions, and miRNA-126 expression were determined in 39 prediabetic (PreDM), 68 type 2 diabetic (T2DM), and 53 control (NGT) subjects, along with main anthropometric and biochemical measurements. MPs were analysed by flow cytometry. miRNA-126 was measured by quantitative real-time PCR. Plasma antioxidant capacity was determined by electronic spin resonance; ICAM-1, and VCAM-1 by ELISA. Results Activated endothelial cell-derived MPs (CD62E+) were significantly increased in PreDM and T2DM in comparison to NGT (p < 0.0001). AnnexinV+/CD62E+ MPs and Annexin V+ MPs were significantly increased in T2DM compared to PreDM and NGT (p < 0.001); other MPs were not significantly different among groups. Plasma antioxidant capacity was significantly decreased in PreDM and T2DM compared to NGT (p = 0.001); VCAM-1 significantly increased in PreDM and T2DM in comparison to NGT (p = 0.001). miR-126-3p expression, but not miR-126-5p, in MPs, decreased significantly and progressively from NGT, to PreDM, and T2DM (p < 0.001). In PreDM and T2DM, CD62E+ MPs level was significantly and negatively associated with plasma glucose (p = 0.004). Conclusion We show for the first time that circulating CD62E+ MPs level and miR-126-3p content in MPs are abnormal in subjects with pre-diabetes; the content of miR-126-3p correlates with markers of endothelial inflammation, such as VCAM-1, plasma antioxidant capacity, and microparticles, well-accepted markers of endothelial dysfunction.
Collapse
Affiliation(s)
| | | | - Lorenzo Franco
- Department of Chemical Sciences, University of Padova, Padua, Italy
| | - Elena Campello
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Paolo Simioni
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Angelo Avogaro
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| | | | - Giulio Ceolotto
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| |
Collapse
|
43
|
Ganjali S, Momtazi AA, Banach M, Kovanen PT, Stein EA, Sahebkar A. HDL abnormalities in familial hypercholesterolemia: Focus on biological functions. Prog Lipid Res 2017; 67:16-26. [DOI: 10.1016/j.plipres.2017.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
|
44
|
Ridger VC, Boulanger CM, Angelillo-Scherrer A, Badimon L, Blanc-Brude O, Bochaton-Piallat ML, Boilard E, Buzas EI, Caporali A, Dignat-George F, Evans PC, Lacroix R, Lutgens E, Ketelhuth DFJ, Nieuwland R, Toti F, Tunon J, Weber C, Hoefer IE. Microvesicles in vascular homeostasis and diseases. Position Paper of the European Society of Cardiology (ESC) Working Group on Atherosclerosis and Vascular Biology. Thromb Haemost 2017; 117:1296-1316. [PMID: 28569921 DOI: 10.1160/th16-12-0943] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/27/2017] [Indexed: 12/15/2022]
Abstract
Microvesicles are members of the family of extracellular vesicles shed from the plasma membrane of activated or apoptotic cells. Microvesicles were initially characterised by their pro-coagulant activity and described as "microparticles". There is mounting evidence revealing a role for microvesicles in intercellular communication, with particular relevance to hemostasis and vascular biology. Coupled with this, the potential of microvesicles as meaningful biomarkers is under intense investigation. This Position Paper will summarise the current knowledge on the mechanisms of formation and composition of microvesicles of endothelial, platelet, red blood cell and leukocyte origin. This paper will also review and discuss the different methods used for their analysis and quantification, will underline the potential biological roles of these vesicles with respect to vascular homeostasis and thrombosis and define important themes for future research.
Collapse
Affiliation(s)
| | - Chantal M Boulanger
- Victoria Ridger, PhD, Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK, E-mail: , or, Chantal M. Boulanger, PhD, INSERM UMR-S 970, Paris Cardiovascular Research Center - PARCC, 56 rue Leblanc, 75015 Paris, France, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Koganti S, Eleftheriou D, Brogan PA, Kotecha T, Hong Y, Rakhit RD. Microparticles and their role in coronary artery disease. Int J Cardiol 2017; 230:339-345. [DOI: 10.1016/j.ijcard.2016.12.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/04/2016] [Accepted: 12/17/2016] [Indexed: 12/16/2022]
|
46
|
Neutrophil-derived microparticles are released into the coronary circulation following percutaneous coronary intervention in acute coronary syndrome patients. Biosci Rep 2017; 37:BSR20160430. [PMID: 27913753 PMCID: PMC5240586 DOI: 10.1042/bsr20160430] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/20/2016] [Accepted: 12/01/2016] [Indexed: 11/17/2022] Open
Abstract
To evaluate (i) local coronary and systemic levels of microparticles (MP) in acute coronary syndrome (ACS) and stable angina pectoris (SAP) patients and (ii) their release after plaque disruption with percutaneous coronary intervention (PCI). MP are small vesicles originating from plasma membranes of cells after activation or apoptosis and are implicated in the pathogenesis of atherosclerosis. Neutrophils play a role in plaque destabilization and shed neutrophil-derived MP that have the potential to drive significant proinflammatory and thrombotic downstream effects. Eight ACS and eight SAP patients were included. Coronary sinus (CS) samples pre-intervention (CS1), 45 s following balloon angioplasty (CS2) and at 45 s intervals following stent deployment (CS3, CS4 and CS5), together with peripheral vein samples, pre- and post-PCI were analysed for neutrophil-derived (CD66b+), endothelial-derived (CD144+), platelet-derived (CD41a+), monocyte-derived (CD14+) and apoptotic (Annexin V+) MP. ELISA for interleukin (IL)-6, myeloperoxidase (MPO) and P-selectin was also performed. CD66b+ MP levels were similar in both groups pre-intervention. Post-PCI, CS levels rose significantly in ACS but not SAP patients (ACS area under the curve (AUC): 549 ± 83, SAP AUC: 24 ± 29, P<0.01). CS CD41a+, CD144+, CD14+ and Annexin V+ MP levels did not differ between groups. Acute neutrophil-derived MP release post-PCI occurs in ACS compared with stable patients, likely to be reflective of plaque MP content in vulnerable lesions.
Collapse
|
47
|
Escate R, Padro T, Borrell-Pages M, Suades R, Aledo R, Mata P, Badimon L. Macrophages of genetically characterized familial hypercholesterolaemia patients show up-regulation of LDL-receptor-related proteins. J Cell Mol Med 2016; 21:487-499. [PMID: 27680891 PMCID: PMC5323824 DOI: 10.1111/jcmm.12993] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022] Open
Abstract
Familial hypercholesterolaemia (FH) is a major risk for premature coronary heart disease due to severe long‐life exposure to high LDL levels. Accumulation of LDL in the vascular wall triggers atherosclerosis with activation of the innate immunity system. Here, we have investigated (i) gene expression of LDLR and LRPs in peripheral blood cells (PBLs) and in differentiated macrophages of young FH‐patients; and (ii) whether macrophage from FH patients have a differential response when exposed to high levels of atherogenic LDL. PBLs in young heterozygous genetically characterized FH patients have higher expression of LRP5 and LRP6 than age‐matched healthy controls or patients with secondary hypercholesterolaemia. LRP1 levels were similar among groups. In monocyte‐derived macrophages (MACs), LRP5 and LRP1 transcript levels did not differ between FHs and controls in resting conditions, but when exposed to agLDL, FH‐MAC showed a highly significant up‐regulation of LRP5, while LRP1 was unaffected. PBL and MAC cells from FH patients had significantly lower LDLR expression than control cells, independently of the lipid‐lowering therapy. Furthermore, exposure of FH‐MAC to agLDL resulted in a reduced expression of CD163, scavenger receptor with anti‐inflammatory and atheroprotective properties. In summary, our results show for first time that LRPs, active lipid‐internalizing receptors, are up‐regulated in innate immunity cells of young FH patients that have functional LDLR mutations. Additionally, their reduced CD163 expression indicates less atheroprotection. Both mechanisms may play a synergic effect on the onset of premature atherosclerosis in FH patients.
Collapse
Affiliation(s)
- Rafael Escate
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Rosa Suades
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Rosa Aledo
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Lina Badimon
- Cardiovascular Research Center (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
48
|
Badimon L, Suades R, Fuentes E, Palomo I, Padró T. Role of Platelet-Derived Microvesicles As Crosstalk Mediators in Atherothrombosis and Future Pharmacology Targets: A Link between Inflammation, Atherosclerosis, and Thrombosis. Front Pharmacol 2016; 7:293. [PMID: 27630570 PMCID: PMC5005978 DOI: 10.3389/fphar.2016.00293] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022] Open
Abstract
Reports in the last decade have suggested that the role of platelets in atherosclerosis and its thrombotic complications may be mediated, in part, by local secretion of platelet-derived microvesicles (pMVs), small cell blebs released during the platelet activation process. MVs are the most abundant cell-derived microvesicle subtype in the circulation. High concentrations of circulating MVs have been reported in patients with atherosclerosis, acute vascular syndromes, and/or diabetes mellitus, suggesting a potential correlation between the quantity of microvesicles and the clinical severity of the atherosclerotic disease. pMVs are considered to be biomarkers of disease but new information indicates that pMVs are also involved in signaling functions. pMVs evoke or promote haemostatic and inflammatory responses, neovascularization, cell survival, and apoptosis, processes involved in the pathophysiology of cardiovascular disease. This review is focused on the complex cross-talk between platelet-derived microvesicles, inflammatory cells and vascular elements and their relevance in the development of the atherosclerotic disease and its clinical outcomes, providing an updated state-of-the art of pMV involvement in atherothrombosis and pMV potential use as therapeutic agent influencing cardiovascular biomedicine in the future.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant PauBarcelona, Spain; Cardiovascular Research Chair, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Teresa Padró
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| |
Collapse
|
49
|
Chiva-Blanch G, Suades R, Padró T, Vilahur G, Peña E, Ybarra J, Pou JM, Badimon L. El ácido acetilsalicílico reduce la liberación de micropartículas eritrocitarias, monocitarias y de células del músculo liso vascular en pacientes diabéticos. Rev Esp Cardiol 2016. [DOI: 10.1016/j.recesp.2015.12.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Endothelial microparticles: Pathogenic or passive players in endothelial dysfunction in autoimmune rheumatic diseases? Vascul Pharmacol 2016; 86:71-76. [PMID: 27291140 DOI: 10.1016/j.vph.2016.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/10/2016] [Accepted: 05/21/2016] [Indexed: 11/24/2022]
Abstract
Autoimmune rheumatic diseases are characterised by systemic inflammation and complex immunopathology, with an increased risk of cardiovascular disease, initiated by endothelial dysfunction in a chronic inflammatory environment. Endothelial microparticles (EMPs) are released into the circulation from activated endothelial cells and may therefore, reflect disease severity, vascular and endothelial dysfunction, that could influence disease pathogenesis via autocrine/paracrine signalling. The exact function of EMPs in rheumatic disease remains unknown, and this has initiated research to elucidate EMP composition and function, which may be determined by the mode of endothelial activation and the micro environment. To date, EMPs are thought to play a role in angiogenesis, thrombosis and inflammation by transferring specific proteins and microRNAs (miRs) to target cells. Here, we review the mechanisms underlying the generation and composition of EMPs and the clinical and experimental studies describing the involvement of EMPs in rheumatic diseases, since we have previously shown endothelial dysfunction and an elevated risk of cardiovascular disease are characteristics in systemic lupus erythematosus. We will also discuss the potential of EMPs as future biomarkers of cardiovascular risk in these diseases.
Collapse
|