1
|
Baker CJ, Min D, Marsh-Wakefield F, Siwan E, Gerofi J, Wang X, Hocking SL, Colagiuri S, Johnson NA, Twigg SM. Circulating CD31 + Angiogenic T cells are reduced in prediabetes and increase with exercise training. J Diabetes Complications 2024; 38:108868. [PMID: 39299028 DOI: 10.1016/j.jdiacomp.2024.108868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
AIMS To investigate circulating angiogenic cells in adults with prediabetes and the effect of a structured exercise program. METHODS A cohort of adults with overweight/obesity and either normal glucose (NG) or prediabetes were randomised to receive exercise (Exercise) (as twice weekly supervised combined high intensity aerobic exercise and progressive resistance training, and once weekly home-based aerobic exercise) or an unsupervised stretching intervention (Control) for 12 weeks. Circulating angiogenic T cells, muscle strength, and cardiovascular disease risk factors, including blood lipids, arterial stiffness, central haemodynamic responses, and cardiorespiratory fitness (VO2peak) in those with prediabetes (n = 35, 16 Control, 19 Exercise) and NG (n = 37, 17 Control, 20 Exercise) were analysed at baseline and after the 12-week intervention. RESULTS At baseline, compared with NG those with prediabetes demonstrated reduced VO2peak, angiogenic CD31+CD8+ T cells and VEGFR2+CD4+ T cells, and increased systolic blood pressure. CD31+ T cells were negatively correlated with cardiovascular disease (CVD) risk. Compared with Control, exercise training increased muscle strength, VO2peak, and CD31+CD4+ and CD31+CD8+ T cells in NG and prediabetes. CONCLUSIONS Circulating angiogenic CD31+ T cells are decreased in people with prediabetes and are enhanced with exercise training. Exercise increases CD31+ T cells, and through this mechanism it is proposed that it may reduce CVD risk. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry number: ACTRN12617000552381.
Collapse
Affiliation(s)
- Callum J Baker
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia
| | - Danqing Min
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Felix Marsh-Wakefield
- Liver Injury and Cancer Program, Centenary Institute, Sydney, NSW, Australia; Human Cancer and Viral Immunology Laboratory, The University of Sydney, Sydney, NSW, Australia
| | - Elisha Siwan
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia
| | - James Gerofi
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia
| | - Xiaoyu Wang
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia
| | - Samantha L Hocking
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia; Boden Initiative, Charles Perkins Centre, University of Sydney, NSW, Australia
| | - Stephen Colagiuri
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Boden Initiative, Charles Perkins Centre, University of Sydney, NSW, Australia
| | - Nathan A Johnson
- Boden Initiative, Charles Perkins Centre, University of Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Stephen M Twigg
- Greg Brown Diabetes & Endocrinology Research Laboratory, Charles Perkins Centre, University of Sydney, Sydney, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
2
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
3
|
Gregory CD, Rimmer MP. Extracellular vesicles arising from apoptosis: forms, functions, and applications. J Pathol 2023; 260:592-608. [PMID: 37294158 PMCID: PMC10952477 DOI: 10.1002/path.6138] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 06/10/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed subcellular bodies produced by most, if not all cells. Research over the last two decades has recognised the importance of EVs in intercellular communication and horizontal transfer of biological material. EVs range in diameter from tens of nanometres up to several micrometres and are able to transfer a spectrum of biologically active cargoes - from whole organelles, through macromolecules including nucleic acids and proteins, to metabolites and small molecules - from their cells of origin to recipient cells, which may consequently become physiologically or pathologically altered. Based on their modes of biogenesis, the most renowned EV classes are (1) microvesicles, (2) exosomes (both produced by healthy cells), and (3) EVs from cells undergoing regulated death by apoptosis (ApoEVs). Microvesicles bud directly from the plasma membrane, while exosomes are derived from endosomal compartments. Current knowledge of the formation and functional properties of ApoEVs lags behind that of microvesicles and exosomes, but burgeoning evidence indicates that ApoEVs carry manifold cargoes, including mitochondria, ribosomes, DNA, RNAs, and proteins, and perform diverse functions in health and disease. Here we review this evidence, which demonstrates substantial diversity in the luminal and surface membrane cargoes of ApoEVs, permitted by their very broad size range (from around 50 nm to >5 μm; the larger often termed apoptotic bodies), strongly suggests their origins through both microvesicle- and exosome-like biogenesis pathways, and indicates routes through which they interact with recipient cells. We discuss the capacity of ApoEVs to recycle cargoes and modulate inflammatory, immunological, and cell fate programmes in normal physiology and in pathological scenarios such as cancer and atherosclerosis. Finally, we provide a perspective on clinical applications of ApoEVs in diagnostics and therapeutics. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Christopher D Gregory
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, University of EdinburghEdinburghUK
| | - Michael P Rimmer
- Centre for Reproductive HealthInstitute for Regeneration and Repair, University of EdinburghEdinburghUK
| |
Collapse
|
4
|
Gonçalves TAF, Lima VS, de Almeida AJPO, de Arruda AV, Veras ACMF, Lima TT, Soares EMC, Santos ACD, Vasconcelos MECD, de Almeida Feitosa MS, Veras RC, de Medeiros IA. Carvacrol Improves Vascular Function in Hypertensive Animals by Modulating Endothelial Progenitor Cells. Nutrients 2023; 15:3032. [PMID: 37447358 DOI: 10.3390/nu15133032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Carvacrol, a phenolic monoterpene, has diverse biological activities, highlighting its antioxidant and antihypertensive capacity. However, there is little evidence demonstrating its influence on vascular regeneration. Therefore, we evaluated the modulation of carvacrol on endothelial repair induced by endothelial progenitor cells (EPC) in hypertension. Twelve-week-old spontaneously hypertensive rats (SHR) were treated with a vehicle, carvacrol (50 or 100 mg/kg/day), or resveratrol (10 mg/kg/day) orally for four weeks. Wistar Kyoto (WKY) rats were used as the normotensive controls. Their systolic blood pressure (SBP) was measured weekly through the tail cuff. The EPCs were isolated from the bone marrow and peripherical circulation and were quantified by flow cytometry. The functionality of the EPC was evaluated after cultivation through the quantification of colony-forming units (CFU), evaluation of eNOS, intracellular detection of reactive oxygen species (ROS), and evaluation of senescence. The superior mesenteric artery was isolated to evaluate the quantification of ROS, CD34, and CD31. Treatment with carvacrol induced EPC migration, increased CFU formation and eNOS expression and activity, and reduced ROS and senescence. In addition, carvacrol reduced vascular ROS and increased CD31 and CD34 expression. This study showed that treatment with carvacrol improved the functionality of EPC, contributing to the reduction of endothelial dysfunction.
Collapse
Affiliation(s)
| | - Viviane Silva Lima
- Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa 58059-900, PB, Brazil
| | | | - Alinne Villar de Arruda
- Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa 58059-900, PB, Brazil
| | | | - Thaís Trajano Lima
- Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa 58059-900, PB, Brazil
| | | | | | | | | | - Robson Cavalcante Veras
- Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa 58059-900, PB, Brazil
| | - Isac Almeida de Medeiros
- Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa 58059-900, PB, Brazil
| |
Collapse
|
5
|
McIntosh R, Hidalgo M, Lobo J, Dillon K, Szeto A, Hurwitz BE. Circulating endothelial and angiogenic cells predict hippocampal volume as a function of HIV status. J Neurovirol 2023; 29:65-77. [PMID: 36418739 DOI: 10.1007/s13365-022-01101-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/27/2022]
Abstract
Circulating endothelial cells (CECs) and myeloid angiogenic cells (MACs) have the capacity to stabilize human blood vessels in vivo. Evidence suggests that these cells are depleted in dementia and in persons living with HIV (PWH), who have a higher prevalence of dementia and other cognitive deficits associated with aging. However, the associations of CECs and MACs with MRI-based measures of aging brain health, such as hippocampal gray matter volume, have not been previously demonstrated. The present study examined differences in these associations in 51 postmenopausal women with and without HIV infection. Gray matter volume was quantified using MRI. CECs and MACs were enumerated using fluorescence-activated cell sorting. Analyses examined the association of these cell counts with left and right hippocampal gray matter volume while controlling for age and hypertension status. The main finding was an interaction suggesting that compared to controls, postmenopausal PWH with greater levels of CECs and MACs had significantly greater hippocampus GMV. Further research is necessary to examine potential underlying pathophysiological mechanisms in HIV infection linking morpho-functional circulatory reparative processes with more diminished hippocampal volume in postmenopausal women.
Collapse
Affiliation(s)
- Roger McIntosh
- Department of Psychology, College of Arts and Sciences, University of Miami, Miami, FL, USA.
- Behavioral Medicine Research Center, University of Miami, Miami, FL, USA.
- Division of Public Health Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Melissa Hidalgo
- Department of Internal Medicine, Broward Health North, Fort Lauderdale, FL, USA
| | - Judith Lobo
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Kaitlyn Dillon
- Department of Psychology, College of Arts and Sciences, University of Miami, Miami, FL, USA
| | - Angela Szeto
- Department of Psychology, College of Arts and Sciences, University of Miami, Miami, FL, USA
| | - Barry E Hurwitz
- Department of Psychology, College of Arts and Sciences, University of Miami, Miami, FL, USA
- Behavioral Medicine Research Center, University of Miami, Miami, FL, USA
- Division of Endocrinology, Diabetes and Metabolism, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
6
|
Salomon C, Das S, Erdbrügger U, Kalluri R, Kiang Lim S, Olefsky JM, Rice GE, Sahoo S, Andy Tao W, Vader P, Wang Q, Weaver AM. Extracellular Vesicles and Their Emerging Roles as Cellular Messengers in Endocrinology: An Endocrine Society Scientific Statement. Endocr Rev 2022; 43:441-468. [PMID: 35552682 PMCID: PMC10686249 DOI: 10.1210/endrev/bnac009] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 12/15/2022]
Abstract
During the last decade, there has been great interest in elucidating the biological role of extracellular vesicles (EVs), particularly, their hormone-like role in cell-to-cell communication. The field of endocrinology is uniquely placed to provide insight into the functions of EVs, which are secreted from all cells into biological fluids and carry endocrine signals to engage in paracellular and distal interactions. EVs are a heterogeneous population of membrane-bound vesicles of varying size, content, and bioactivity. EVs are specifically packaged with signaling molecules, including lipids, proteins, and nucleic acids, and are released via exocytosis into biofluid compartments. EVs regulate the activity of both proximal and distal target cells, including translational activity, metabolism, growth, and development. As such, EVs signaling represents an integral pathway mediating intercellular communication. Moreover, as the content of EVs is cell-type specific, it is a "fingerprint" of the releasing cell and its metabolic status. Recently, changes in the profile of EV and bioactivity have been described in several endocrine-related conditions including diabetes, obesity, cardiovascular diseases, and cancer. The goal of this statement is to highlight relevant aspects of EV research and their potential role in the field of endocrinology.
Collapse
Affiliation(s)
- Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Saumya Das
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Uta Erdbrügger
- Department of Medicine, Nephrology Division, University of Virginia, Charlottesville, VA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Jerrold M Olefsky
- Department of Medicine, University of California-San Diego, La Jolla, CA, USA
| | | | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Pieter Vader
- CDL Research, Division LAB, UMC Utrecht, Utrecht, the Netherlands Faculty of Medicine, Utrecht University, Utrecht, the Netherlands; Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, The Netherlands
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
7
|
Stampouloglou PK, Siasos G, Bletsa E, Oikonomou E, Vogiatzi G, Kalogeras K, Katsianos E, Vavuranakis MA, Souvaliotis N, Vavuranakis M. The Role of Cell Derived Microparticles in Cardiovascular Diseases: Current Concepts. Curr Pharm Des 2022; 28:1745-1757. [DOI: 10.2174/1381612828666220429081555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 12/07/2022]
Abstract
Abstract:
Cardiovascular disease remains the main cause of human morbidity and mortality in the developed countries. Microparticles (MPs) are small vesicles originating from the cell membrane as a result of various stimuli and particularly of biological processes that constitute the pathophysiology of atherosclerosis, such as endothelial damage. They form vesicles that can transfer various molecules and signals to remote target cells without direct cell to cell interaction. Circulating microparticles have been associated with cardiovascular diseases. Therefore, many studies have been designed to further investigate the role of microparticles as biomarkers for diagnosis, prognosis, and disease monitoring. To this concept the pro-thrombotic and atherogenic potential of platelets and endothelial derived MPs has gain research interest especially concerning accelerate atherosclerosis and acute coronary syndrome triggering and prognosis. MPs especially of endothelial origin have been investigated in different clinical scenarios of heart failure and in association of left ventricular loading conditions. Finally, most cardiovascular risk factors present unique patterns of circulating MPs population, highlighting their pathophysiologic link to cardiovascular disease progression. In this review article we present a synopsis of the biogenesis and characteristics of microparticles, as well as the most recent data concerning their implication in the cardiovascular settings.
Collapse
Affiliation(s)
- Panagiota K. Stampouloglou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Evanthia Bletsa
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Georgia Vogiatzi
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Efstratios Katsianos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Nektarios Souvaliotis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| |
Collapse
|
8
|
Mas-Bargues C, Alique M, Barrús-Ortiz MT, Borrás C, Rodrigues-Díez R. Exploring New Kingdoms: The Role of Extracellular Vesicles in Oxi-Inflamm-Aging Related to Cardiorenal Syndrome. Antioxidants (Basel) 2021; 11:78. [PMID: 35052582 PMCID: PMC8773353 DOI: 10.3390/antiox11010078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of age associated chronic diseases has increased in recent years. Although several diverse causes produce these phenomena, abundant evidence shows that oxidative stress plays a central role. In recent years, numerous studies have focused on elucidating the role of oxidative stress in the development and progression of both aging and chronic diseases, opening the door to the discovery of new underlying mechanisms and signaling pathways. Among them, senolytics and senomorphics, and extracellular vesicles offer new therapeutic strategies to slow the development of aging and its associated chronic diseases by decreasing oxidative stress. In this review, we aim to discuss the role of extracellular vesicles in human cardiorenal syndrome development and their possible role as biomarkers, targets, or vehicles of drugs to treat this syndrome.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Madrid, Spain;
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - María Teresa Barrús-Ortiz
- Área de Fisiología, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Univesidad Rey Juan Carlos, Avenida de Atenas s/n, 28922 Madrid, Spain
| | - Consuelo Borrás
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Raquel Rodrigues-Díez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Hospital La Paz (IdiPAZ), 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 08036 Barcelona, Spain
| |
Collapse
|
9
|
Lia G, Giaccone L, Leone S, Bruno B. Biomarkers for Early Complications of Endothelial Origin After Allogeneic Hematopoietic Stem Cell Transplantation: Do They Have a Potential Clinical Role? Front Immunol 2021; 12:641427. [PMID: 34093530 PMCID: PMC8170404 DOI: 10.3389/fimmu.2021.641427] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/04/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial cell (EC) dysfunction causes a number of early and life-threatening post hematopoietic stem cell transplant (HCT) complications that result in a rapid clinical decline. The main early complications are graft-vs.-host disease (GVHD), transplant associated thrombotic microangiopathy (TA-TMA), and sinusoidal obstruction syndrome (SOS). Post-HCT endothelial dysfunction occurs as a result of chemotherapy, infections, and allogeneic reactivity. Despite major advances in transplant immunology and improvements in supportive care medicine, these complications represent a major obstacle for successful HCT. In recent years, different biomarkers have been investigated for early detection of post-transplant endothelial cell dysfunction, but few have been validated. In this review we will define GVHD, TA-TMA and SOS, summarize the current data available in HCT biomarker research and identify promising biomarkers for detection and diagnosis of early HCT complications.
Collapse
Affiliation(s)
- Giuseppe Lia
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luisa Giaccone
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Sarah Leone
- Department of Internal Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States
| |
Collapse
|
10
|
Liu ZZ, Jose PA, Yang J, Zeng C. Importance of extracellular vesicles in hypertension. Exp Biol Med (Maywood) 2021; 246:342-353. [PMID: 33517775 DOI: 10.1177/1535370220974600] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hypertension affects approximately 1.13 billion adults worldwide and is the leading global risk factor for cardiovascular, cerebrovascular, and kidney diseases. There is emerging evidence that extracellular vesicles participate in the development and progression of hypertension. Extracellular vesicles are membrane-enclosed structures released from nearly all types of eukaryotic cells. During their formation, extracellular vesicles incorporate various parent cell components, including proteins, lipids, and nucleic acids that can be transferred to recipient cells. Extracellular vesicles mediate cell-to-cell communication in a variety of physiological and pathophysiological processes. Therefore, studying the role of circulating and urinary extracellular vesicles in hypertension has the potential to identify novel noninvasive biomarkers and therapeutic targets of different hypertension phenotypes. This review discusses the classification and biogenesis of three EV subcategories (exosomes, microvesicles, and apoptotic bodies) and provides a summary of recent discoveries in the potential impact of extracellular vesicles on hypertension with a specific focus on their role in the blood pressure regulation by organs-artery and kidney, as well as renin-angiotensin-system.
Collapse
Affiliation(s)
- Zhi Z Liu
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400714, P.R. China.,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, P. R. China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, P.R. China
| | - Chunyu Zeng
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400714, P.R. China.,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, P. R. China.,Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
11
|
Uil M, Hau CM, Ahdi M, Mills JD, Kers J, Saleem MA, Florquin S, Gerdes VEA, Nieuwland R, Roelofs JJTH. Cellular origin and microRNA profiles of circulating extracellular vesicles in different stages of diabetic nephropathy. Clin Kidney J 2021; 14:358-365. [PMID: 33564439 PMCID: PMC7857783 DOI: 10.1093/ckj/sfz145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes and the main cause of end-stage renal disease. Extracellular vesicles (EVs) are small cell-derived vesicles that can alter disease progression by microRNA (miRNA) transfer. METHODS In this study, we aimed to characterize the cellular origin and miRNA content of EVs in plasma samples of type 2 diabetes patients at various stages of DN. Type 2 diabetes patients were classified in three groups: normoalbuminuria, microalbuminuria and macroalbuminuria. The concentration and cellular origin of plasma EVs were measured by flow cytometry. A total of 752 EV miRNAs were profiled in 18 subjects and differentially expressed miRNAs were validated. RESULTS Diabetic patients with microalbuminuria and/or macroalbuminuria showed elevated concentrations of total EVs and EVs from endothelial cells, platelets, leucocytes and erythrocytes compared with diabetic controls. miR-99a-5p was upregulated in macroalbuminuric patients compared with normoalbuminuric and microalbuminuric patients. Transfection of miR-99a-5p in cultured human podocytes downregulated mammalian target of rapamycin (mTOR) protein expression and downregulated the podocyte injury marker vimentin. CONCLUSIONS Type 2 diabetes patients with microalbuminuria and macroalbuminuria display differential EV profiles. miR-99a-5p expression is elevated in EVs from macroalbuminuria and mTOR is its validated mRNA target.
Collapse
Affiliation(s)
- Melissa Uil
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chi M Hau
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mohamed Ahdi
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Van’t Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, The Netherlands
| | - Moin A Saleem
- Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, UK
| | - Sandrine Florquin
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Victor E A Gerdes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Sesn2 attenuates the damage of endothelial progenitor cells induced by angiotensin II through regulating the Keap1/Nrf2 signal pathway. Aging (Albany NY) 2020; 12:25505-25527. [PMID: 33231566 PMCID: PMC7803511 DOI: 10.18632/aging.104156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023]
Abstract
Endothelial progenitor cell (EPC) dysfunction is an important physiopathological mechanism in the dynamics of the formation of atherosclerosis. It has been reported that angiotensin II (Ang-II) damages the function of EPCs in atherosclerotic plaque through induction of oxidative stress. Sestrin 2 (Sesn2) serves as an antioxidant role in oxidative stress, however, the exact mechanisms underlying the dynamics of how Sesn2 may factor into EPCs after Ang-II treatments needs to be illustrated. We isolated EPCs from human umbilical cord blood samples and treated with Ang-II. Western blotting, qRT-PCR, transwell assays, immunofluorescence and so on were used to investigate the mechanisms underlying the roles of Sesn2 in EPCs treated with Ang-II. Ang-II was found to promote the apoptosis of EPCs as well as inhibited the mRNA and protein expression of Sesn2. Upregulation of Sesn2 attenuated the negative effect of Ang-II. Sesn2 increased the protein expression of Nrf2 by enhancing P62-dependent autophagy. Silencing of Nrf2 enhanced the degree of apoptosis of EPCs as well as resulted in the impairment of EPC functions through inducing the promotion of (reactive oxygen species) ROS production. Our study results indicated that Sesn2 facilitated the viability of EPCs After treatment with Ang-II, as well as provided a potential therapeutic target to alleviate the progression of atherosclerosis.
Collapse
|
13
|
Martinez-Arroyo O, Ortega A, Redon J, Cortes R. Therapeutic Potential of Extracellular Vesicles in Hypertension-Associated Kidney Disease. Hypertension 2020; 77:28-38. [PMID: 33222549 DOI: 10.1161/hypertensionaha.120.16064] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension-mediated organ damage frequently includes renal function decline in which several mechanisms are involved. The present review outlines the state of the art on extracellular vesicles in hypertension and hypertension-related renal damage. Emerging evidence indicates that extracellular vesicles, small vesicles secreted by most cell types and body fluids, are involved in cell-to-cell communication and are key players mediating biological processes such as inflammation, endothelial dysfunction or fibrosis, mechanisms present the onset and progression of hypertension-associated kidney disease. We address the potential use of extracellular vesicles as markers of hypertension-mediated kidney damage severity and their application as therapeutic agents in hypertension-associated renal damage. The capacity of exosomes to deliver a wide variety of cargos to the target cell efficiently makes them a potential drug delivery system for treatment of renal diseases.
Collapse
Affiliation(s)
- Olga Martinez-Arroyo
- From the Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Valencia, Spain (O.M.-A., A.O., J.R., R.C.)
| | - Ana Ortega
- From the Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Valencia, Spain (O.M.-A., A.O., J.R., R.C.)
| | - Josep Redon
- From the Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Valencia, Spain (O.M.-A., A.O., J.R., R.C.).,Internal Medicine, Clinic Universitary Hospital, Valencia, Spain (J.R.).,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Minister of Health, Madrid, Spain (J.R.)
| | - Raquel Cortes
- From the Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Valencia, Spain (O.M.-A., A.O., J.R., R.C.)
| |
Collapse
|
14
|
NOX5-induced uncoupling of endothelial NO synthase is a causal mechanism and theragnostic target of an age-related hypertension endotype. PLoS Biol 2020; 18:e3000885. [PMID: 33170835 PMCID: PMC7654809 DOI: 10.1371/journal.pbio.3000885] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hypertension is the most important cause of death and disability in the elderly. In 9 out of 10 cases, the molecular cause, however, is unknown. One mechanistic hypothesis involves impaired endothelium-dependent vasodilation through reactive oxygen species (ROS) formation. Indeed, ROS forming NADPH oxidase (Nox) genes associate with hypertension, yet target validation has been negative. We re-investigate this association by molecular network analysis and identify NOX5, not present in rodents, as a sole neighbor to human vasodilatory endothelial nitric oxide (NO) signaling. In hypertensive patients, endothelial microparticles indeed contained higher levels of NOX5—but not NOX1, NOX2, or NOX4—with a bimodal distribution correlating with disease severity. Mechanistically, mice expressing human Nox5 in endothelial cells developed—upon aging—severe systolic hypertension and impaired endothelium-dependent vasodilation due to uncoupled NO synthase (NOS). We conclude that NOX5-induced uncoupling of endothelial NOS is a causal mechanism and theragnostic target of an age-related hypertension endotype. Nox5 knock-in (KI) mice represent the first mechanism-based animal model of hypertension. The causes of hypertension are not understood; treatments are symptomatic and prevent only few of the associated risks. This study applies network medicine to identify a subgroup of patients with NADPH oxidase 5-induced uncoupling of nitric oxide synthase as the cause of age-related hypertension, enabling a first-in-class mechanism-based treatment of hypertension.
Collapse
|
15
|
La Salvia S, Gunasekaran PM, Byrd JB, Erdbrügger U. Extracellular Vesicles in Essential Hypertension: Hidden Messengers. Curr Hypertens Rep 2020; 22:76. [PMID: 32880744 DOI: 10.1007/s11906-020-01084-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Hypertension affects about half of all Americans, yet in the vast majority of cases, the factors causing the hypertension cannot be clearly delineated. Developing a more precise understanding of the molecular pathogenesis of HTN and its various phenotypes is therefore a pressing priority. Circulating and urinary extracellular vesicles (EVs) are potential novel candidates as biomarkers and bioactivators in HTN. EVs are a heterogeneous population of small membrane fragments shed from various cell types into various body fluids. As EVs carry protein, RNA, and lipids, they also play a role as effectors and novel cell-to-cell communicators. In this review, we discuss the diagnostic, functional, and regenerative role of EVs in essential HTN and focus on EV protein and RNA cargo as the most extensively studied EV cargo. RECENT FINDINGS The field of EVs in HTN is still a young one and earlier studies have not used the novel EV detection tools currently available. More rigor and transparency in EV research are needed. Current data suggest that EVs represent potential novel biomarkers in HTN. EVs correlate with HTN severity and possibly end-organ damage. However, it has yet to be discerned which specific subtype(s) of EV reflects best HTN pathophysiology. Evolving studies are also showing that EVs might be novel regulators in vascular and renal tubular function and also be therapeutic. RNA in EVs has been studied in the context of hypertension, largely in the form of studies of miRNA, which are reviewed herein. Beyond miRNAs, mRNA in urinary EVs changed in response to sodium loading in humans. EVs represent promising novel biomarkers and bioactivators in essential HTN. Novel tools are being developed to apply more rigor in EV research including more in vivo models and translation to humans.
Collapse
Affiliation(s)
- Sabrina La Salvia
- Department of Internal Medicine, Division of Nephrology, University of Virginia Health System, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908-0133, USA.
| | - Pradeep Moon Gunasekaran
- Department of Internal Medicine, Division of Cardiovascular Medicine, Medical School, University of Michigan Medical School, 5570C MSRB II, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - James Brian Byrd
- Department of Internal Medicine, Division of Cardiovascular Medicine, Medical School, University of Michigan Medical School, 5570C MSRB II, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Uta Erdbrügger
- Department of Internal Medicine, Division of Nephrology, University of Virginia Health System, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908-0133, USA
| |
Collapse
|
16
|
Mir B, Goettsch C. Extracellular Vesicles as Delivery Vehicles of Specific Cellular Cargo. Cells 2020; 9:cells9071601. [PMID: 32630649 PMCID: PMC7407641 DOI: 10.3390/cells9071601] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) mediate cell-to-cell communication via the transfer of biomolecules locally and systemically between organs. It has been elucidated that the specific EV cargo load is fundamental for cellular response upon EV delivery. Therefore, revealing the specific molecular machinery that functionally regulates the precise EV cargo intracellularly is of importance in understanding the role of EVs in physiology and pathophysiology and conveying therapeutic use. The purpose of this review is to summarize recent findings on the general rules, as well as specific modulator motifs governing EV cargo loading. Finally, we address available information on potential therapeutic strategies to alter cargo loading.
Collapse
|
17
|
Highton PJ, White AEM, Nixon DGD, Wilkinson TJ, Neale J, Martin N, Bishop NC, Smith AC. Influence of acute moderate- to high-intensity aerobic exercise on markers of immune function and microparticles in renal transplant recipients. Am J Physiol Renal Physiol 2020; 318:F76-F85. [DOI: 10.1152/ajprenal.00332.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Renal transplant recipients (RTRs) and patients with nondialysis chronic kidney disease display elevated circulating microparticle (MP) counts, while RTRs display immunosuppression-induced infection susceptibility. The impact of aerobic exercise on circulating immune cells and MPs is unknown in RTRs. Fifteen RTRs [age: 52.8 ± 14.5 yr, estimated glomerular filtration rate (eGFR): 51.7 ± 19.8 mL·min−1·1.73 m−2 (mean ± SD)] and 16 patients with nondialysis chronic kidney disease (age: 54.8 ± 16.3 yr, eGFR: 61.9 ± 21.0 mL·min−1·1.73 m−2, acting as a uremic control group), and 16 healthy control participants (age: 52.2 ± 16.2 yr, eGFR: 85.6 ± 6.1 mL·min−1·1.73 m−2) completed 20 min of walking at 60–70% peak O2 consumption. Venous blood samples were taken preexercise, postexercise, and 1 h postexercise. Leukocytes and MPs were assessed using flow cytometry. Exercise increased classical ( P = 0.001) and nonclassical ( P = 0.002) monocyte subset proportions but decreased the intermediate subset ( P < 0.001) in all groups. Exercise also decreased the percentage of platelet-derived MPs that expressed tissue factor in all groups ( P = 0.01), although no other exercise-dependent effects were observed. The exercise-induced reduction in intermediate monocyte percentage suggests an anti-inflammatory effect, although this requires further investigation. The reduction in the percentage of tissue factor-positive platelet-derived MPs suggests reduced prothrombotic potential, although further functional assays are required. Exercise did not cause aberrant immune cell activation, suggesting its safety from an immunological standpoint (ISRCTN38935454).
Collapse
Affiliation(s)
- Patrick J. Highton
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Alice E. M. White
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Daniel G. D. Nixon
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Thomas J. Wilkinson
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Jill Neale
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Naomi Martin
- Leicester School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Nicolette C. Bishop
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Alice C. Smith
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
18
|
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: 0.4] [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
|
19
|
Dec-Gilowska M, Trojnar M, Makaruk B, Czop M, Przybylska-Kuc S, Mosiewicz-Madejska B, Dzida G, Mosiewicz J. Circulating Endothelial Microparticles and Aortic Stiffness in Patients with Type 2 Diabetes Mellitus. ACTA ACUST UNITED AC 2019; 55:medicina55090596. [PMID: 31527473 PMCID: PMC6780956 DOI: 10.3390/medicina55090596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/30/2019] [Accepted: 09/09/2019] [Indexed: 01/14/2023]
Abstract
Diabetes mellitus represents a metabolic disorder the incidence of which has been on the increase in recent years. The well-known long-term complications of this disease encompass a wide spectrum of renal, neurological and cardiovascular conditions. The aim of the study was to investigate the serum concentration of endothelial microparticles (EMPs) as well as selected noninvasive parameters of the ascending aorta stiffness calculated with echocardiography. In this study, 58 patients were enrolled-38 subjects diagnosed with type 2 diabetes mellitus (T2DM) and 20 healthy controls. The analyzed populations did not differ significantly with respect to age, renal function, systolic and diastolic blood pressure. The patients with diabetes and concomitant hypertension presented higher levels of EMPs in comparison with diabetic normotensive subjects. Among patients with diabetes and hypertension, aortic stiffness assessed with the elasticity index (Ep) was higher and the aortic compliance index (D) lower than in the diabetic normotensive group. No correlation between the amount of EMPs and lipid profile, C-reactive protein (CRP) level and glycemia, was observed in the studied group. There was, however, a statistically significant positive correlation between the creatinine level and amount of EMPs, while the negative relationship was documented for EMPs level and the estimated glomerular filtration rate (eGFR).
Collapse
Affiliation(s)
- Malgorzata Dec-Gilowska
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
- Chair and Department of Internal Diseases and Diabetology, Medical University of Warsaw, 02-091 Warsaw, Poland.
| | - Marcin Trojnar
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
| | - Boguslaw Makaruk
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
| | - Marcin Czop
- Department of Clinical Genetics, Medical University of Lublin, 20-059 Lublin, Poland.
| | - Sylwia Przybylska-Kuc
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
| | - Barbara Mosiewicz-Madejska
- Chair and Department of Internal Diseases, Medical University of Lublin, Students Medical Association, 20-059 Lublin, Poland.
| | - Grzegorz Dzida
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
| | - Jerzy Mosiewicz
- Chair and Department of Internal Diseases, Medical University of Lublin, 20-059 Lublin, Poland.
| |
Collapse
|
20
|
Bratseth V, Chiva-Blanch G, Byrkjeland R, Solheim S, Arnesen H, Seljeflot I. Elevated levels of circulating microvesicles in coronary artery disease patients with type 2 diabetes and albuminuria: Effects of exercise training. Diab Vasc Dis Res 2019; 16:431-439. [PMID: 31023084 DOI: 10.1177/1479164119843094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Circulating microvesicles, released from activated/apoptotic cells, are involved in vascular complications and may be looked upon as biomarkers. Albuminuria is characteristic of disease progression in type 2 diabetes mellitus. We aimed to investigate quantitative and qualitative differences of circulating microvesicles in type 2 diabetes mellitus with and without albuminuria and whether 12-month exercise training influenced expression of circulating microvesicles. METHODS Coronary artery disease patients with type 2 diabetes mellitus (n = 75), of which 25 had albuminuria, were included. Annexin V+ (AV+) circulating microvesicles were analysed by flow cytometry in citrated plasma. The exercise volume was 150 min per week. RESULTS In albuminuria patients, circulating microvesicles from endothelial-(CD146+/CD62E+/AV+) and endothelial-progenitor-(CD309+/CD34+/AV+) cells were significantly higher compared to those without (p ⩽ 0.01, both). Receiver operating characteristic curve analysis of the endothelial circulating microvesicles shows an area under the curve of 0.704 (95% confidence interval: 0.57-0.84; p = 0.004). Albuminuria patients had more circulating microvesicles derived from activated leukocytes and monocytes and monocytes carrying tissue factor (CD11b+/AV+, CD11b+/CD14+/AV+, CD142+/CD14+/AV+, respectively, p ⩽ 0.05, all) and higher number of circulating microvesicles from activated platelets (CD62P+/AV+). Within exercising patients, circulating microvesicles from progenitor cells increased (p = 0.023), however, not significantly different from controls. CONCLUSION Coronary artery disease patients with type 2 diabetes mellitus and albuminuria had elevated number of circulating microvesicles from activated blood and vascular cells, rendering them as potential predictors of disease severity. The circulating microvesicles were limitedly affected by long-term exercise training in our population.
Collapse
Affiliation(s)
- Vibeke Bratseth
- 1 Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
- 2 Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gemma Chiva-Blanch
- 3 Cardiovascular Program - ICCC - IR Hospital Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Rune Byrkjeland
- 1 Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Svein Solheim
- 1 Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Harald Arnesen
- 1 Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
- 2 Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- 1 Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
- 2 Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
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: 1.0] [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
|
22
|
Erdbrügger U, Le TH. Extracellular vesicles as a novel diagnostic and research tool for patients with HTN and kidney disease. Am J Physiol Renal Physiol 2019; 317:F641-F647. [PMID: 31313949 DOI: 10.1152/ajprenal.00071.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hypertension (HTN) affects one in three adults in the United States and is a major risk factor for cardiovascular disease and kidney failure. There is emerging evidence that more intense blood pressure lowering reduces mortality in patients with kidney disease who are at risk of cardiovascular disease and progression to end-stage renal disease. However, the ideal blood pressure threshold for patients with kidney disease remains a question of debate. Novel tools to more precisely diagnose HTN, tailor treatment, and predict the risk of end-organ damage such as kidney disease are needed. Analysis of circulating and urinary extracellular vesicles (EVs) and their cargo (protein and RNA) has the potential to identify novel noninvasive biomarkers that can also reflect a specific pathological mechanism of different HTN phenotypes. We will discuss the use of extracellular vesicles as markers of HTN severity and explain their profile change with antihypertensive medicine and potential to detect early end-organ damage. However, more studies with enhanced rigor in this field are needed to define the blood pressure threshold to prevent or delay kidney disease progression and decrease cardiovascular risk.
Collapse
Affiliation(s)
- Uta Erdbrügger
- Division of Nephrology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York
| |
Collapse
|
23
|
Berezin AE, Kremzer AA, Samura TA, Berezina TA. Altered signature of apoptotic endothelial cell-derived microvesicles predicts chronic heart failure phenotypes. Biomark Med 2019; 13:737-750. [PMID: 31157550 DOI: 10.2217/bmm-2018-0449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/02/2019] [Indexed: 02/06/2023] Open
Abstract
Aim: to evaluate the associations between signatures of apoptotic endothelial cell-derived microvesicles (MVs) with phenotypes of chronic heart failure (HF). Methods: The study cohort consisted of 388 prospectively involved subjects with HF patients with predominantly reduced left ventricular ejection fraction (HFrEF), HF with preserved ejection fraction (HFpEF) and HF with mid-range ejection fraction (HFmrEF). All biomarkers were measured at baseline. Results: The number of circulating CD31+/annexin V+ MVs in HFrEF and HFmrEF patients was similar. The number of circulating CD144+/annexin V+ MVs in HFrEF patients was significantly higher than HFmrEF and HFpEF. We determined that a combination of number of circulating CD31+/annexin V+ MVs and Gal-3 was the best predictor of HFpEF and that number of circulating CD144+/annexin V+ MVs is able to increase predictive capabilities of soluble ST2 (sST2) and Gal-3 for HFrEF. Conclusion: We found that the number of circulating CD31+/annexin V+ MVs may improve a predictive capacity for conventional HF biomarkers.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, State Medical University, Zaporozhye, 69035, Ukraine
| | - Alexander A Kremzer
- Clinical Pharmacology Department, State Medical University, Zaporozhye, 69035, Ukraine
| | - Tatyana A Samura
- Clinical Pharmacology Department, State Medical University, Zaporozhye, 69035, Ukraine
| | | |
Collapse
|
24
|
Abstract
Microparticles are a distinctive group of small vesicles, without nucleus, which are involved as significant modulators in several physiological and pathophysiological mechanisms. Plasma microparticles from various cellular lines have been subject of research. Data suggest that they are key players in development and manifestation of cardiovascular diseases and their presence, in high levels, is associated with chronic inflammation, endothelial damage and thrombosis. The strong correlation of microparticle levels with several outcomes in cardiovascular diseases has led to their utilization as biomarkers. Despite the limited clinical application at present, their significance emerges, mainly because their detection and enumeration methods are improving. This review article summarizes the evidence derived from research, related with the genesis and the function of microparticles in the presence of various cardiovascular risk factors and conditions. The current data provide a substrate for several theories of how microparticles influence various cellular mechanisms by transferring biological information.
Collapse
Affiliation(s)
- Christos Voukalis
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK
| | - Eduard Shantsila
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK
| | - Gregory Y H Lip
- b Liverpool Centre for Cardiovascular Science , University of Liverpool and Liverpool Heart & Chest Hospital , Liverpool , UK.,c Department of Clinical Medicine, Aalborg Thrombosis Research Unit , Aalborg University , Aalborg , Denmark
| |
Collapse
|
25
|
Barrachina MN, Calderón-Cruz B, Fernandez-Rocca L, García Á. Application of Extracellular Vesicles Proteomics to Cardiovascular Disease: Guidelines, Data Analysis, and Future Perspectives. Proteomics 2019; 19:e1800247. [PMID: 30467982 DOI: 10.1002/pmic.201800247] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/09/2018] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of vesicles composed of a lipid bilayer that carry a large repertoire of molecules including proteins, lipids, and nucleic acids. In this review, some guidelines for plasma-derived EVs isolation, characterization, and proteomic analysis, and the application of the above to cardiovascular disease (CVD) studies are provided. For EVs analysis, blood samples should be collected using a 21-gauge needle, preferably in citrate tubes, and plasma stored for up to 1 year at -80°, using a single freeze-thaw cycle. For proteomic applications, differential centrifugation (including ultracentrifugation steps) is a good option for EVs isolation. EVs characterization is done by transmission electron microscopy, particle enumeration techniques (nanoparticle-tracking analysis, dynamic light scattering), and flow cytometry. Regarding the proteomics strategy, a label-free and gel-free quantitative method is a good choice due to its accuracy and because it minimizes the amount of sample required for clinical applications. Besides the above, main EVs proteomic findings in cardiovascular-related diseases are presented and analyzed in this review, paying especial attention to overlapping results between studies. The latter might offer new insights into the clinical relevance and potential of novel EVs biomarkers identified to date in the context of CVD.
Collapse
Affiliation(s)
- Maria N Barrachina
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade Santiago de Compostela, Santiago de Compostela, 15782, Spain.,Instituto de Investigación, Sanitaria de Santiago (IDIS), Santiago de Compostela, 15706, Spain
| | - Beatriz Calderón-Cruz
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade Santiago de Compostela, Santiago de Compostela, 15782, Spain.,Instituto de Investigación, Sanitaria de Santiago (IDIS), Santiago de Compostela, 15706, Spain
| | - Lucía Fernandez-Rocca
- Clinical Analysis Laboratory, Maciel Hospital, Faculty of Chemistry, University of the Republic, Montevideo, 11000, Uruguay
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade Santiago de Compostela, Santiago de Compostela, 15782, Spain.,Instituto de Investigación, Sanitaria de Santiago (IDIS), Santiago de Compostela, 15706, Spain
| |
Collapse
|
26
|
Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatr Nephrol 2019; 34:11-30. [PMID: 29181712 PMCID: PMC6244861 DOI: 10.1007/s00467-017-3816-z] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles are cell-derived membrane particles ranging from 30 to 5,000 nm in size, including exosomes, microvesicles, and apoptotic bodies. They are released under physiological conditions, but also upon cellular activation, senescence, and apoptosis. They play an important role in intercellular communication. Their release may also maintain cellular integrity by ridding the cell of damaging substances. This review describes the biogenesis, uptake, and detection of extracellular vesicles in addition to the impact that they have on recipient cells, focusing on mechanisms important in the pathophysiology of kidney diseases, such as thrombosis, angiogenesis, tissue regeneration, immune modulation, and inflammation. In kidney diseases, extracellular vesicles may be utilized as biomarkers, as they are detected in both blood and urine. Furthermore, they may contribute to the pathophysiology of renal disease while also having beneficial effects associated with tissue repair. Because of their role in the promotion of thrombosis, inflammation, and immune-mediated disease, they could be the target of drug therapy, whereas their favorable effects could be utilized therapeutically in acute and chronic kidney injury.
Collapse
|
27
|
Liu D, Baqar S, Lincz LL, Ekinci EI. Sodium Intake, Circulating Microvesicles and Cardiovascular Outcomes in Type 2 Diabetes. Curr Diabetes Rev 2019; 15:435-445. [PMID: 30747074 DOI: 10.2174/1573399815666190212120822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/22/2019] [Accepted: 02/08/2019] [Indexed: 11/22/2022]
Abstract
There is ongoing debate surrounding the complex relationship between dietary sodium intake and cardiovascular morbidity and mortality. The existing literature consists largely of observational studies that have demonstrated positive, negative, U-/J-shaped or unclear associations between sodium intake and cardiovascular outcomes. Our group and others have previously demonstrated an inverse relationship between dietary sodium intake and cardiovascular outcomes in people with type 2 diabetes. Increased activity of the renin-angiotensin-aldosterone system and sympathetic nervous system is postulated to contribute to these paradoxical findings through endothelial dysfunction, a precursor to the development of cardiovascular disease. Microvesicles are submicron (0.1 - 1.0μm) vesicles that form during cellular activation, injury or death with endothelial microvesicles being recognized markers of endothelial dysfunction. They are pathologically elevated in a variety of vascular-related conditions including type 2 diabetes. Lower habitual sodium intake in type 2 diabetes has been associated with higher pro-coagulant platelet microvesicles levels but not with endothelial microvesicles. Research utilizing endothelial microvesicles to evaluate the mechanistic relationship between dietary sodium intake and adverse cardiovascular outcomes in type 2 diabetes remains scarce.
Collapse
Affiliation(s)
- Dorothy Liu
- Department of Medicine, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Sara Baqar
- Department of Medicine, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Lisa L Lincz
- Hunter Haematology Research Group, Calvary Mater Newcastle, New South Wales, Australia
| | - Elif I Ekinci
- Department of Medicine, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
28
|
Abbasian N, Herbert KE, Pawluczyk I, Burton JO, Bevington A. Vesicles bearing gifts: the functional importance of micro-RNA transfer in extracellular vesicles in chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1430-F1443. [PMID: 30110570 DOI: 10.1152/ajprenal.00318.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including microparticles (MPs) and exosomes (EXOs), are derived from a wide range of mammalian cells including blood platelets, endothelial cells, and kidney cells and can be detected in body fluids including blood and urine. While EVs are well established as diagnostic markers under pathophysiological and stress conditions, there is also mounting evidence of their functional significance as vehicles for communication between cells mediated by the presence of nucleic acids, especially microRNAs (miRs), encapsulated in the EVs. miRs regulate gene expression, are transported both in MPs and EXOs, and exert profound effects in the kidney. Here we review current understanding of the links between EVs and miRs, discuss the importance of miRs in kidney disease, and shed light on the role of EVs in transferring miRs through the circulation among the renal, vascular, and inflammatory cell populations that are functionally important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester National Institute of Health Research Cardiovascular Biomedical Research Unit , Leicester , United Kingdom
| | - Izabella Pawluczyk
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - James O Burton
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom.,John Walls Renal Unit, University Hospitals of Leicester , Leicester , United Kingdom
| | - Alan Bevington
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
29
|
Jalal D, Renner B, Laskowski J, Stites E, Cooper J, Valente K, You Z, Perrenoud L, Le Quintrec M, Muhamed I, Christians U, Klawitter J, Lindorfer MA, Taylor RP, Holers VM, Thurman JM. Endothelial Microparticles and Systemic Complement Activation in Patients With Chronic Kidney Disease. J Am Heart Assoc 2018; 7:e007818. [PMID: 30006493 PMCID: PMC6064828 DOI: 10.1161/jaha.117.007818] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/28/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Endothelial microparticles are associated with chronic kidney disease (CKD) and complement activation. We hypothesized that the complement pathway is activated in patients with CKD via endothelial microparticles and that complement activation correlates with endothelial dysfunction in CKD. METHODS AND RESULTS We analyzed complement data of 30 healthy subjects, 30 patients with stage III/IV CKD, and 30 renal transplant recipients with stage III/IV CKD, evaluating the potential correlation of complement fragments with brachial artery flow-mediated dilation, Chronic Kidney Disease Epidemiology Collaboration glomerular filtration rate, and urinary albumin/creatinine ratio. Endothelial microparticles were characterized via proteomic analysis and compared between study groups. Complement fragment Ba was significantly increased in CKD and post-kidney transplant CKD. Plasma Ba levels correlated significantly with lower brachial artery flow-mediated dilation, lower Chronic Kidney Disease Epidemiology Collaboration glomerular filtration rate, and higher urinary albumin/creatinine ratio. Factor D levels were significantly higher in the plasma microparticles of patients with CKD versus healthy controls. Plasma microparticles isolated from patients with CKD and containing factor D activated the alternative pathway in vitro. CONCLUSION The alternative complement pathway is activated in CKD and correlates with endothelial dysfunction and markers of CKD. Future studies are needed to evaluate whether endothelial microparticles with increased factor D play a pathologic role in CKD-associated vascular disease. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02230202.
Collapse
Affiliation(s)
- Diana Jalal
- Division of Nephrology, Carver College of Medicine University of Iowa, Iowa City, IA
| | - Brandon Renner
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Jennifer Laskowski
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Erik Stites
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - James Cooper
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Karissa Valente
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Zhiying You
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Loni Perrenoud
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Moglie Le Quintrec
- Department of Nephrology and Renal Transplantation, Lapeyronnie Hospital and INSERM U1183 IRMB, Montpellier, France
| | - Ismaeel Muhamed
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, North Carolina State University and University of North Carolina-Chapel Hill, NC
| | - Uwe Christians
- iC42 Clinical Research and Development, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jelena Klawitter
- iC42 Clinical Research and Development, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Joshua M Thurman
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO
| |
Collapse
|
30
|
Said AS, Rogers SC, Doctor A. Physiologic Impact of Circulating RBC Microparticles upon Blood-Vascular Interactions. Front Physiol 2018; 8:1120. [PMID: 29379445 PMCID: PMC5770796 DOI: 10.3389/fphys.2017.01120] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Here, we review current data elucidating the role of red blood cell derived microparticles (RMPs) in normal vascular physiology and disease progression. Microparticles (MPs) are submicron-size, membrane-encapsulated vesicles derived from various parent cell types. MPs are produced in response to numerous stimuli that promote a sequence of cytoskeletal and membrane phospholipid changes and resulting MP genesis. MPs were originally considered as potential biomarkers for multiple disease processes and more recently are recognized to have pleiotropic biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in initiating apoptosis. RMPs, specifically, form normally during RBC maturation in response to injury during circulation, and are copiously produced during processing and storage for transfusion. Notably, several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs markedly from that of intact RBCs and the nature/composition of RMP components are affected by the specific circumstances of RMP genesis. Described RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion as well as influence upon vasoregulation via influence upon nitric oxide (NO) bioavailability. Of particular relevance, RMPs scavenge NO more avidly than do intact RBCs; this physiology has been proposed to contribute to the impaired oxygen delivery homeostasis that may be observed following transfusion. In summary, RMPs are submicron particles released from RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in normal and patho-physiology and in transfusion recipients is an area of continued investigation.
Collapse
Affiliation(s)
- Ahmed S Said
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Stephen C Rogers
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Allan Doctor
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States.,Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
31
|
de la Cuesta F, Baldan-Martin M, Moreno-Luna R, Alvarez-Llamas G, Gonzalez-Calero L, Mourino-Alvarez L, Sastre-Oliva T, López JA, Vázquez J, Ruiz-Hurtado G, Segura J, Vivanco F, Ruilope LM, Barderas MG. Kalirin and CHD7: novel endothelial dysfunction indicators in circulating extracellular vesicles from hypertensive patients with albuminuria. Oncotarget 2017; 8:15553-15562. [PMID: 28152519 PMCID: PMC5362505 DOI: 10.18632/oncotarget.14948] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Despite of the great advances in anti-hypertensive therapies, many patients under Renin-Angiotensin- System (RAS) suppression develop albuminuria, which is a clear indicator of therapeutic inefficiency. Hence, indicators of vascular function are needed to assess patients’ condition and help deciding future therapies. Proteomic analysis of circulating extracellular vesicles (EVs) showed two proteins, kalirin and chromodomain-helicase-DNA-binding protein 7 (CHD7), increased in albuminuric patients. A positive correlation of both with the expression of the endothelial activation marker E-selectin was found in EVs. In vitro analysis using TNFα-treated adult human endothelial cells proved their involvement in endothelial cell activation. Hence, we propose protein levels of kalirin and CHD7 in circulating EVs as novel endothelial dysfunction markers to monitor vascular condition in hypertensive patients with albuminuria.
Collapse
Affiliation(s)
- Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain.,Current address: Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Montserrat Baldan-Martin
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | - Rafael Moreno-Luna
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | | | | | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | | | | | - Gema Ruiz-Hurtado
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Julian Segura
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundacion Jimenez Diaz, Madrid, Spain.,Departamento de Bioquimica y Biologia Molecular I, Universidad Complutense, Madrid, Spain
| | - Luis M Ruilope
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| |
Collapse
|
32
|
Abstract
Extracellular vesicles, such as exosomes and microvesicles, are host cell-derived packages of information that allow cell-cell communication and enable cells to rid themselves of unwanted substances. The release and uptake of extracellular vesicles has important physiological functions and may also contribute to the development and propagation of inflammatory, vascular, malignant, infectious and neurodegenerative diseases. This Review describes the different types of extracellular vesicles, how they are detected and the mechanisms by which they communicate with cells and transfer information. We also describe their physiological functions in cellular interactions, such as in thrombosis, immune modulation, cell proliferation, tissue regeneration and matrix modulation, with an emphasis on renal processes. We discuss how the detection of extracellular vesicles could be utilized as biomarkers of renal disease and how they might contribute to disease processes in the kidney, such as in acute kidney injury, chronic kidney disease, renal transplantation, thrombotic microangiopathies, vasculitides, IgA nephropathy, nephrotic syndrome, urinary tract infection, cystic kidney disease and tubulopathies. Finally, we consider how the release or uptake of extracellular vesicles can be blocked, as well as the associated benefits and risks, and how extracellular vesicles might be used to treat renal diseases by delivering therapeutics to specific cells.
Collapse
Affiliation(s)
- Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| | - Anne-Lie Ståhl
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| | - Ida Arvidsson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| |
Collapse
|
33
|
Endothelial progenitor cells and hypertension: current concepts and future implications. Clin Sci (Lond) 2017; 130:2029-2042. [PMID: 27729472 DOI: 10.1042/cs20160587] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
Abstract
The discovery of endothelial progenitor cells (EPCs), a group of cells that play important roles in angiogenesis and the maintenance of vascular endothelial integrity, has led to considerable improvements in our understanding of the circulatory system and the regulatory mechanisms of vascular homoeostasis. Despite lingering disputes over where EPCs actually originate and how they facilitate angiogenesis, extensive research in the past decade has brought about significant advancements in this field of research, establishing EPCs as an essential element in the pathogenesis of various diseases. EPC and hypertensive disorders, especially essential hypertension (EH, also known as primary hypertension), represent one of the most appealing branches in this area of research. Chronic hypertension remains a major threat to public health, and the exact pathologic mechanisms of EH have never been fully elucidated. Is there a relationship between EPC and hypertension? If so, what is the nature of such relationship-is it mediated by blood pressure alterations, or other factors that lie in between? How can our current knowledge about EPCs be utilized to advance the prevention and clinical management of hypertension? In this review, we set out to answer these questions by summarizing the current concepts about EPC pathophysiology in the context of hypertension, while attempting to point out directions for future research on this subject.
Collapse
|
34
|
Influence of red blood cell-derived microparticles upon vasoregulation. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2017; 15:522-534. [PMID: 28686154 DOI: 10.2450/2017.0353-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022]
Abstract
Here we review recent data and the evolving understanding of the role of red blood cell-derived microparticles (RMPs) in normal physiology and in disease progression. Microparticles (MPs) are small membrane vesicles derived from various parent cell types. MPs are produced in response to a variety of stimuli through several cytoskeletal and membrane phospholipid changes. MPs have been investigated as potential biomarkers for multiple disease processes and are thought to have biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in apoptosis. Specifically, RMPs are produced normally during RBC maturation and their production is accelerated during processing and storage for transfusion. Several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs from that of intact RBCs, and the nature and composition of RMP components are affected by both storage duration and the character of storage solutions. Recognised RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion, as well as influence upon vasoregulation via nitric oxide (NO) scavenging. Of particular relevance, RMPs are more avid NO scavengers than intact RBCs and this feature has been proposed as a mechanism for the impaired oxygen delivery homeostasis that has been observed following transfusion. Preliminary human studies demonstrate that circulating RMP abundance increases with RBC transfusion and is associated with altered plasma vasoactivity and abnormal vasoregulation. In summary, RMPs are submicron particles released from stored RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in transfusion recipients is an area of continued investigation.
Collapse
|
35
|
Abstract
Heart failure (HF) continues to have a sufficient impact on morbidity, mortality, and disability in developed countries. Growing evidence supports the hypothesis that microparticles (MPs) might contribute to the pathogenesis of the HF development playing a pivotal role in the regulation of the endogenous repair system, thrombosis, coagulation, inflammation, immunity, and metabolic memory phenomenon. Therefore, there is a large body of data clarifying the predictive value of MP numerous in circulation among subjects with HF. Although the determination of MP signature is better than measurement of single MP circulating level, there is not yet close confirmation that immune phenotype of cells produced MPs are important for HF prediction and development. The aim of the chapter is to summarize knowledge regarding the role of various MPs in diagnosis and prognosis of HF. The role of MPs as a delivery vehicle for drugs attenuated cardiac remodeling is considered.
Collapse
|
36
|
Cherré S, Fernandes E, Germano J, Dias T, Cardoso S, Piedade MS, Rozlosnik N, Oliveira MI, Freitas PP. Rapid and specific detection of cell-derived microvesicles using a magnetoresistive biochip. Analyst 2017; 142:979-986. [DOI: 10.1039/c6an02651f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Specific and sensitive detection of endothelial MVs within physiologically relevant concentrations using a magnetoresistive biochip platform.
Collapse
Affiliation(s)
- Solène Cherré
- Department of Micro- and Nanotechnology
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| | | | - José Germano
- INESC Microsystems and Nanotechnologies and Instituto de Nanociencias e Nanotecnologias
- 1000-029 Lisbon
- Portugal
| | - Tomás Dias
- INESC Microsystems and Nanotechnologies and Instituto de Nanociencias e Nanotecnologias
- 1000-029 Lisbon
- Portugal
- Instituto Superior Tecnico
- Universidade de Lisboa
| | - Susana Cardoso
- INESC Microsystems and Nanotechnologies and Instituto de Nanociencias e Nanotecnologias
- 1000-029 Lisbon
- Portugal
- Instituto Superior Tecnico
- Universidade de Lisboa
| | - Moisés S. Piedade
- INESC Microsystems and Nanotechnologies and Instituto de Nanociencias e Nanotecnologias
- 1000-029 Lisbon
- Portugal
- Instituto de Engenharia de Sistemas e Computadores-Investigaçao e Desenvolvimento (INESC ID)
- Lisbon
| | - Noemi Rozlosnik
- Department of Micro- and Nanotechnology
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| | - Marta I. Oliveira
- International Iberian Nanotechnology Laboratory
- 4715-330, Braga
- Portugal
| | - Paulo P. Freitas
- International Iberian Nanotechnology Laboratory
- 4715-330, Braga
- Portugal
| |
Collapse
|
37
|
Murphy TP, Cooper CJ, Pencina KM, D'Agostino R, Massaro J, Cutlip DE, Jamerson K, Matsumoto AH, Henrich W, Shapiro JI, Tuttle KR, Cohen DJ, Steffes M, Gao Q, Metzger DC, Abernethy WB, Textor SC, Briguglio J, Hirsch AT, Tobe S, Dworkin LD. Relationship of Albuminuria and Renal Artery Stent Outcomes: Results From the CORAL Randomized Clinical Trial (Cardiovascular Outcomes With Renal Artery Lesions). Hypertension 2016; 68:1145-1152. [PMID: 27647847 DOI: 10.1161/hypertensionaha.116.07744] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/15/2016] [Indexed: 11/16/2022]
Abstract
Randomized clinical trials have not shown an additional clinical benefit of renal artery stent placement over optimal medical therapy alone. However, studies of renal artery stent placement have not examined the relationship of albuminuria and treatment group outcomes. The CORAL study (Cardiovascular Outcomes in Renal Atherosclerotic Lesions) is a prospective clinical trial of 947 participants with atherosclerotic renal artery stenosis randomized to optimal medical therapy with or without renal artery stent which showed no treatment differences (3(5.8% and 35.1% event rate at mean 43-month follow-up). In a post hoc analysis, the study population was stratified by the median baseline urine albumin/creatinine ratio (n=826) and analyzed for the 5-year incidence of the primary end point (myocardial infarction, hospitalization for congestive heart failure, stroke, renal replacement therapy, progressive renal insufficiency, or cardiovascular disease- or kidney disease-related death), for each component of the primary end point, and overall survival. When baseline urine albumin/creatinine ratio was ≤ median (22.5 mg/g, n=413), renal artery stenting was associated with significantly better event-free survival from the primary composite end point (73% versus 59% at 5 years; P=0.02), cardiovascular disease-related death (93% versus 85%; P≤ 0.01), progressive renal insufficiency (91% versus 77%; P=0.03), and overall survival (89% versus 76%; P≤0.01), but not when baseline urine albumin/creatinine ratio was greater than median (n=413). These data suggest that low albuminuria may indicate a potentially large subgroup of those with renal artery stenosis that could experience improved event-free and overall-survival after renal artery stent placement plus optimal medical therapy compared with optimal medical therapy alone. Further research is needed to confirm these preliminary observations. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT00081731.
Collapse
Affiliation(s)
- Timothy P Murphy
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada.
| | - Christopher J Cooper
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Karol M Pencina
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Ralph D'Agostino
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Joseph Massaro
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Donald E Cutlip
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Kenneth Jamerson
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Alan H Matsumoto
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - William Henrich
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Joseph I Shapiro
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Katherine R Tuttle
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - David J Cohen
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Michael Steffes
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Qi Gao
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - D Christopher Metzger
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - William B Abernethy
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Stephen C Textor
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - John Briguglio
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Alan T Hirsch
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Sheldon Tobe
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| | - Lance D Dworkin
- From the Departments of Diagnostic Imaging (T.P.M.) and Medicine (L.D.D.), Rhode Island Hospital, Providence; Alpert Medical School of Brown University, Providence, RI (T.P.M., L.D.D.); Department of Medicine, University of Toledo, OH (C.J.C.); Departments of Statistics (K.M.P., R.B.D, J.M.M.), Medicine (D.E.C.), and Biostatistics (Q.G.), Harvard Clinical Research Institute, Boston, MA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (D.E.C.); Department of Mathematics and Statistics, Boston University, MA (R.B.D.); Department of Medicine, University of Michigan, Ann Arbor (K.J.); Department of Radiology, University of Virginia, Charlottesville (A.H.M.); Department of Medicine, University of Texas Health Science Center, San Antonio (W.H.); Department of Medicine, Marshall University, Huntington, WV (J.I.S.); Department of Medicine, Providence Health Care and University of Washington School of Medicine, Spokane (K.R.T.); Department of Medicine, St. Luke's Hospital, Kansas City, MO (D.J.C.); Departments of Pathology (M.S.) and Medicine (A.H.), The University of Minnesota Medical School, Minneapolis; Department of Medicine, Wellmont-Holston Valley Medical Center, Kingsport, TN (D.C.M.); Department of Medicine, Asheville Cardiology Associates, NC (W.B.A.); Department of Medicine, Mayo Clinic, Rochester, MN (S.C.T.); Department of Radiology, Lancaster General Hospital, PA (J.B.); and Department of Medicine, Sunnybrook Research Institute (S.T.), Toronto, Ontario, Canada
| |
Collapse
|
38
|
Durante A, Zalewski J. Microparticles: A Novel Player in Cardiovascular Diseases. Cardiology 2016; 132:249-51. [PMID: 26329533 DOI: 10.1159/000437045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 11/19/2022]
|
39
|
Hsu CY, Huang PH, Chen TH, Chiang CH, Leu HB, Huang CC, Chen JW, Lin SJ. Increased Circulating Visfatin Is Associated With Progression of Kidney Disease in Non-Diabetic Hypertensive Patients. Am J Hypertens 2016; 29:528-36. [PMID: 26298010 DOI: 10.1093/ajh/hpv132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/20/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUD Declining renal function is an independent risk factor for all-cause mortality in cardiovascular disease. Visfatin has been described as a marker of inflammation and endothelial dysfunction, but whether circulating visfatin levels are predictive to a subsequent decline in renal function remains unclear. METHODS In total, 200 nondiabetic, non-proteinuric hypertensive outpatients with initial serum creatinine (Scr) ≤1.5 mg/dl were enrolled. Plasma visfatin concentration and endothelial function estimated by brachial artery flow-mediated dilatation (FMD) were determined in the study subjects. The primary endpoints were the occurrence of renal events including doubling of Scr, 25% loss of glomerular filtration rate (GFR) from baseline values, and the occurrence of end-stage renal disease during follow-up. RESULTS The mean annual rate of GFR decline (ΔGFR/y) was -1.26±2.76 ml/min/1.73 m(2) per year during follow-up (8.6±2.5 years). At baseline, plasma visfatin was negatively correlated with estimated GFR. In longitudinal analysis, the ΔGFR/y was correlated with visfatin, baseline GFR, FMD, systolic blood pressure, and fasting blood glucose (FBG). Multivariate analysis indicated that increased visfatin (r = -0.331, P <0.001), baseline GFR (r = -0.234, P = 0.001), FMD (r = 0.163, P = 0.015), and FBG (r = -0.160, P = 0.015) are independent predictors of ΔeGFR/y. Cox regression model analysis showed that visfatin (hazard ratio (HR), 1.09; 95% confidence interval (CI), 1.05-1.13, P <0.001), FBG (HR, 1.01; 95% CI, 1.00-1.02, P = 0.020), and FMD (HR, 0.87; 95% CI, 0.76-1.00, P = 0.049) were independently associated with the risk of developing future renal events. CONCLUSIONS Increased circulating visfatin are associated with subsequent decline in renal function in nondiabetic hypertensive patients.
Collapse
Affiliation(s)
- Chien-Yi Hsu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medicine, Taipei Veterans General Hospital Yuli Branch, Hualien, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan;
| | - Tz-Heng Chen
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Hung Chiang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Bang Leu
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Chou Huang
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
40
|
Helmke A, von Vietinghoff S. Extracellular vesicles as mediators of vascular inflammation in kidney disease. World J Nephrol 2016; 5:125-38. [PMID: 26981436 PMCID: PMC4777783 DOI: 10.5527/wjn.v5.i2.125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/18/2015] [Accepted: 01/08/2016] [Indexed: 02/06/2023] Open
Abstract
Vascular inflammation is a common cause of renal impairment and a major cause of morbidity and mortality of patients with kidney disease. Current studies consistently show an increase of extracellular vesicles (EVs) in acute vasculitis and in patients with atherosclerosis. Recent research has elucidated mechanisms that mediate vascular wall leukocyte accumulation and differentiation. This review addresses the role of EVs in this process. Part one of this review addresses functional roles of EVs in renal vasculitis. Most published data address anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis and indicate that the number of EVs, mostly of platelet origin, is increased in active disease. EVs generated from neutrophils by activation by ANCA can contribute to vessel damage. While EVs are also elevated in other types of autoimmune vasculitis with renal involvement such as systemic lupus erythematodes, functional consequences beyond intravascular thrombosis remain to be established. In typical hemolytic uremic syndrome secondary to infection with shiga toxin producing Escherichia coli, EV numbers are elevated and contribute to toxin distribution into the vascular wall. Part two addresses mechanisms how EVs modulate vascular inflammation in atherosclerosis, a process that is aggravated in uremia. Elevated numbers of circulating endothelial EVs were associated with atherosclerotic complications in a number of studies in patients with and without kidney disease. Uremic endothelial EVs are defective in induction of vascular relaxation. Neutrophil adhesion and transmigration and intravascular thrombus formation are critically modulated by EVs, a process that is amenable to therapeutic interventions. EVs can enhance monocyte adhesion to the endothelium and modulate macrophage differentiation and cytokine production with major influence on the local inflammatory milieu in the plaque. They significantly influence lipid phagocytosis and antigen presentation by mononuclear phagocytes. Finally, platelet, erythrocyte and monocyte EVs cooperate in shaping adaptive T cell immunity. Future research is needed to define changes in uremic EVs and their differential effects on inflammatory leukocytes in the vessel wall.
Collapse
|
41
|
Berezin AE, Kremzer AA, Martovitskaya YV, Berezina TA, Gromenko EA. Pattern of endothelial progenitor cells and apoptotic endothelial cell-derived microparticles in chronic heart failure patients with preserved and reduced left ventricular ejection fraction. EBioMedicine 2016; 4:86-94. [PMID: 26981573 PMCID: PMC4776070 DOI: 10.1016/j.ebiom.2016.01.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/01/2016] [Accepted: 01/14/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chronic heart failure (HF) remains a leading cause of cardiovascular (CV) mortality and morbidity worldwide. The aim of the study was to investigate whether the pattern of angiogenic endothelial progenitor cells (EPCs) and apoptotic endothelial cell-derived microparticles (EMPs) would be able to differentiate HF with reduced (HFrEF) and preserved (HFpEF) ejection fraction. METHODS One hundred sixty four chronic HF subjects met inclusion criteria. Patients with global left ventricular ejection fraction ≥ 50% were categorized as the HFpEF group (n = 79) and those with ≤ 45% as the HFrEF group (n = 85). Therefore, to compare the circulating levels of biological markers 35 control subjects without HF were included in the study. All control individuals were age- and sex-matched chronic HF patients. The serum level of biomarkers was measured at baseline. The flow cytometric technique was used for predictably distinguishing circulating cell subsets depending on expression of CD45, CD34, CD14, Tie-2, and CD309 antigens and determining endothelial cell-derived microparticles. CD31(+)/annexin V(+) was defined as apoptotic endothelial cell-derived MPs, MPs labeled for CD105(+) or CD62E(+) were determined as MPs produced due to activation of endothelial cells. RESULTS In multivariate logistic regression model T2DM (R(2) = 0.26; P = 0.001), obesity (R(2) = 0.22; P = 0.001), previous MI (R(2) = 0.17; P = 0.012), galectin-3 (R(2) = 0.67; P = 0.012), CD31(+)/annexin V(+) EMPs (R(2) = 0.11; P = 0.001), NT-proBNP (R(2) = 0.11; P = 0.046), CD14(+) CD309(+) cells (R(2) = 0.058; P = 0.001), and CD14(+) СD309(+) Tie-2(+) cells (R(2) = 0.044; P = 0.028) were found as independent predictors of HFpEF. Using multivariate Cox-regression analysis adjusted etiology (previous myocardial infarction), cardiovascular risk factors (obesity, type 2 diabetes mellitus) we found that NT-proBNP (OR 1.08; 95% CI = 1.03-1.12; P = 0.001) and CD31(+)/annexin V(+) EMPs to CD14(+) CD309(+) cell ratio (OR 1.06; 95% CI = 1.02-1.11; P = 0.02) were independent predictors for HFpEF. CONCLUSION We found that CD31(+)/annexin V(+) EMPs to CD14(+) CD309(+) cell ratio added to NT-proBNP, clinical data, and cardiovascular risk factors has exhibited the best discriminate value and higher reliability to predict HFpEF compared with NT-proBNP and clinical data/cardiovascular risk factors alone.
Collapse
Affiliation(s)
- Alexander E. Berezin
- Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye UA-69035, Ukraine
| | | | - Yulia V. Martovitskaya
- Pathology and Immunology Department, Clinical Laboratory “Dia-Service”, Zaporozhye, Ukraine
| | | | | |
Collapse
|
42
|
de la Cuesta F, Baldan-Martin M, Mourino-Alvarez L, Sastre-Oliva T, Alvarez-Llamas G, Gonzalez-Calero L, Ruiz-Hurtado G, Segura J, Vivanco F, Ruilope LM, Barderas MG. [Cardiovascular risk study in patients with renin-angiotensin system blockade by means of the proteone of circulating extracellular vesicles]. HIPERTENSION Y RIESGO VASCULAR 2016; 33:21-7. [PMID: 26826536 DOI: 10.1016/j.hipert.2015.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are released to the bloodstream by certain cell types due to transport, activation and cell death processes. Blood count of EVs from platelet and endothelial origin has been proved to be a cardiovascular risk biomarker. Thus, EVs proteome might reflect the underlying cellular processes in hypertensive patients with albuminuria. MATERIAL AND METHODS Protein content of circulating EVs was analyzed by liquid chromatography coupled to mass spectrometry. EVs were isolated by an ultracentrifugation protocol optimized in order to avoid contamination by blood plasma proteins. Purity of the isolated fraction was verified by electronic and confocal microscopy, and by flow cytometry. RESULTS We hereby show a method to isolate circulating EVs from hypertensive patients with/without albuminuria with high yield and purity. Besides, we provide a reference proteome of the EVs of these patients, composed of 2,463 proteins, and prove that the proteins carried by these vesicles are associated with crucial processes involved in the inherent cardiovascular risk. CONCLUSION The proteome of circulating EVs is an interesting source of indicators in the evaluation of cardiovascular risk in hypertensive patients with renin-angiotensin system blockage.
Collapse
Affiliation(s)
- F de la Cuesta
- Laboratorio de Fisiopatología Vascular, Hospital Nacional de Parapléjicos (HNP), Servicio de Salud de Castilla la Mancha (SESCAM), Toledo, España.
| | - M Baldan-Martin
- Laboratorio de Fisiopatología Vascular, Hospital Nacional de Parapléjicos (HNP), Servicio de Salud de Castilla la Mancha (SESCAM), Toledo, España
| | - L Mourino-Alvarez
- Laboratorio de Fisiopatología Vascular, Hospital Nacional de Parapléjicos (HNP), Servicio de Salud de Castilla la Mancha (SESCAM), Toledo, España
| | - T Sastre-Oliva
- Laboratorio de Fisiopatología Vascular, Hospital Nacional de Parapléjicos (HNP), Servicio de Salud de Castilla la Mancha (SESCAM), Toledo, España
| | - G Alvarez-Llamas
- Departamento de Inmunología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - L Gonzalez-Calero
- Departamento de Inmunología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - G Ruiz-Hurtado
- Departamento de Riesgo Cardiovascular e Hipertensión, IIS-Hospital 12 de Octubre, Madrid, España
| | - J Segura
- Unidad de Hipertensión, Hospital 12 de Octubre, Madrid, España
| | - F Vivanco
- Departamento de Inmunología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - L M Ruilope
- Departamento de Riesgo Cardiovascular e Hipertensión, IIS-Hospital 12 de Octubre, Madrid, España
| | - M G Barderas
- Laboratorio de Fisiopatología Vascular, Hospital Nacional de Parapléjicos (HNP), Servicio de Salud de Castilla la Mancha (SESCAM), Toledo, España
| |
Collapse
|
43
|
Fonseca HAR, Fonseca FA, Lins LC, Monteiro AM, Bianco HT, Brandão SA, Povoa RM, Juliano L, Figueiredo-Neto AM, Boschcov P, Gidlund M, Izar MC. Antihypertensive therapy increases natural immunity response in hypertensive patients. Life Sci 2015; 143:124-30. [PMID: 26514303 DOI: 10.1016/j.lfs.2015.10.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/04/2015] [Accepted: 10/23/2015] [Indexed: 12/21/2022]
Abstract
AIMS The aim of this work was to evaluate the effects of treatment of hypertension on the autoantibodies to apolipoprotein B-derived peptides (anti-ApoB-D peptide Abs) response, inflammation markers and vascular function. MAIN METHODS Eighty-eight patients with hypertension (stage 1 or 2) were recruited and advised to receive perindopril (4mg), hydrochlorothiazide (25mg), or indapamide (1.5mg) for 12weeks in a blinded fashion. Office and 24-h ambulatory blood pressure monitoring (24h ABPM), flow-mediated dilatation (FMD), nitrate-induced dilatation (NID), titers of IgG and IgM anti-ApoB-D peptide Abs, hsCRP, and interleukins (IL-8 and IL-10) were evaluated at baseline and 12weeks after therapies. KEY FINDINGS All treatments reduced office BP, and improved FMD (P<0.05 vs. baseline). The NID was improved only in the perindopril arm (P<0.05 vs. baseline). The 24h-ABPM was reduced with perindopril and hydrochlorothiazide therapies (P<0.05 vs. baseline), but not with indapamide, and this effect was followed by increase in titers of IgM Anti-ApoB-D peptide Abs (P<0.05 vs. baseline), without modifications in titers IgG Anti-ApoB-D peptide Abs and interleukins. Multivariable regression analysis has shown that change in the titers of IgM anti-ApoB-D peptide was associated with the changes in FMD (β -0.347; P<0.05). SIGNIFICANCE These findings shed light to a possible modulator effect of the antihypertensive therapy on the natural immunity responses and vascular function.
Collapse
Affiliation(s)
| | - Francisco A Fonseca
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Lívia C Lins
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Andrea M Monteiro
- Department of Immunology, Institute of Biomedical Sciences IV, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Henrique T Bianco
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Sergio A Brandão
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rui M Povoa
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Luiz Juliano
- Department of Biophysics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Paulo Boschcov
- Department of Biophysics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Magnus Gidlund
- Department of Immunology, Institute of Biomedical Sciences IV, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria C Izar
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
44
|
Shih CL, Chong KY, Hsu SC, Chien HJ, Ma CT, Chang JWC, Yu CJ, Chiou CC. Development of a magnetic bead-based method for the collection of circulating extracellular vesicles. N Biotechnol 2015; 33:116-22. [PMID: 26409934 DOI: 10.1016/j.nbt.2015.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/28/2015] [Accepted: 09/17/2015] [Indexed: 12/19/2022]
Abstract
Cells release different types of extracellular vesicles (EVs). These EVs contain biomolecules, including proteins and nucleic acids, from their parent cells, which can be useful for diagnostic applications. The aim of this study was to develop a convenient procedure to collect circulating EVs with detectable mRNA or other biomolecules. Magnetic beads coated with annexin A5 (ANX-beads), which bound to phosphatidylserine moieties on the surfaces of most EVs, were tested for their ability to capture induced apoptotic bodies in vitro and other phosphatidylserine-presenting vesicles in body fluids. Our results show that up to 60% of induced apoptotic bodies could be captured by the ANX-beads. The vesicles captured from cultured media or plasma contained amplifiable RNA. Suitable blood samples for EV collection included EDTA-plasma and serum but not heparin-plasma. In addition, EVs in plasma were labile to freeze-and-thaw cycles. In rodents xenografted with human cancer cells, tumor-derived mRNA could be detected in EVs captured from serum samples. Active proteins could be detected in EVs captured from ascites but not from plasma. In conclusion, we have developed a magnetic bead-based procedure for the collection of EVs from body fluids and proved that captured EVs contain biomolecules from their parent cells, and therefore have great potential for disease diagnosis.
Collapse
Affiliation(s)
- Chun-Liang Shih
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kowit-Yu Chong
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Che Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Jung Chien
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Ting Ma
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Jung Yu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chiuan-Chian Chiou
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
45
|
Microparticle levels after arterial injury and NO therapy in diabetes. J Surg Res 2015; 200:722-31. [PMID: 26490225 DOI: 10.1016/j.jss.2015.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/16/2015] [Accepted: 08/17/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Little is known about how arterial injury, nitric oxide (NO), or the diabetic milieu impact microparticle (MP) levels in the vasculature. We hypothesized that MP levels would increase following local arterial injury, and that NO would modify MP levels differently based on the metabolic environment. MATERIALS AND METHODS Type 1 diabetes was induced in male Lean Zucker (LZ) rats with streptozotocin, and type 2 diabetes was induced in male Zucker diabetic fatty rats through diet. Lean Zucker rats served as nondiabetic controls. The rat carotid balloon injury was performed ± NO (n > 4/group). Blood was obtained at intervals from baseline to 14 d after injury and analyzed for platelet MP (PMP), leukocyte MP (LMP), and endothelial MP (EMP) using fluorescence-activated cell sorting (FACS) analysis. RESULTS At baseline, type 1 diabetic rats had the highest EMP levels (P < 0.05). After arterial injury, type 1 and type 2 diabetic rats had a transient increase in EMP levels (P < 0.05) before decreasing below baseline levels. Both LMP and PMP levels generally declined after injury in all three animal models but were the lowest in both type 1 and type 2 diabetic rats. NO therapy had little impact on MP levels in nondiabetic and type 1 diabetic rats after injury. Conversely, NO caused a dramatic increase in EMP, LMP, and PMP levels in type 2 diabetic animals at early time points after injury (P < 0.05). CONCLUSIONS These data demonstrate that the diabetic milieu impacts MP levels at baseline, after arterial injury and with NO treatment.
Collapse
|
46
|
Wu TC, Chan JS, Lee CY, Leu HB, Huang PH, Chen JS, Lin SJ, Chen JW. Rivaroxaban, a factor Xa inhibitor, improves neovascularization in the ischemic hindlimb of streptozotocin-induced diabetic mice. Cardiovasc Diabetol 2015; 14:81. [PMID: 26077117 PMCID: PMC4473833 DOI: 10.1186/s12933-015-0243-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/06/2015] [Indexed: 01/13/2023] Open
Abstract
Background Factor Xa inhibitor is used for preventing venous thromboembolism (VTE) in adult patients receiving orthopedic operation. However, the role of factor Xa inhibitor, rivaroxaban, in angiogenesis is still unknown. Methods and results Streptozotocin (STZ)–induced diabetic mice with model of hind-limb ischemia, were divided into non-diabetic control, diabetic control, and low- and high-dose rivaroxaban treatment groups, in order to evaluate the effect of rivaroxaban in angiogenesis. Doppler perfusion imaging showed that blood flow recovery was significantly increased, and more capillary density occurred in the rivaroxaban treatment group. In vitro studies, human endothelial progenitor cells (EPCs) treated with rivaroxaban had significant functional improvement in migration and senescence under hyperglycemic conditions. Rivaroxaban also increased endothelial nitric oxide synthase (eNOS) as well as vascular endothelial growth factor (VEGF) expressions in hyperglycemia-stimulated EPCs. Conclusions Rivaroxaban promoted vessel formation in diabetic mice and improved endothelial progenitor cell function under hyperglycemic conditions. These effects may be associated with enhancement of expression of eNOS and VEGF.
Collapse
Affiliation(s)
- Tao-Cheng Wu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Renal Division, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan County, Taiwan
| | - Chiu-Yang Lee
- Division of Cardiovascular Surgery, Department of Surgery, Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiovascular Surgery, Department of Surgery, Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Jia-Shiong Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research and Education, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei 112, Taiwan ROC
| | - Jaw-Wen Chen
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Department of Medical Research and Education, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei 112, Taiwan ROC. .,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
47
|
Berezin A, Zulli A, Kerrigan S, Petrovic D, Kruzliak P. Predictive role of circulating endothelial-derived microparticles in cardiovascular diseases. Clin Biochem 2015; 48:562-568. [PMID: 25697107 DOI: 10.1016/j.clinbiochem.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 12/31/2022]
Abstract
Endothelial-derived microparticles (EMPs) are a novel biological marker of endothelium injury and vasomotion disorders that are involved in pathogenesis of cardiovascular, metabolic, and inflammatory diseases. Circulating levels of EMPs are thought to reflect a balance between cell stimulation, proliferation, apoptosis, and cell death. Increased EMPs may be defined in several cardiovascular diseases, such as stable and unstable coronary artery disease, acute and chronic heart failure, hypertension, arrhythmias, thromboembolism, asymptomatic atherosclerosis as well as renal failure, metabolic disorders (including type two diabetes mellitus, abdominal obesity, metabolic syndrome, insulin resistance) and dyslipidemia. This review highlights the controversial opinions regarding impact of circulating EMPs in major cardiovascular and metabolic diseases and summarizes the perspective implementation of the EMPs in risk stratification models.
Collapse
Affiliation(s)
- Alexander Berezin
- Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | - Anthony Zulli
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Australia
| | - Steve Kerrigan
- Molecular and Cellular Therapeutics Department, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Daniel Petrovic
- Department of Histology and Embryology, School of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Kruzliak
- International Clinical Research Center, St. Anne's University Hospital, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
48
|
Association between microalbuminuria predicting in-stent restenosis after myocardial infarction and cellular senescence of endothelial progenitor cells. PLoS One 2015; 10:e0123733. [PMID: 25874702 PMCID: PMC4395282 DOI: 10.1371/journal.pone.0123733] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/26/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Relationship between microalbuminuria and worse outcome of coronary artery disease patients is discussed, but its underlying pathophysiological mechanism remains unclear. We investigated the role of microalbuminuria to the function of endothelial progenitor cells (EPCs), that might affect to outcome of acute myocardial infarction (AMI) patients. METHODS Forty-five AMI patients were divided into two groups according to their urinary albumin excretion: normal (n = 24) and microalbuminuria (>30 mg/day, n = 21). At day-2 and day-7 after AMI onset, circulating-EPCs (CD34+ Flk1+) were quantified by flow cytometry. The number of lectin-acLDL-positive cultured-EPCs immobilized on fibronectin was determined. To assess the cellular senescence of cultured-EPCs, the expression level of sirtuin-1 mRNA and the number of SA-β-gal positive cell were evaluated. Angiographic late in-stent loss after percutaneous coronary intervention (PCI) was evaluated at a six-month follow-up. RESULTS No significant differences in coronary risk and the extent of myocardial damage were observed between the two groups. Late in-stent loss at the six-month follow-up was significantly higher in the microalbuminuria group (normal:microalbuminuria = 0.76±0.34:1.18±0.57 mm, p=0.021). The number of circulating-EPCs was significantly increased in microalbuminuria group at day-7, however, improved adhesion of EPCs was observed in normal group but not in microalbuminuria group from baseline to day-7 (+3.1±8.3:-1.3±4.4%: p<0.05). On the other hand, in microalbuminuria group at day-7, the level of sirtuin-1 mRNA expression of cultured-EPCs was significantly decreased (7.1±8.9:2.5±3.7 fold, p<0.05), which was based on the negative correlation between the level of sirtuin-1 mRNA expression and the extent of microalbuminuria. The ratio of SA-β-gal-positive cells in microalbuminuria group was increased compared to that of normal group. CONCLUSIONS Microalbuminuria in AMI patients is closely associated with functional disorder of EPCs via cellular senescence, that predicts the aggravation of coronary remodeling after PCI.
Collapse
|
49
|
Michelucci A, Cesari F, Ricciardi G, Attanà P, Pieragnoli P, Ristalli F, Padeletti L, Gori AM, Gensini GF, Abbate R. Left ventricular mass and progenitor cells in chronic heart failure patients. Intern Emerg Med 2015; 10:329-35. [PMID: 25387824 DOI: 10.1007/s11739-014-1149-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/27/2014] [Indexed: 01/19/2023]
Abstract
The aim of the study was to evaluate the association between circulating (CPCs) and endothelial (EPCs) progenitor cells and left ventricular (LV) remodeling in chronic heart failure (HF). 85 HF patients, ranging 29-89 years, 83.5% males, 45.9% ischemic, NYHA functional class II-IV, with a LV ejection fraction ≤40% were studied. LV ejection fraction, LV end-diastolic and end-systolic (LVESV) volumes, LV mass and tricuspid annular plane systolic excursion (TAPSE) were evaluated, and, when indicated, indexed for body surface area (BSA). CPCs and EPCs number was assessed using flow cytometry. CPCs were defined as CD34+, CD133+ and CD34+/CD133+. EPCs, identified through their expression of KDR, were defined as CD34+/KDR+, CD133+/KDR+ and CD34+/CD133+/KDR+. All EPCs were negatively related to LVESV/BSA (r = -0.24, p = 0.02 for all EPC's populations), and to LVmass/BSA (CD34+KDR+; r = -0.30, p = 0.005; CD133+KDR+; r = -0.31, p = 0.004; CD34+CD133+KDR+; r = -0.29, p = 0.007). No differences in EPCs levels in relation to cardiovascular risk factors, medications, etiology, age or gender were observed. CPCs number was higher in women, and lower in ischemic patients. In logistic regression analyses, the low EPCs' number was associated with an increased likelihood of abnormal LVmass/BSA. CPCs proved to be higher and EPCs lower in patients with severely abnormal LVmass/BSA (gr/m(2), ≥122 in women and ≥149 in men). Our results suggest a correlation between LV remodeling and progenitor cells. This is noteworthy considering that it has been suggested that bone marrow-derived EPCs participate in cardiac regeneration and function recovery in the setting of progressive HF.
Collapse
Affiliation(s)
- Antonio Michelucci
- Section of Arrhythmology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Muñoz-Hernandez R, Vallejo-Vaz AJ, Sanchez Armengol A, Moreno-Luna R, Caballero-Eraso C, Macher HC, Villar J, Merino AM, Castell J, Capote F, Stiefel P. Obstructive sleep apnoea syndrome, endothelial function and markers of endothelialization. Changes after CPAP. PLoS One 2015; 10:e0122091. [PMID: 25815511 PMCID: PMC4376903 DOI: 10.1371/journal.pone.0122091] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/22/2015] [Indexed: 01/07/2023] Open
Abstract
Study objectives This study tries to assess the endothelial function in vivo using flow-mediated dilatation (FMD) and several biomarkers of endothelium formation/restoration and damage in patients with obstructive sleep apnoea (OSA) syndrome at baseline and after three months with CPAP therapy. Design Observational study, before and after CPAP therapy. Setting and Patients We studied 30 patients with apnoea/hypopnoea index (AHI) >15/h that were compared with themselves after three months of CPAP therapy. FMD was assessed non-invasively in vivo using the Laser-Doppler flowmetry. Circulating cell-free DNA (cf-DNA) and microparticles (MPs) were measured as markers of endothelial damage and the vascular endothelial growth factor (VEGF) was determined as a marker of endothelial restoration process. Measurements and results After three month with CPAP, FMD significantly increased (1072.26 ± 483.21 vs. 1604.38 ± 915.69 PU, p< 0.005) cf-DNA and MPs significantly decreased (187.93 ± 115.81 vs. 121.28 ± 78.98 pg/ml, p<0.01, and 69.60 ± 62.60 vs. 39.82 ± 22.14 U/μL, p<0.05, respectively) and VEGF levels increased (585.02 ± 246.06 vs. 641.11 ± 212.69 pg/ml, p<0.05). These changes were higher in patients with more severe disease. There was a relationship between markers of damage (r = -0.53, p<0.005) but not between markers of damage and restoration, thus suggesting that both types of markers should be measured together. Conclusions CPAP therapy improves FMD. This improvement may be related to an increase of endothelial restoration process and a decrease of endothelial damage.
Collapse
Affiliation(s)
- Rocio Muñoz-Hernandez
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Antonio J Vallejo-Vaz
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Angeles Sanchez Armengol
- Unidad del Sueño, Unidad Medico Quirúrgica de Enfermedades Respiratorias, Hospital Virgen del Rocío, Sevilla, Spain
| | - Rafael Moreno-Luna
- Departamento de Fisiopatología Vascular, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Candela Caballero-Eraso
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Unidad del Sueño, Unidad Medico Quirúrgica de Enfermedades Respiratorias, Hospital Virgen del Rocío, Sevilla, Spain
| | - Hada C Macher
- Servicio de Bioquímica Clínica, Hospital Virgen del Rocío, Sevilla, Spain
| | - Jose Villar
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Unidad Clínico Experimental de Riesgo Vascular (UCAMI), Hospital Virgen del Rocío, Sevilla, Spain
| | - Ana M Merino
- Instituto de Investigación Biomédica Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Castell
- UGC de Radiología, Hospital Virgen del Rocío, Sevilla, Spain
| | - Francisco Capote
- Unidad del Sueño, Unidad Medico Quirúrgica de Enfermedades Respiratorias, Hospital Virgen del Rocío, Sevilla, Spain
| | - Pablo Stiefel
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Unidad Clínico Experimental de Riesgo Vascular (UCAMI), Hospital Virgen del Rocío, Sevilla, Spain
| |
Collapse
|