1
|
Justa HCD, Baldissera AB, Machado MI, Souza SH, Polli NLC, Boia-Ferreira M, Schluga PHDC, Donatti L, Wille ACM, Minozzo JC, Gremski LH, Veiga SS. Induction of ectosome formation by binding of phospholipases D from Loxosceles venoms to endothelial cell surface: Mechanism of interaction. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159579. [PMID: 39547302 DOI: 10.1016/j.bbalip.2024.159579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Members of the phospholipase D (PLD) superfamily found in Loxosceles spider venoms are potent toxins with inflammatory and necrotizing activities. They degrade phospholipids in cell membranes, generating bioactive molecules that activate skin cells. These skin cells, in turn, activate leukocytes involved in dermonecrosis, characterized by aseptic coagulative necrosis. Although the literature has advanced in understanding the structure-function relationship, the cell biology resulting from the interactions of these molecules with cells remains poorly understood. In this study, we show that different cells exposed to recombinant PLDs bind these molecules to their plasma membrane, leading to the subsequent organization of extracellular microvesicles/ectosomes. The binding occurs as quickly as five minutes or less after exposure, increases over time, and eventually, the PLDs are expelled from the cell surface without generating cytotoxicity. PLDs are not endocytosed, nor do they spatially colocalize with acidic organelles in the intracellular environment. At least two regions of PLDs - the domain involved in magnesium ion coordination and the choline binding site - appear to play a role in cell surface binding and ectosome organization. However, the amino acids involved in catalysis do not participate in these events. The binding of these PLDs to the cell membrane, independent of catalytic activity, is sufficient to trigger intracellular signaling and enhance the expression of the pro-inflammatory IL-8 gene. These results are supported by the observation that isoforms of PLDs lacking catalytic activity induce an inflammatory response in vivo when injected into the skin of rabbits, without causing dermonecrosis. Our data indicate that these PLDs bind to the surface of target cells, promoting the organization of extracellular vesicles/ectosomes. Subsequently, these events activate pro-inflammatory genes and induce an inflammatory response in vivo. The binding to cells is not dependent on amino acids involved in catalysis but rather on amino acids involved in magnesium coordination. The binding of PLDs to the cell surface, formation of ectosomes, and activation of cells appear to initiate signals involved in inflammatory responses that can lead to dermonecrosis in accidents. This correlation is supported by experimental observations indicating that the events of toxin binding to cells, formation of microvesicles, and inflammatory responses observed both in vitro and in vivo are interconnected.
Collapse
Affiliation(s)
- Hanna Câmara da Justa
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | | | - Samira Hajjar Souza
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Marianna Boia-Ferreira
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Lucelia Donatti
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Ana Carolina M Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa (UEPG), Ponta Grossa 84030-900, Brazil
| | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products (CPPI), State Department of Health, Piraquara 83302-200, Brazil
| | - Luiza Helena Gremski
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Silvio S Veiga
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil.
| |
Collapse
|
2
|
Xagorari A, Iskas M, Papadopoulos V, Dimosthenous C, Gavriilaki E, Bougiouklis D, Sakellari I, Sotiropoulos D. Post-transplantation monitoring and quantitation of microparticles in allogeneic hematopoietic cell transplantation. Transpl Immunol 2024; 87:102133. [PMID: 39307436 DOI: 10.1016/j.trim.2024.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (allo-HCT) represents a curative treatment for various blood-related disorders, including hematological malignancies and genetic disorders. The success of this procedure hinges on the efficacy of the conditioning regimen and the graft's ability to engraft and function properly. Microparticles (MPs), small vesicles produced from stimulated, apoptotic, or activated cells, are involved in both physiological and pathological processes. However, the impact of MPs on allo-HCT remains poorly understood. OBJECTIVES This study aimed to investigate the presence of MPs from different cell types in grafts and patient plasma after allo-HCT, as well as their association with various parameters. We measured MPs from CD34+, CD56+, CD3+, CD19+, and CD33+ cells in grafts and patient plasma from day 0 to day 60 after transplantation. METHODS 224 blood samples were collected from 19 consecutive allo -HCT recipients at 0, +4, +14,+30 and + 60 day as well as from their grafts. MPs isolated from the plasma and quantified by flow cytometry analysis. RESULTS MP levels varied over time. Notably, CD34+ MP levels were linked to both early and late engraftment of neutrophils and platelets. Furthermore, grafts with high CD34+ and CD56+ MP levels in patient plasma on days 0 and + 4 were associated with late engraftment, whereas high CD33+ MP levels in both graft and patient plasma on day +4 were associated with early engraftment. Conditioning regimen affected CD19+ MP levels at day +14, and the number of CD34+, CD56+, and CD19+ MPs 30 days after transplantation was correlated with acute graft-versus-host disease. CONCLUSION These findings suggest that MPs derived from hematopoietic cells may play a significant role in the clinical course of patients following allo-HCT.
Collapse
Affiliation(s)
- A Xagorari
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece; Public Cord Blood Bank, Hematology Department, "G.Papanicolaou" Hospital, Thessaloniki, Greece.
| | - M Iskas
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece
| | - V Papadopoulos
- General Hospital of Pella - Giannitsa Unit, Giannitsa, Greece
| | - C Dimosthenous
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece
| | - E Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Greece
| | - D Bougiouklis
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece
| | - I Sakellari
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece
| | - D Sotiropoulos
- Bone Marrow Transplantation Unit, Hematology Department "G.Papanicolaou" Hospital, Thessaloniki, Greece; Public Cord Blood Bank, Hematology Department, "G.Papanicolaou" Hospital, Thessaloniki, Greece
| |
Collapse
|
3
|
Featherby SJ, Ettelaie C. Endothelial-derived microvesicles promote pro-migratory cross-talk with smooth muscle cells by a mechanism requiring tissue factor and PAR2 activation. Front Cardiovasc Med 2024; 11:1365008. [PMID: 38966751 PMCID: PMC11222581 DOI: 10.3389/fcvm.2024.1365008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Microvesicles (MV) released by endothelial cells (EC) following injury or inflammation contain tissue factor (TF) and mediate communication with the underlying smooth muscle cells (SMC). Ser253-phosphorylated TF co-localizes with filamin A at the leading edge of migrating SMC. In this study, the influence of endothelial-derived TF-MV, on human coronary artery SMC (HCASMC) migration was examined. Methods and Results MV derived from human coronary artery EC (HCAEC) expressing TFWt accelerated HCASMC migration, but was lower with cytoplasmic domain-deleted TF. Furthermore, incubation with TFAsp253-MV, or expression of TFAsp253 in HCASMC, reduced cell migration. Blocking TF-factor VIIa (TF-fVIIa) procoagulant/protease activity, or inhibiting PAR2 signaling on HCASMC, abolished the accelerated migration. Incubation with fVIIa alone increased HCASMC migration, but was significantly enhanced on supplementation with TF. Neither recombinant TF alone, factor Xa, nor PAR2-activating peptide (SLIGKV) influenced cell migration. In other experiments, HCASMC were transfected with peptides corresponding to the cytoplasmic domain of TF prior to stimulation with TF-fVIIa. Cell migration was suppressed only when the peptides were phosphorylated at position of Ser253. Expression of mutant forms of filamin A in HCASMC indicated that the enhancement of migration by TF but not by PDGF-BB, was dependent on the presence of repeat-24 within filamin A. Incubation of HCASMC with TFWt-MV significantly reduced the levels of Smoothelin-B protein, and upregulated FAK expression. Discussion In conclusion, Ser253-phosphorylated TF and fVIIa released as MV-cargo by EC, act in conjunction with PAR2 on SMC to promote migration and may be crucial for normal arterial homeostasis as well as, during development of vascular disease.
Collapse
|
4
|
Schiavello M, Vizio B, Bosco O, Pivetta E, Mariano F, Montrucchio G, Lupia E. Extracellular Vesicles: New Players in the Mechanisms of Sepsis- and COVID-19-Related Thromboinflammation. Int J Mol Sci 2023; 24:ijms24031920. [PMID: 36768242 PMCID: PMC9916541 DOI: 10.3390/ijms24031920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Sepsis and COVID-19 patients often manifest an imbalance in inflammation and coagulation, a complex pathological mechanism also named thromboinflammation, which strongly affects patient prognosis. Extracellular vesicles (EVs) are nanoparticles released by cells into extracellular space that have a relevant role in cell-to-cell communication. Recently, EVs have been shown to act as important players in a variety of pathologies, including cancer and cardiovascular disease. The biological properties of EVs in the mechanisms of thromboinflammation during sepsis and COVID-19 are still only partially known. Herein, we summarize the current experimental evidence on the role of EVs in thromboinflammation, both in bacterial sepsis and in COVID-19. A better understanding of EV involvement in these processes could be useful in describing novel diagnostic and therapeutic applications of EVs in these diseases.
Collapse
|
5
|
Increased Plasma Concentrations of Extracellular Vesicles Are Associated with Pro-Inflammatory and Pro-Thrombotic Characteristics of Left and Right Ventricle Mechanical Support Devices. J Cardiovasc Dev Dis 2023; 10:jcdd10010021. [PMID: 36661916 PMCID: PMC9866833 DOI: 10.3390/jcdd10010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023] Open
Abstract
Mechanical circulatory support (MCS) allows for functional left and right heart ventricle replacement. MCS induces a systemic inflammatory reaction and prothrombotic state leading to an increased risk of thrombus formation. The extracellular vesicles (EVs) are nanoparticles released from active/injured cells characterized by prothrombotic properties. Simple inflammatory parameters from whole blood count analysis have established a clinical role in everyday practice to describe immune-inflammatory activation. We hypothesized that increased plasma concentrations of EVs might be associated with the proinflammatory and pro-thrombotic characteristics of left ventricle assist device (LVAD) and right ventricle assist device (RVAD) devices. We presented a pilot study showing the concentration of peripheral blood serum, right and left ventricle mechanical assist device extracellular concentration in relation to thrombotic complication in patients treated with a biventricular pulsatile assist device (BIVAD). The observation was based on 12 replacements of pulsatile pumps during 175 days of observation. The proinflammatory characteristics of LVAD were noted. The proinflammatory and procoagulant activation by RVAD was observed. The results may provide possible explanations for the worse results of right-sided mechanical supports observed in clinical practice.
Collapse
|
6
|
Malingen SA, Rangamani P. Modelling membrane curvature generation using mechanics and machine learning. J R Soc Interface 2022; 19:20220448. [PMID: 36128706 PMCID: PMC9490339 DOI: 10.1098/rsif.2022.0448] [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: 06/15/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022] Open
Abstract
The deformation of cellular membranes regulates trafficking processes, such as exocytosis and endocytosis. Classically, the Helfrich continuum model is used to characterize the forces and mechanical parameters that cells tune to accomplish membrane shape changes. While this classical model effectively captures curvature generation, one of the core challenges in using it to approximate a biological process is selecting a set of mechanical parameters (including bending modulus and membrane tension) from a large set of reasonable values. We used the Helfrich model to generate a large synthetic dataset from a random sampling of realistic mechanical parameters and used this dataset to train machine-learning models. These models produced promising results, accurately classifying model behaviour and predicting membrane shape from mechanical parameters. We also note emerging methods in machine learning that can leverage the physical insight of the Helfrich model to improve performance and draw greater insight into how cells control membrane shape change.
Collapse
Affiliation(s)
- S. A. Malingen
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - P. Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Mei C, Peng F, Yin W, Xu W, Yao R, Li B, Zhou R, Fan X, Li N. Increased suicidal erythrocyte death in patients with hepatitis B-related acute-on-chronic liver failure. Am J Physiol Gastrointest Liver Physiol 2022; 323:G9-G20. [PMID: 35411804 DOI: 10.1152/ajpgi.00050.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Anemia is a common complication of hepatitis B-related acute-on-chronic liver failure (HB-ACLF). Eryptosis, a suicidal erythrocyte death characterized by phosphatidylserine (PS) externalization and red blood cell-derived microparticle (RMP) generation, decreases erythrocyte lifespan. Herein, we investigated whether enhanced eryptosis is involved in the anemia pathophysiology associated with HB-ACLF. PS exposure, cell volume, cytosolic Ca2+, and reactive oxygen species (ROS) production were determined using flow cytometry. RMPs were extracted using a polyethylene glycol (PEG)-based method. We found that hemoglobin (Hb) and hematocrit (Hct) were significantly lower in patients with HB-ACLF than in healthy controls (HC), patients with chronic hepatitis B (CHB), and patients with cirrhosis. The direct antiglobulin test positive rate was 75.9% in patients with HB-ACLF while its intensity was associated with anemia. The ratio of abnormal erythrocytes was higher in patients with HB-ACLF than in HC, CHB, and cirrhosis. The percentage of PS-exposed erythrocytes was higher in patients with HB-ACLF (2.07 ± 0.11%) compared with HC (0.37 ± 0.05%), CHB (0.38 ± 0.03%), and cirrhosis (0.38 ± 0.04%). The cytosolic Ca2+ and ROS abundance were also higher in patients with HB-ACLF compared with HC, patients with CHB, and patients with cirrhosis, and were inversely correlated with the anemia in patients with HB-ACLF. PS exposure of erythrocytes collected from HC was significantly pronounced following incubation in plasma from patients with HB-ACLF compared with incubation in plasma from HC. The protein concentration and RMPs size significantly increased in patients with HB-ACLF compared with HC. Thus, the anemia in patients with HB-ACLF is associated with increased eryptosis, which is partially triggered by increased cytosolic Ca2+ and oxidative stress.NEW & NOTEWORTHY Acute chronic liver failure (ACLF) is a critical syndrome characterized by multiple organ failures and high short-term mortality. A common complication of HB-ACLF is anemia, however, the mechanism of anemia in HB-ACLF remains to be elucidated. We confirm that the accelerated eryptosis is involved in the pathophysiology of anemia associated with HB-ACLF, which progressively aggravates the clinical outcome. Our study illustrates the mechanism regarding the anemia pathogenesis of HB-ACLF, which may be utilized further toward therapeutic ends.
Collapse
Affiliation(s)
- Cheng Mei
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China
| | - Fang Peng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenyu Yin
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China
| | - Wei Xu
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China
| | - Run Yao
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China
| | - Bijuan Li
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China
| | - Rongrong Zhou
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Hwang W, Shimizu M, Lee JW. Role of extracellular vesicles in severe pneumonia and sepsis. Expert Opin Biol Ther 2022; 22:747-762. [PMID: 35418256 PMCID: PMC9971738 DOI: 10.1080/14712598.2022.2066470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Extracellular vesicles (EV) released constitutively or following external stimuli from structural and immune cells are now recognized as important mediators of cell-to-cell communication. They are involved in the pathogenesis of pneumonia and sepsis, leading causes of acute respiratory distress syndrome (ARDS) where mortality rates remain up to 40%. Multiple investigators have demonstrated that one of the underlying mechanisms of the effects of EVs is through the transfer of EV content to host cells, resulting in apoptosis, inflammation, and permeability in target organs. AREAS COVERED The current review focuses on preclinical research examining the role of EVs released into the plasma and injured alveolus during pneumonia and sepsis. EXPERT OPINION Inflammation is associated with elevated levels of circulating EVs that are released by activated structural and immune cells and can have significant proinflammatory, procoagulant, and pro-permeability effects in critically ill patients with pneumonia and/or sepsis. However, clinical translation of the use of EVs as biomarkers or potential therapeutic targets may be limited by current methodologies used to identify and quantify EVs accurately (whether from host cells or infecting organisms) and lack of understanding of the role of EVs in the reparative phase during recovery from pneumonia and/or sepsis.
Collapse
Affiliation(s)
- Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s hospital, Catholic College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Masaru Shimizu
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Jae-Woo Lee
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California.,Jae-Woo Lee, MD, Professor, University of California San Francisco, Department of Anesthesiology, 505 Parnassus Ave., Box 0648, San Francisco, CA 94143, Telephone: (415) 476-0452, Fax: (415) 514-2999,
| |
Collapse
|
9
|
Redman CW, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the convergence point for multiple pathways. Am J Obstet Gynecol 2022; 226:S907-S927. [PMID: 33546842 DOI: 10.1016/j.ajog.2020.09.047] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Preeclampsia evolves in 2 stages: a placental problem that generates signals to the mother to cause a range of responses that comprise the second stage (preeclampsia syndrome). The first stage of early-onset preeclampsia is poor placentation, which we here call malplacentation. The spiral arteries are incompletely remodeled, leading to later placental malperfusion, relatively early in the second half of pregnancy. The long duration of the first stage (several months) is unsurprisingly associated with fetal growth restriction. The first stage of late-onset preeclampsia, approximately 80% of total cases, is shorter (several weeks) and part of a process that is common to all pregnancies. Placental function declines as it outgrows uterine capacity, with increasing chorionic villous packing, compression of the intervillous space, and fetal hypoxia, and causes late-onset clinical presentations such as "unexplained" stillbirths, late-onset fetal growth restriction, or preeclampsia. The second stages of early- and late-onset preeclampsia share syncytiotrophoblast stress as the most relevant feature that causes the maternal syndrome. Syncytiotrophoblast stress signals in the maternal circulation are probably the most specific biomarkers for preeclampsia. In addition, soluble fms-like tyrosine kinase-1 (mainly produced by syncytiotrophoblast) is the best-known biomarker and is routinely used in clinical practice in many locations. How the stress signals change over time in normal pregnancies indicates that syncytiotrophoblast stress begins on average at 30 to 32 weeks' gestation and progresses to term. At term, syncytiotrophoblast shows increasing markers of stress, including apoptosis, pyroptosis, autophagy, syncytial knots, and necrosis. We label this phenotype the "twilight placenta" and argue that it accounts for the clinical problems of postmature pregnancies. Senescence as a stress response differs in multinuclear syncytiotrophoblast from that of mononuclear cells. Syncytiotrophoblast irreversibly acquires part of the senescence phenotype (cell cycle arrest) when it is formed by cell fusion. The 2 pathways converge on the common pathologic endpoint, syncytiotrophoblast stress, and contribute to preeclampsia subtypes. We highlight that the well-known heterogeneity of the preeclampsia syndrome arises from different pathways to this common endpoint, influenced by maternal genetics, epigenetics, lifestyle, and environmental factors with different fetal and maternal responses to the ensuing insults. This complexity mandates a reassessment of our approach to predicting and preventing preeclampsia, and we summarize research priorities to maximize what we can learn about these important issues.
Collapse
|
10
|
Caillon A, Trimaille A, Favre J, Jesel L, Morel O, Kauffenstein G. Role of neutrophils, platelets, and extracellular vesicles and their interactions in COVID-19-associated thrombopathy. J Thromb Haemost 2022; 20:17-31. [PMID: 34672094 PMCID: PMC8646423 DOI: 10.1111/jth.15566] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic extended all around the world causing millions of deaths. In addition to acute respiratory distress syndrome, many patients with severe COVID-19 develop thromboembolic complications associated to multiorgan failure and death. Here, we review evidence for the contribution of neutrophils, platelets, and extracellular vesicles (EVs) to the thromboinflammatory process in COVID-19. We discuss how the immune system, influenced by pro-inflammatory molecules, EVs, and neutrophil extracellular traps (NETs), can be caught out in patients with severe outcomes. We highlight how the deficient regulation of the innate immune system favors platelet activation and induces a vicious cycle amplifying an immunothrombogenic environment associated with platelet/NET interactions. In light of these considerations, we discuss potential therapeutic strategies underlining the modulation of purinergic signaling as an interesting target.
Collapse
Affiliation(s)
- Antoine Caillon
- Lady Davis Institute for Medical Research, McGill University, Montréal, Quebec, Canada
| | - Antonin Trimaille
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Julie Favre
- INSERM, UMR S 1121, Biomaterials and Bioengineering, CRBS, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | - Laurence Jesel
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Olivier Morel
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | | |
Collapse
|
11
|
Puhm F, Flamand L, Boilard E. Platelet extracellular vesicles in COVID-19: Potential markers and makers. J Leukoc Biol 2021; 111:63-74. [PMID: 34730839 PMCID: PMC8667644 DOI: 10.1002/jlb.3mir0221-100r] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Platelets and platelet extracellular vesicles (pEV) are at the crossroads of coagulation and immunity. Extracellular vesicles are messengers that not only transmit signals between cells, but also provide information about the status of their cell of origin. Thus, pEVs have potential as both biomarkers of platelet activation and contributors to pathology. Coronavirus Disease‐19 (COVID‐19), caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), is a complex disease affecting multiple organs and is characterized by a high degree of inflammation and risk of thrombosis in some patients. In this review, we introduce pEVs as valuable biomarkers in disease with a special focus on their potential as predictors of and contributors to COVID‐19.
Collapse
Affiliation(s)
- Florian Puhm
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| | - Louis Flamand
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| |
Collapse
|
12
|
Zhang CN, Li FJ, Zhao ZL, Zhang JN. The role of extracellular vesicles in traumatic brain injury-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2021; 321:L885-L891. [PMID: 34549593 DOI: 10.1152/ajplung.00023.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acute lung injury (ALI), a common complication after traumatic brain injury (TBI), can evolve into acute respiratory distress syndrome (ARDS) and has a mortality rate of 30%-40%. Secondary ALI after TBI exhibits the following typical pathological features: infiltration of neutrophils into the alveolar and interstitial space, alveolar septal thickening, alveolar edema, and hemorrhage. Extracellular vesicles (EVs) were recently identified as key mediators in TBI-induced ALI. Due to their small size and lipid bilayer, they can pass through the disrupted blood-brain barrier (BBB) into the peripheral circulation and deliver their contents, such as genetic material and proteins, to target cells through processes such as fusion, receptor-mediated interactions, and uptake. Acting as messengers, EVs contribute to mediating brain-lung cross talk after TBI. In this review, we aim to summarize the mechanism of EVs in TBI-induced ALI, which may provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Chao-Nan Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Fan-Jian Li
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Zi-Long Zhao
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Jian-Ning Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Amiral J. Extra cellular vesicles in blood circulation as biomarkers and messengers of patho-hysiological activity and alterations. Transfus Apher Sci 2021; 60:103209. [PMID: 34244081 DOI: 10.1016/j.transci.2021.103209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is an increasing interest in Extracellular Vesicles released by many cells through membrane shedding. In addition to cell signaling, these particles are true messenger cargos, which can carry cell surface proteins, miRNAs and non-coding RNAs to other and distant cells. They are part of the inter-cellular crosstalk and they contribute to transferring biological messages far away from the triggering event. EVs are biomarkers of many diseases, including thrombo-embolic pathology, infections, neurological or metabolic disorders, and malignancy. Their role and significance are presented and discussed in this short review, as consequences of disease and causes of its progression. But they can also be beneficial for tissue healing or repair, and they can be prepared in vitro to be used for cell- targeted treatments. Many identification and measurement methods for EV's are sophisticated, which restricts their use to research studies, but they have, nevertheless, a high laboratory potential for diagnosis, prognosis and evolution as follow-up of many pathologies. New emerging laboratory tools offer more friendly and easy applications for characterizing EVs and testing their associated activity, especially for the procoagulant ones.
Collapse
Affiliation(s)
- Jean Amiral
- SH/Scientific-Hemostasis, Scientific Director and Consultant in Hemostasis and Thrombosis Diagnostics, Franconville, France.
| |
Collapse
|
14
|
Processing methods and storage duration impact extracellular vesicle counts in red blood cell units. Blood Adv 2021; 4:5527-5539. [PMID: 33166402 DOI: 10.1182/bloodadvances.2020001658] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are active components of red blood cell (RBC) concentrates and may be associated with beneficial and adverse effects of transfusion. Elucidating controllable factors associated with EV release in RBC products is thus important to better manage the quality and properties of RBC units. Erythrocyte-derived EVs (EEVs) and platelet-derived EVs (PEVs) were counted in 1226 RBC units (administered to 280 patients) using a standardized cytometry-based method. EV size and CD47 and annexin V expression were also measured. The effects of donor characteristics, processing methods, and storage duration on EV counts were analyzed by using standard comparison tests, and analysis of covariance was used to determine factors independently associated with EV counts. PEV as well as EEV counts were higher in whole-blood-filtered RBC units compared with RBC-filtered units; PEV counts were associated with filter type (higher with filters associated with higher residual platelets), and CD47 expression was higher on EEVs in RBC units stored longer. Multivariate analysis showed that EEV counts were strongly associated with filter type (P < .0001), preparation, and storage time (+25.4 EEV/µL per day [P = .01] and +42.4 EEV/µL per day [P < .0001], respectively). The only independent factor associated with PEV counts was the residual platelet count in the unit (+67.1 PEV/µL; P < .0001). Overall, processing methods have an impact on EV counts and characteristics, leading to large variations in EV quantities transfused into patients. RBC unit processing methods might be standardized to control the EV content of RBC units if any impacts on patient outcomes can be confirmed. The IMIB (Impact of Microparticles in Blood) study is ancillary to the French ABLE (Age of Transfused Blood in Critically Ill Adults) trial (ISRCTN44878718).
Collapse
|
15
|
Nazari M, Javandoost E, Talebi M, Movassaghpour A, Soleimani M. Platelet Microparticle Controversial Role in Cancer. Adv Pharm Bull 2021; 11:39-55. [PMID: 33747851 PMCID: PMC7961228 DOI: 10.34172/apb.2021.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are a group of micrometer-scale extracellular vesicles released by platelets upon activation that are responsible for the majority of microvesicles found in plasma. PMPs' physiological properties and functions have long been investigated by researchers. In this regard, a noticeable area of studies has been devoted to evaluating the potential roles and effects of PMPs on cancer progression. Clinical and experimental evidence conflictingly implicates supportive and suppressive functions for PMPs regarding cancer. Many of these functions could be deemed as a cornerstone for future considerations of PMPs usage in cancer targeted therapy. This review discusses what is currently known about PMPs and provides insights for new and possible research directions for further grasping the intricate interplay between PMPs and cancer.
Collapse
Affiliation(s)
- Mahnaz Nazari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Javandoost
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. Introduction
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
16
|
Molecular Mechanisms Underlying the Cardiovascular Toxicity of Specific Uremic Solutes. Cells 2020; 9:cells9092024. [PMID: 32887404 PMCID: PMC7565564 DOI: 10.3390/cells9092024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Mounting evidence strongly suggests a causal link between chronic kidney disease (CKD) and cardiovascular disease (CVD). Compared with non-CKD patients, patients with CKD suffer disproportionately from CVD and derive suboptimal benefits from interventions targeting conventional CVD risk factors. Uremic toxins (UTs), whose plasma levels rapidly rise as CKD progresses, represent a unique risk factor in CKD, which has protean manifestations on CVD. Among the known UTs, tryptophan metabolites and trimethylamine N-oxide are well-established cardiovascular toxins. Their molecular mechanisms of effect warrant special consideration to draw translational value. This review surveys current knowledge on the effects of specific UTs on different pathways and cell functions that influence the integrity of cardiovascular health, with implication for CVD progression. The effect of UTs on cardiovascular health is an example of a paradigm in which a cascade of molecular and metabolic events induced by pathology in one organ in turn induces dysfunction in another organ. Deciphering the molecular mechanisms underlying such cross-organ pathologies will help uncover therapeutic targets to improve the management of CVD in patients with CKD.
Collapse
|
17
|
Investigation of Changes in Exosomes Profile During Storage Period of Erythrocyte Suspensions. Indian J Hematol Blood Transfus 2020. [DOI: 10.1007/s12288-020-01336-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
18
|
Relja B, Land WG. Damage-associated molecular patterns in trauma. Eur J Trauma Emerg Surg 2020; 46:751-775. [PMID: 31612270 PMCID: PMC7427761 DOI: 10.1007/s00068-019-01235-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
In 1994, the "danger model" argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of "strange" molecules. Thus, an alternative to the "self versus non-self recognition model" has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their "friend or foe" role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Collapse
Affiliation(s)
- Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt, Germany.
| | - Walter Gottlieb Land
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
19
|
Zhang Z, Xiao C, Yong T, Yang X, Gan L, Li Z. Cellular microparticles for tumor targeting delivery: from bench to bedside. Chem Commun (Camb) 2020; 56:6171-6188. [PMID: 32478343 DOI: 10.1039/d0cc02333g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Within the past few years, cell derived microparticles (MPs) have emerged as a highly potent natural drug delivery system for tumor therapy. We and others have engineered different cells to obtain eleven kinds of MPs, which efficiently delivered antitumor agents to recipient cells and achieved ideal treatment outcomes in a great number of rodent tumor models. More significantly, autologous tumor cell derived MPs packaging chemotherapeutic drugs were demonstrated to be safe and tolerable and accomplished decent objective clinical response in lung cancer patients in clinical settings, leading to their approval as a novel biotherapy for treating malignancies in China. In this feature article, we review MP mediated tumor targeting delivery. The impact of tumor cell derived MPs on tumor progression and metastasis will be briefly summarized. Different means to prepare, label and characterize MPs will be introduced. Advantages and limitations of distinctive cargo encapsulation strategies will be outlined and compared. Tumor cell MP mediated in vivo transport processes will be reviewed comprehensively. Representative MPs shed by platelets, endothelial cells, macrophages, and dendritic cells will be selected to showcase their advantages in tumor targeting delivery. Multifarious therapeutic agents, including chemotherapeutic drugs, oncolytic adenovirus, nucleic acids, antigens, antibodies, to name a few, have been transported to their targets for corresponding cancer therapies. Current challenges and future opportunities on translating MPs for tumor targeting delivery will be provided in the end.
Collapse
Affiliation(s)
- Zhijie Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China.
| | - Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China.
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China. and Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China and Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China. and Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China and Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China. and Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China and Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
| |
Collapse
|
20
|
Renaud-Picard B, Vallière K, Toussaint J, Kreutter G, El-Habhab A, Kassem M, El-Ghazouani F, Olland A, Hirschi S, Porzio M, Chenard MP, Toti F, Kessler L, Kessler R. Epithelial-mesenchymal transition and membrane microparticles: Potential implications for bronchiolitis obliterans syndrome after lung transplantation. Transpl Immunol 2020; 59:101273. [PMID: 32097721 DOI: 10.1016/j.trim.2020.101273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Long term survival post lung transplantation (LTx) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). One mechanism involved is the epithelial-mesenchymal transition (EMT). Membrane microparticles (MPs) are known to be involved in some respiratory diseases and in other organs allograft rejection episodes. We hypothesized that leukocyte-derived MPs likely contribute to EMT. To emphasize this physiological concept, our objectives were to: (1) confirm the presence of EMT on explanted lungs from patients who underwent a second LTx for BOS; 2) characterize circulating MPs in transplanted patients, with or without BOS; (3) evaluate in vitro the effect of monocyte-derived MPs in EMT of human bronchial epithelial cells. Our IHC analysis on explanted graft lungs revealed significant pathological signs of EMT with an inhomogeneous destruction of the bronchial epithelium, with decreased expression of the epithelial protein E-cadherin and increased expression of the mesenchymal protein Vimentin. The immunophenotyping of MPs demonstrated that the concentration of MPs carrying E-cadherin was lower in patients affected by BOS (p = .007). In vitro, monocyte-derived MPs produced with LPS were associated with decreased E-cadherin expression (p < .05) along with significant morphological and functional cell modifications. MPs may play a role in EMT onset in bronchial epithelium following LTx.
Collapse
Affiliation(s)
- Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France.
| | - Kevin Vallière
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Justine Toussaint
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Guillaume Kreutter
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Ali El-Habhab
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Mohamad Kassem
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Fatiha El-Ghazouani
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France; Department of Thoracic Surgery and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Sandrine Hirschi
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Michele Porzio
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | | | - Florence Toti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Laurence Kessler
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| | - Romain Kessler
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| |
Collapse
|
21
|
Xu B, Ma L, Zhang N, Guo W, Luo LM, Wang C, Jiang Y, Liu LG. Increased microparticle levels in middle-aged and elderly patients with insomnia may be involved in the pathogenesis of arteriosclerosis. INT ANGIOL 2020; 39:252-260. [PMID: 32052947 DOI: 10.23736/s0392-9590.19.04261-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Insomnia may affect vascular factors and promote arteriosclerosis. Microparticles (MPs) are a heterogeneous group of bioactive small vesicles that can be found in blood and body fluids following activation, necrosis or apoptosis of virtually any eukaryotic cells. MPs are believed to participate in the pathogenesis of atherosclerosis. Few studies have been concerned with the microparticle level in patients with sleep disorder. The purpose of the present study is to measure the levels of endothelial microparticles (EMPs), platelet microparticles (PMPs) and leukocyte-derived microparticles (LMPs) in middle-aged and elderly patients with or without insomnia. METHODS Patients with insomnia (N.=30) and without insomnia (N.=18) were enrolled. The insomnia group covered patients with chronic insomnia (N.=16) and acute insomnia (N.=14). Levels of EMPs (CD31 +, CD62E +) and PMPs (CD41a +, CD42a +) and granulocyte-derived (CD11a +) MPs were measured. Flow cytometry was performed on the Beckman Coulter analyzer. Reference gate was defined for the level of MPs using 0.22-0.45-0.88μm microspheres, and the size gate for MPs was 0.5-1.0μm. RESULTS Of all types of MPs detected, the levels of CD31 +MPs, CD62E +MPs and CD11a +MPs were significantly higher in the insomnia group than in the non-insomnia group (P<0.05). Besides, compared with acute insomnia, the levels of CD31 + MPs and CD11a +MPs were significantly higher in chronic insomnia (P<0.001). CONCLUSIONS In insomnia patients, atherosclerosis progression may be increased by the CD31+ EMPs-mediated apoptosis and endothelial injury. The level of CD11a+ LMPs kept increasing as insomnia persisted, which may indicate atherosclerosis progression.
Collapse
Affiliation(s)
- Bin Xu
- Department of General Practice, School of General Practice and Continuing Education, Capital Medical University, Beijing, China.,Department of Emergency, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Li Ma
- Department of General Practice, School of General Practice and Continuing Education, Capital Medical University, Beijing, China.,Department of General Practice, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Internal Medicine-Neurology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Wei Guo
- Department of Emergency, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Li-Ming Luo
- Puhuangyu Community Service Center, Fengtai District, Beijing, China
| | - Chen Wang
- Department of General Practice, School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| | - Yue Jiang
- Department of General Practice, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Li-Ge Liu
- Department of General Practice, School of General Practice and Continuing Education, Capital Medical University, Beijing, China - .,Department of Internal Medicine, Health Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Zacharia E, Zacharias K, Papamikroulis GA, Bertsias D, Miliou A, Pallantza Z, Papageorgiou N, Tousoulis D. Cell-Derived Microparticles and Acute Coronary Syndromes: Is there a Predictive Role for Microparticles? Curr Med Chem 2020; 27:4440-4468. [PMID: 31838988 DOI: 10.2174/0929867327666191213104841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 09/09/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Despite the recent advances in the treatment of Acute Coronary Syndromes (ACS), patients with ACS are still exposed to an increased risk for adverse cardiovascular events, while their prognosis is difficult to determine. Experimental and clinical studies have shown that cell-derived Microparticles (MPs) are associated with the underlying pathophysiological processes that are responsible for atherogenesis and may be causally implicated in the induction of atherothrombosis. OBJECTIVE In the present article, we aimed to review the available evidence regarding the predictive role of MPs in patients with ACS. RESULTS Evidence suggests that endothelial MPs are associated with future adverse cardiovascular events in patients with ACS. Platelet-derived MPs have been excessively studied, since they have been found to trigger the coagulation cascade; however, their role as predictors of future cardiovascular events remains debatable. The role of red blood cell-derived MPs is more intriguing; they have been proposed as markers of ongoing thrombosis in patients with ACS, while previous studies have shown that they have anti-coagulant properties in healthy individuals. Leukocyte-derived MPs may also have a predictive role, although the studies regarding these are still limited. Last but not least, it was an interesting discovery that circulating MPs can provide information regarding the angiographic lesions in patients with ACS. CONCLUSION The concept of MPs as potential circulating biomarkers in patients with ACS holds much promise. However, large-scale clinical studies are required to evaluate whether the measurement of plasma MPs could be of clinical significance and, thus, dictate a more aggressive treatment strategy in patients with high levels of circulating MPs.
Collapse
Affiliation(s)
- Effimia Zacharia
- 1st Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | | | | | | | - Antigoni Miliou
- 1st Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | - Zoi Pallantza
- 1st Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | | | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| |
Collapse
|
23
|
Quaglia M, Dellepiane S, Guglielmetti G, Merlotti G, Castellano G, Cantaluppi V. Extracellular Vesicles as Mediators of Cellular Crosstalk Between Immune System and Kidney Graft. Front Immunol 2020; 11:74. [PMID: 32180768 PMCID: PMC7057849 DOI: 10.3389/fimmu.2020.00074] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are known immune-modulators exerting a critical role in kidney transplantation (KT). EV bioactive cargo includes graft antigens, costimulatory/inhibitory molecules, cytokines, growth factors, and functional microRNAs (miRNAs) that may modulate expression of recipient cell genes. As paracrine factors, neutrophil- and macrophage-derived EVs exert immunosuppressive and immune-stimulating effects on dendritic cells, respectively. Dendritic cell-derived EVs mediate alloantigen spreading and modulate antigen presentation to T lymphocytes. At systemic level, EVs exert pleiotropic effects on complement and coagulation. Depending on their biogenesis, they can amplify complement activation or shed complement inhibitors and prevent cell lysis. Likewise, endothelial- and platelet-derived EVs can exert procoagulant/prothrombotic effects and also promote endothelial survival and angiogenesis after ischemic injury. Kidney endothelial- and tubular-derived EVs play a key role in ischemia-reperfusion injury (IRI) and during the healing process; additionally, they can trigger rejection by inducing both alloimmune and autoimmune responses. Endothelial EVs have procoagulant/pro-inflammatory effects and can release sequestered self-antigens, generating a tissue-specific autoimmunity. Renal tubule-derived EVs shuttle pro-fibrotic mediators (TGF-β and miR-21) to interstitial fibroblasts and modulate neutrophil and T-lymphocyte influx. These processes can lead to peritubular capillary rarefaction and interstitial fibrosis-tubular atrophy. Different EVs, including those from mesenchymal stromal cells (MSCs), have been employed as a therapeutic tool in experimental models of rejection and IRI. These particles protect tubular and endothelial cells (by inhibition of apoptosis and inflammation-fibrogenesis or by inducing autophagy) and stimulate tissue regeneration (by triggering angiogenesis, cell proliferation, and migration). Finally, urinary and serum EVs represent potential biomarkers for delayed graft function (DGF) and acute rejection. In conclusion, EVs sustain an intricate crosstalk between graft tissue and innate/adaptive immune systems. EVs play a major role in allorecognition, IRI, autoimmunity, and alloimmunity and are promising as biomarkers and therapeutic tools in KT.
Collapse
Affiliation(s)
- Marco Quaglia
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Sergio Dellepiane
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
- Division of Hematology/Medical Oncology, Icahn School of Medicine at Mount Sinai Hospital, The Tisch Cancer Institute, New York, NY, United States
| | - Gabriele Guglielmetti
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Guido Merlotti
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Giuseppe Castellano
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
- *Correspondence: Vincenzo Cantaluppi
| |
Collapse
|
24
|
Ability of Fibrin Monomers to Predict Progressive Hemorrhagic Injury in Patients with Severe Traumatic Brain Injury. Neurocrit Care 2019; 33:182-195. [PMID: 31797276 DOI: 10.1007/s12028-019-00882-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Progressive hemorrhagic injury (PHI) is common in patients with severe traumatic brain injury (TBI) and is associated with poor outcomes. TBI-associated coagulopathy is frequent and has been described as risk factor for PHI. This coagulopathy is a dynamic process involving hypercoagulable and hypocoagulable states either one after the other either concomitant. Fibrin monomers (FMs) are a direct marker of thrombin action and thus reflect coagulation activation. This study sought to determine the ability of FM to predict PHI after severe TBI. METHODS We conducted a prospective, observational study including all severe TBI patients admitted in the trauma center. Between September 2011 and September 2016, we enrolled patients with severe TBI into the derivation cohort. Between October 2016 and December 2018, we recruited the validation cohort on the same basis. Study protocol included FM measurements and standard coagulation test at admission and two computed tomography (CT) scans (upon arrival and at least 6 h thereafter). A PHI was defined by an increment in size of initial lesion (25% or more) or the development of a new hemorrhage in the follow-up CT scan. Multivariate logistic regression analysis was applied to identify predictors of PHI. RESULTS Overall, 106 patients were included in the derivation cohort. Fifty-four (50.9%) experienced PHI. FM values were higher in these patients (151 [136.8-151] vs. 120.5 [53.3-151], p < 0.0001). The ROC curve demonstrated that FM had a fair accuracy to predict the occurrence of PHI with an area under curve of 0.7 (95% CI [0.6-0.79]). The best threshold was determined at 131.7 μg/ml. In the validation cohort of 54 patients, this threshold had a negative predictive value of 94% (95% CI [71-100]) and a positive predictive value of 49% (95% CI [32-66]). The multivariate logistic regression analysis identified 2 parameters associated with PHI: FM ≥ 131.7 (OR 6.8; 95% CI [2.8-18.1]) and Marshall category (OR 1.7; 95% CI [1.3-2.2]). Coagulopathy was not associated with PHI (OR 1.3; 95% CI [0.5-3.0]). The proportion of patients with an unfavorable functional neurologic outcome at 6-months follow-up was higher in patients with positive FM: 59 (62.1%) versus 16 (29.1%), p < 0.0001. CONCLUSIONS FM levels at admission had a fair accuracy to predict PHI in patients with severe TBI. FM values ≥ 131.7 μg/ml are independently associated with the occurrence of PHI.
Collapse
|
25
|
Ageing enhances the shedding of splenocyte microvesicles with endothelial pro-senescent effect that is prevented by a short-term intake of omega-3 PUFA EPA:DHA 6:1. Biochem Pharmacol 2019; 173:113734. [PMID: 31811867 DOI: 10.1016/j.bcp.2019.113734] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/25/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Ageing is associated with progressive endothelial senescence and dysfunction, and cardiovascular risk. Circulating endothelial microvesicles (MVs) are pro-senescent and pro-inflammatory endothelial effectors in acute coronary syndrome. Omega-3 PUFA intake was claimed beneficial in cardiovascular prevention. PURPOSE To investigate whether the intake of the omega-3 formulation EPA:DHA 6:1 by middle-aged and old rats reduces the shedding of pro-senescent microvesicles from cultured spleen leukocytes (SMVs) and clarify the underlying mechanisms in target coronary primary endothelial cells (ECs). METHODS Middle-aged male Wistar rats (M, 48-week old) received 500 mg/kg/d of either EPA:DHA 6:1, EPA:DHA 1:1, or vehicle (CTL) for 7 days, old rats (72-week old) for 14 days. Spleen-derived leukocytes were prepared and cultured for 24 h and MVs collected from supernatants (SMVs). Cultured ECs were prepared from freshly isolated porcine coronary arteries. Senescence-associated β-galactosidase activity (SA-β-gal) was assessed by C12FDG, protein expression by Western blot analysis, oxidative stress by dihydroethidium using confocal microscopy, and procoagulant MVs by prothrombinase assay. The pro-senescent potential of SMVs from middle-aged rats (M-SMVs) was analyzed by comparison with young (Y, 12-week) and old (O) rats. RESULTS The shedding of SMVs significantly increased with age and was inhibited by EPA:DHA 6:1 intake that also prevented ROS accumulation in spleen. Incubation of ECs with 10 nM SMVs from middle-aged and old but not those from young rats induced premature senescence after 48 h. The pro-senescent effect of M-SMVs was prevented by Losartan and associated with endothelial oxidative stress. M-SMVs induced an up-regulation of senescence markers (p16, p21, p53), pro-atherothrombotic (VCAM-1, ICAM-1, tissue factor) and pro-inflammatory markers (pNF-κB, COX-2) and proteins of the angiotensin system (ACE, AT1-R). Conversely, endothelial NO synthase was down-regulated. Intake of EPA:DHA 1:1 and 6:1 by middle-aged rats decreased SMV shedding by 14% and 24%, respectively. Only EPA:DHA 6:1 intake abolished the M-SMVs-induced endothelial senescence and reduced the pro-senescent action of O-SMVs by 45%. Protection of ECs was not observed in response to SMVs from EPA:DHA 1:1 treated rats. CONCLUSION Ingestion of EPA:DHA 6:1 by middle-aged or old rats, respectively abolished or limited both the shedding of SMVs and their pro-senescent, pro-thrombotic and pro-inflammatory effects in ECs, most likely by triggering the local angiotensin system. EPA:DHA 6:1 may help to delay ageing-related endothelial dysfunction.
Collapse
|
26
|
Molecular Targets of Aspirin and Prevention of Preeclampsia and Their Potential Association with Circulating Extracellular Vesicles during Pregnancy. Int J Mol Sci 2019; 20:ijms20184370. [PMID: 31492014 PMCID: PMC6769718 DOI: 10.3390/ijms20184370] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/30/2019] [Accepted: 08/26/2019] [Indexed: 12/26/2022] Open
Abstract
Uncomplicated healthy pregnancy is the outcome of successful fertilization, implantation of embryos, trophoblast development and adequate placentation. Any deviation in these cascades of events may lead to complicated pregnancies such as preeclampsia (PE). The current incidence of PE is 2–8% in all pregnancies worldwide, leading to high maternal as well as perinatal mortality and morbidity rates. A number of randomized controlled clinical trials observed the association between low dose aspirin (LDA) treatment in early gestational age and significant reduction of early onset of PE in high-risk pregnant women. However, a substantial knowledge gap exists in identifying the particular mechanism of action of aspirin on placental function. It is already established that the placental-derived exosomes (PdE) are present in the maternal circulation from 6 weeks of gestation, and exosomes contain bioactive molecules such as proteins, lipids and RNA that are a “fingerprint” of their originating cells. Interestingly, levels of exosomes are higher in PE compared to normal pregnancies, and changes in the level of PdE during the first trimester may be used to classify women at risk for developing PE. The aim of this review is to discuss the mechanisms of action of LDA on placental and maternal physiological systems including the role of PdE in these phenomena. This review article will contribute to the in-depth understanding of LDA-induced PE prevention.
Collapse
|
27
|
Eguchi A, Franz N, Kobayashi Y, Iwasa M, Wagner N, Hildebrand F, Takei Y, Marzi I, Relja B. Circulating Extracellular Vesicles and Their miR "Barcode" Differentiate Alcohol Drinkers With Liver Injury and Those Without Liver Injury in Severe Trauma Patients. Front Med (Lausanne) 2019; 6:30. [PMID: 30859103 PMCID: PMC6397866 DOI: 10.3389/fmed.2019.00030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 02/01/2019] [Indexed: 12/16/2022] Open
Abstract
Short Summary: Extracellular vesicles (EVs), released during tissue/cell injury, contain a “barcode” indicating specific microRNAs (miRs) that can uncover their origin. We examined whether systemic EVs possessing hepatic miR-signatures would indicate ongoing liver injury and clinical complications in trauma patients (TP). We grouped the patients of alcoholic drinkers into “alcohol-drinkers with liver injury (LI)” (EtOH with LI) or “alcohol-drinkers without LI” (EtOH w/o LI) and we compared these groups to “non-drinkers” (no EtOH). When we examined patient blood from the EtOH with LI group we found the total number of EVs to be increased, along with an increase in miR-122 and let7f—two EV-associated miRNAs—and several inflammation-associating cytokines, such as interleukin (IL)-6 and IL-33. In contrast, all of the aforementioned readouts were found to be decreased in the EtOH w/o LI group. These novel data demonstrate that hepatocyte damage in alcohol-intoxicated trauma patients presenting with liver injury can be reflected by an increase in circulating serum EVs, their specific miR-“barcode” and the concomitant increase of systemic inflammatory markers IL-6 and IL-33. Anti-inflammatory effect of alcohol-drinking in EtOH w/o LI can be presented by a reduced number of hepato-derived EVs, no upregulation of IL-6 and IL-33, and a miR “barcode” different from patients presenting with liver injury. Background: Alcohol abuse is associated with (neuro)protective effects related to (head) injuries, and with negative effects regarding infection rates and survival in severely injured trauma patients (TP). Extracellular vesicles (EVs), which are released during tissue and/or cell injury, can contain a “barcode” including specific microRNAs (miRs) that uncover their origin. We examined whether EVs with a hepatic miR signature can be systemically measured, and whether they can indicate ongoing liver injury in alcohol-intoxicated TP and foretell clinical complications. Patients/Methods: We enrolled 35 TP and measured blood EVs, IL-6, TNF-alpha, IL-1beta, IL-10 and IL-33, alcohol (ethanol, EtOH) concentration (BAC), GLDH, GGT, AST, ALT, leukocytes, platelets, and bilirubin. Within circulating EVs we measured the expression levels of miR-122, let7f, miR21, miR29a, miR-155, and miR-146a. Patients of alcohol-drinkers were grouped into “alcohol drinkers with liver injury (LI)” (EtOH with LI) or “alcohol drinkers without LI” (EtOH w/o LI) and compared to “non-drinkers” (no EtOH). We assessed systemic injury characteristics and the outcome of hospitalization with regard to sepsis, septic shock, pneumonia, or mortality. Results: EtOH with LI patients had significantly increased rates of pneumonia vs. the EtOH w/o LI group. EVs, IL-6, and IL-33 levels were significantly increased in EtOH with LI vs. EtOH w/o LI group (p < 0.05). EV number correlated positively with ALT and IL-6 (p < 0.0001). Two miRs, miR-122 and let7f, were increased only in the blood EVs from the EtOH with LI group (p < 0.05). Five miRs, miR-122, let7f, miR-21, miR-29a, and miR-146a, were reduced in the blood EVs from the EtOH w/o LI group, vs. no EtOH (p < 0.05). Notably miR-122 correlated significantly with increased bilirubin levels in the EtOH with LI group (p < 0.05). Conclusions: Liver injury in alcohol-intoxicated TP is reflected by increased EV numbers, their specific miR barcode, and the correlated increase of systemic inflammatory markers IL-6 and IL-33. Interestingly, severely injured TP without liver injury were found to have a reduced number of liver-derived EVs, no observed inflammatory infiltration and reduced specific miR “barcode.”
Collapse
Affiliation(s)
- Akiko Eguchi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan.,JST, PRESTO, Saitama, Japan
| | - Niklas Franz
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Yoshinao Kobayashi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Nils Wagner
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Frank Hildebrand
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
28
|
Fogli S, Neri T, Nuti E, Mattii L, Camodeca C, Rossello A. Matrix metalloproteinase inhibitors prevent the release and proteolytic activity of monocyte/macrophage-derived microparticles. Pharmacol Rep 2019; 71:485-490. [PMID: 31005034 DOI: 10.1016/j.pharep.2019.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/10/2019] [Accepted: 01/30/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The role of monocyte/macrophage-derived microparticles (MPs) in the pathophysiology of cancer and chronic inflammatory diseases has been reported; nevertheless, the mechanism underlying microparticles release is currently unclear. The aim of the current study was to investigate whether matrix metalloproteinase (MMP) inhibitors could prevent MP shedding from stimulated human monocyte/macrophage. METHODS Microparticles were obtained by isolated peripheral blood mononuclear cells after stimulation with the calcium ionophore, A23187. MP shedding, intracellular calcium concentration, analysis of RhoA expression, and proteolytic activities of isolated MPs were assessed in the absence or presence of MMP inhibitors. RESULTS We demonstrated that MMP inhibitors remarkably prevented MP shedding in a concentration-dependent manner with IC50 values in the nano- to micromolar range. Such an effect was related to their ability to reduce the intracellular Ca2+ levels induced by the calcium ionophore and the consequent translocation of RhoA from cytosol to membrane. Furthermore, MMP inhibitors could inhibit the proteolytic activity of cell-derived MPs. CONCLUSIONS The current study provide evidence that MMP inhibitors can prevent MPs shedding from stimulated human monocyte/macrophage and the proteolytic activity of released MPs. Finally, the most active compound tested might represent the lead compound of a new class of molecules with therapeutic potential in cancer and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Stefano Fogli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Tommaso Neri
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Letizia Mattii
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
29
|
Afonyushkin T, Binder CJ. Extracellular Vesicles Act as Messengers of Macrophages Sensing Atherogenic Stimuli. Arterioscler Thromb Vasc Biol 2018; 38:2-3. [PMID: 29282243 DOI: 10.1161/atvbaha.117.310257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Taras Afonyushkin
- From the Department of Laboratory Medicine, Medical University of Vienna, Austria; and Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph J Binder
- From the Department of Laboratory Medicine, Medical University of Vienna, Austria; and Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
30
|
Iba T, Ogura H. Role of extracellular vesicles in the development of sepsis-induced coagulopathy. J Intensive Care 2018; 6:68. [PMID: 30377532 PMCID: PMC6194680 DOI: 10.1186/s40560-018-0340-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background The advances of research on extracellular vesicles (EVs) are of particular interest to the clinicians as well as the researchers who are studying coagulation disorder in sepsis. Here, we intend to update the latest knowledge and currently unsolved problems that should be addressed. Main body Secreted membrane-enclosed vesicles including apoptotic bodies, exosomes, ectosomes, microvesicles, and microparticles are generically called EVs. Though the basic structure of these vesicles is the same, i.e., originating from the plasma membrane, their characteristics differ significantly depending on their surface structures and interior components. Numerous studies have shown elevated levels of circulating EVs that exhibit proinflammatory and procoagulant properties during sepsis. These EVs are known to play important roles in the development of coagulation disorder and organ dysfunction in sepsis. Coagulation disorder in sepsis is characterized by activated coagulation, disrupted anticoagulant systems, and imbalanced fibrinolytic systems. These processes collaborate with one another and contribute to the development of disseminated intravascular coagulation (DIC), with devastating consequences. As part of this pathogenesis, the membrane-exposed tissue factor, phosphatidylserine and bioactive substances contained within the vesicles, such as histones, nucleosomes, and high-mobility group box 1, contribute to the development of DIC. EVs not only upregulate the procoagulant systems by themselves, but they also disseminate prothrombotic activities by transferring their procoagulant properties to distant target cells. Though the basic concept behind the role of procoagulant properties, EVs in the development of sepsis-induced coagulopathy has started to be unveiled, knowledge of the actual status is far from satisfactory, mainly because of the lack of standardized assay procedures. Recent advances and current problems that remain to be resolved are introduced in this review. Conclusion The recent studies succeeded to elucidate the important roles of EVs in the progress of coagulation disorder in sepsis. However, further harmonization in terminology, methodology, and evaluation methods is required for future studies.
Collapse
Affiliation(s)
- Toshiaki Iba
- 1Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Hiroshi Ogura
- 2Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
31
|
Record M, Silvente-Poirot S, Poirot M, Wakelam MJO. Extracellular vesicles: lipids as key components of their biogenesis and functions. J Lipid Res 2018; 59:1316-1324. [PMID: 29764923 PMCID: PMC6071772 DOI: 10.1194/jlr.e086173] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Intercellular communication has been known for decades to involve either direct contact between cells or to operate via circulating molecules, such as cytokines, growth factors, or lipid mediators. During the last decade, we have begun to appreciate the increasing importance of intercellular communication mediated by extracellular vesicles released by viable cells either from plasma membrane shedding (microvesicles, also named microparticles) or from an intracellular compartment (exosomes). Exosomes and microvesicles circulate in all biological fluids and can trigger biological responses at a distance. Their effects include a large variety of biological processes, such as immune surveillance, modification of tumor microenvironment, or regulation of inflammation. Extracellular vesicles can carry a large array of active molecules, including lipid mediators, such as eicosanoids, proteins, and nucleic acids, able to modify the phenotype of receiving cells. This review will highlight the role of the various lipidic pathways involved in the biogenesis and functions of microvesicles and exosomes.
Collapse
Affiliation(s)
- Michel Record
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | - Sandrine Silvente-Poirot
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | - Marc Poirot
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | | |
Collapse
|
32
|
Munrós J, Martínez-Zamora MA, Tàssies D, Reverter JC, Rius M, Gracia M, Ros C, Carmona F. Total Circulating Microparticle Levels After Laparoscopic Surgical Treatment for Endometrioma: A Pilot, Prospective, Randomized Study Comparing Stripping with CO 2 Laser Vaporization. J Minim Invasive Gynecol 2018; 26:450-455. [PMID: 29803597 DOI: 10.1016/j.jmig.2018.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/10/2018] [Accepted: 05/17/2018] [Indexed: 01/17/2023]
Abstract
STUDY OBJECTIVE To evaluate serial generation of microparticles (MPs) after laparoscopic stripping or CO2 laser vaporization in the surgical treatment of patients with ovarian endometrioma (OE). DESIGN A prospective, randomized, blinded, pilot study (Canadian Task Force classification I). SETTING Tertiary care university hospital from December 2014 to July 2016. PATIENTS Thirty women with unilateral OE undergoing laparoscopic surgery. INTERVENTION Patients were randomly selected to undergo either CO2 laser vaporization (L group) or laparoscopic stripping (S group) of OE. MEASUREMENTS AND MAIN RESULTS Blood samples were collected before surgery and at 2 hours, 24 hours, 1 month, and 3 months after surgery. An MP generation curve after OE surgery was created. MP generation was greater in the S group than in the L group at all time points evaluated. The MP generation curve showed a significantly higher area under the curve after excisional surgery (p <.05). CONCLUSION The higher MP levels in the S group suggest an increased inflammation and procoagulant response after this procedure.
Collapse
Affiliation(s)
- Jordina Munrós
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria-Angeles Martínez-Zamora
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Dolors Tàssies
- Department of Hemotherapy and Hemostasis, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Juan Carlos Reverter
- Department of Hemotherapy and Hemostasis, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Mariona Rius
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Meritxell Gracia
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina Ros
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Carmona
- Department of Gynecology, Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic of Barcelona, Faculty of Medicine, University of Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
33
|
Chang YH, Wu KC, Harn HJ, Lin SZ, Ding DC. Exosomes and Stem Cells in Degenerative Disease Diagnosis and Therapy. Cell Transplant 2018; 27:349-363. [PMID: 29692195 PMCID: PMC6038041 DOI: 10.1177/0963689717723636] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stroke can cause death and disability, resulting in a huge burden on society. Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor dysfunction. Osteoarthritis (OA) is a progressive degenerative joint disease characterized by cartilage destruction and osteophyte formation in the joints. Stem cell therapy may provide a biological treatment alternative to traditional pharmacological therapy. Mesenchymal stem cells (MSCs) are preferred because of their differentiation ability and possible derivation from many adult tissues. In addition, the paracrine effects of MSCs play crucial anti-inflammatory and immunosuppressive roles in immune cells. Extracellular vesicles (EVs) are vital mediators of cell-to-cell communication. Exosomes contain various molecules such as microRNA (miRNA), which mediates biological functions through gene regulation. Therefore, exosomes carrying miRNA or other molecules can enhance the therapeutic effects of MSC transplantation. MSC-derived exosomes have been investigated in various animal models representing stroke, PD, and OA. Exosomes are a subtype of EVs. This review article focuses on the mechanism and therapeutic potential of MSC-derived exosomes in stroke, PD, and OA in basic and clinical aspects.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- 1 Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.,2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Kung-Chi Wu
- 3 Department of Orthopedics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Horng-Jyh Harn
- 4 Department of Pathology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shinn-Zong Lin
- 5 Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Dah-Ching Ding
- 2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,6 Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
34
|
Karasu E, Eisenhardt SU, Harant J, Huber-Lang M. Extracellular Vesicles: Packages Sent With Complement. Front Immunol 2018; 9:721. [PMID: 29696020 PMCID: PMC5904200 DOI: 10.3389/fimmu.2018.00721] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/23/2018] [Indexed: 12/20/2022] Open
Abstract
Cells communicate with other cells in their microenvironment by transferring lipids, peptides, RNA, and sugars in extracellular vesicles (EVs), thereby also influencing recipient cell functions. Several studies indicate that these vesicles are involved in a variety of critical cellular processes including immune, metabolic, and coagulatory responses and are thereby associated with several inflammatory diseases. Furthermore, EVs also possess anti-inflammatory properties and contribute to immune regulation, thus encouraging an emerging interest in investigating and clarifying mechanistic links between EVs and innate immunity. Current studies indicate complex interactions of the complement system with EVs, with a dramatic influence on local and systemic inflammation. During inflammatory conditions with highly activated complement, including after severe tissue trauma and during sepsis, elevated numbers of EVs were found in the circulation of patients. There is increasing evidence that these shed vesicles contain key complement factors as well as complement regulators on their surface, affecting inflammation and the course of disease. Taken together, interaction of EVs regulates complement activity and contributes to the pro- and anti-inflammatory immune balance. However, the molecular mechanisms behind this interaction remain elusive and require further investigation. The aim of this review is to summarize the limited current knowledge on the crosstalk between complement and EVs. A further aspect is the clinical relevance of EVs with an emphasis on their capacity as potential therapeutic vehicles in the field of translational medicine.
Collapse
Affiliation(s)
- Ebru Karasu
- Institute of Clinical and Experimental Trauma-Immunology, Universitätsklinikum Ulm, Ulm, Germany
| | - Steffen U Eisenhardt
- Division of Reconstructive Microsurgery, Department of Plastic and Hand Surgery, University of Freiburg Faculty of Medicine, University of Freiburg Medical Centre, Freiburg, Germany
| | - Julia Harant
- Institute of Clinical and Experimental Trauma-Immunology, Universitätsklinikum Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Universitätsklinikum Ulm, Ulm, Germany
| |
Collapse
|
35
|
Burbano C, Villar-Vesga J, Orejuela J, Muñoz C, Vanegas A, Vásquez G, Rojas M, Castaño D. Potential Involvement of Platelet-Derived Microparticles and Microparticles Forming Immune Complexes during Monocyte Activation in Patients with Systemic Lupus Erythematosus. Front Immunol 2018; 9:322. [PMID: 29545790 PMCID: PMC5837989 DOI: 10.3389/fimmu.2018.00322] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/05/2018] [Indexed: 11/13/2022] Open
Abstract
Microparticles (MPs) are vesicles derived from the plasma membrane of different cells, are considered a source of circulating autoantigens, and can form immune complexes (MPs-ICs). The number of MPs and MPs-ICs increases in patients with systemic lupus erythematosus (SLE). MPs activate myeloid cells by inducing IL-6 and TNF-α in both SLE and other diseases. Therefore, we propose that the recognition of MPs-ICs by monocytes rather that MPs may define their phenotype and contribute to the inflammatory process in patients with SLE. Thus, the aims of this study were to evaluate the association among circulating MPs-ICs from different cell sources, alterations observed in monocyte subsets, and disease activity in patients with SLE and to establish whether monocytes bind and respond to MPs-ICs in vitro. Circulating MPs and monocyte subsets were characterized in 60 patients with SLE and 60 healthy controls (HCs) using multiparametric flow cytometry. Patients had higher MP counts and frequencies of MPs-CD41a + (platelet-derived) compared with HCs, regardless of disease activity. MPs from patients with SLE were C1q + and formed ICs with IgM and IgG. MPs-IgG + were positively correlated with active SLE (aSLE), whereas MPs-IgM + were negatively correlated. Most of the circulating total ICs-IgG + were located on MPs. The proportion and number of non-classical monocytes were significantly decreased in patients with SLE compared with HCs and in patients with aSLE compared with patients with the inactive disease. Non-classical monocytes obtained from patients with SLE exhibited increased levels of CD64 associated with MPs-IgG +, MPs-C1q +, total circulating ICs-IgG +, and disease activity. The direct effects of MPs and MPs-IgG + on monocytes were evaluated in cell culture. Monocytes from both HCs and patients bound to and internalized MPs and MPs-IgG + independent of CD64. These vesicles derived from platelets (PMPs), mainly PMPs-IgG +, activated monocytes in vitro and increased the expression of CD69, CD64, and pro-inflammatory cytokines such as IL-1β, TNF-α, and IFN-α. Therefore, MPs are one of the most representative sources of the total amount of circulating ICs-IgG + in patients with SLE. MPs-IgG + are associated with SLE activity, and PMPs-IgG + stimulate monocytes, changing their phenotype and promoting pro-inflammatory responses related to disease activity.
Collapse
Affiliation(s)
- Catalina Burbano
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Unidad de Citometría de Flujo, Sede de Investigación Universitaria, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Juan Villar-Vesga
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Janine Orejuela
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Carlos Muñoz
- Sección de Reumatología, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Adriana Vanegas
- Sección de Reumatología, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Gloria Vásquez
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Unidad de Citometría de Flujo, Sede de Investigación Universitaria, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Diana Castaño
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
36
|
Microparticles produced by human papillomavirus type 16 E7-expressing cells impair antigen presenting cell function and the cytotoxic T cell response. Sci Rep 2018; 8:2373. [PMID: 29402982 PMCID: PMC5799164 DOI: 10.1038/s41598-018-20779-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/23/2018] [Indexed: 01/08/2023] Open
Abstract
High-risk, cancer-causing human papillomaviruses (HPV) cause infections of the epidermis that may progress to cancer, including cervical cancer. Viral persistence, contributed to by viral evasion of the host immune response, is associated with the likelihood of cancer developing. Langerhans cells (LCs) are the only professional antigen presenting cells located in the epidermis, therefore may influence the antiviral immune response. Microparticles, or microvesicles, are small membrane particles shed by cells that can exert effects on other cells at both a local and systemic level. We found increased numbers of microparticles were shed from human or mouse keratinocytes expressing the HPV16 E7 oncoprotein, compared with control keratinocytes. Co-culture of LCs with microparticles from E7-expressing cells suppressed the cytotoxic T cell response. We attributed this, at least in part, to the reduction in surface of CD40 and intracellular pro-inflammatory cytokine IL-12 p40 subunit that we measured in the LCs. The evidence provided here shows that co-culture of E7-microparticles with LCs inhibits antigen-specific cytotoxicity. This is an important finding, suggesting that microparticles from HPV-infected cells could suppress the T cell response by regulating LCs, potentially contributing to persistence of HPV infection and cancer.
Collapse
|
37
|
Przybyla B, Pinomäki A, Petäjä J, Joutsi-Korhonen L, Strandberg K, Hillarp A, Öhlin AK, Ruutu T, Volin L, Lassila R. Coordinated responses of natural anticoagulants to allogeneic stem cell transplantation and acute GVHD - A longitudinal study. PLoS One 2017; 12:e0190007. [PMID: 29272282 PMCID: PMC5741247 DOI: 10.1371/journal.pone.0190007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/06/2017] [Indexed: 12/27/2022] Open
Abstract
Background Allogeneic stem cell transplantation (SCT) enhances coagulation via endothelial perturbation and inflammation. Role of natural anticoagulants in interactions between coagulation and inflammation as well as in acute graft-versus-host disease (GVHD) are not well known. The purpose of this study was to define changes in natural anticoagulants over time in association with GVHD. Patients and methods This prospective study included 30 patients who received grafts from siblings (n = 19) or unrelated donors (n = 11). Eight patients developed GVHD. Standard clinical assays were applied to measure natural anticoagulants, represented by protein C (PC), antithrombin (AT), protein S (PS), complex of activated PC with its inhibitor (APC-PCI) and by markers of endothelial activation: Factor VIII coagulant activity (FVIII:C) and soluble thrombomodulin (s-TM) at 6–8 time points over three months. Results Overall, PC, AT and FVIII:C increased in parallel after engraftment. Significant correlations between PC and FVIII:C (r = 0.64–0.82, p<0.001) and between PC and AT (r = 0.62–0.81, p<0.05) were observed at each time point. Patients with GVHD had 21% lower PC during conditioning therapy and 55% lower APC-PCI early after transplantation, as well as 37% higher values of s-TM after engraftment. The GVHD group had also increases of PC (24%), FVIII: C (28%) and AT (16%) three months after transplantation. Conclusion The coordinated activation of natural anticoagulants in our longitudinal study indicates the sustained ability of adaptation to endothelial and inflammatory activation during allogenic SCT treatment. The suboptimal control of coagulation by natural anticoagulants at early stage of SCT may contribute to onset of GVHD.
Collapse
Affiliation(s)
- Beata Przybyla
- Coagulation Disorders Unit, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Anne Pinomäki
- Stem Cell Transplantation Unit, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Jari Petäjä
- Department of Paediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- Hematology and Clinical Chemistry and HUSLAB Laboratory Services, Helsinki University Hospital, Helsinki, Finland
| | - Karin Strandberg
- Department of Clinical Chemistry, Lund University, Lund University Hospital, Lund, Sweden
- University and Regional Laboratories, Skane County Council, Coagulation Laboratory, Clinical Chemistry, Malmö, Sweden
| | - Andreas Hillarp
- Department of Clinical Chemistry, Lund University, Lund University Hospital, Lund, Sweden
- University and Regional Laboratories, Skane County Council, Coagulation Laboratory, Clinical Chemistry, Malmö, Sweden
| | - Ann-Kristin Öhlin
- Department of Clinical Chemistry, Lund University, Lund University Hospital, Lund, Sweden
- University and Regional Laboratories, Skane County Council, Coagulation Laboratory, Clinical Chemistry, Malmö, Sweden
| | - Tapani Ruutu
- Stem Cell Transplantation Unit, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Liisa Volin
- Stem Cell Transplantation Unit, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Riitta Lassila
- Coagulation Disorders Unit, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Hematology and Clinical Chemistry and HUSLAB Laboratory Services, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
38
|
Gritti BB, Binder C. Oxidation-specific epitopes are major targets of innate immunity in atherothrombosis. Hamostaseologie 2017; 36:89-96. [DOI: 10.5482/hamo-14-11-0069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/30/2015] [Indexed: 01/08/2023] Open
Abstract
ZusammenfassungAtherosklerose ist eine chronisch-entzündliche Erkrankung der Gefäßwände, die durch das Zusammenspiel von Dyslipidämie und vermehrtem oxidativen Stress verursacht wird. Die damit verbundene Lipidperoxidation führt zu einer Reihe von Abbauprodukten von Membranlipiden, sogenannten oxidations-spezifischen Epitopen (OSE). OSE finden sich in oxidierten Lipoproteinen und auf der Oberfläche absterbender Zellen, und ihre Fähigkeit inflammatorische und thrombogene Reaktionen auszulösen ist weithin bekannt. Jüngste Studien konnten zeigen, daß OSE spezifische Zielstrukturen für eine Reihe von zellulären und humoralen Rezeptoren des angeborenen Immunsystems darstellen. Dadurch kann das Immunsystem, metabolische Abbaubprodukte erkennen und wichtige physiologische “Haushaltsfunktionen” vermitteln, z.B. durch die kontrollierte Entsorgung abgestorbener Zellen und oxidierten Moleküle. So wurde gezeigt, daß natürliche IgM Antikörper mit Spezifität für OSE Mäuse vor der Entstehung atherosklerotischer Läsionen schützen. So können spezifische natürliche IgM Antikörper die pro-inflammatorischen und pro-thrombotischen Effekte von OSE neutralisieren, währenddessen niedrige Plasmaspiegel OSE-spezifischer IgM Antikörper mit einem erhöhten Risiko für Myokardinfarkt assoziiert sind. Schlussfolgerung: Das Verständnis der molekularen Komponenten und Mechanismen, die an diesem Prozess beteiligt sind, werden in Zukunft dazu beitragen, Personen mit einem erhöhten Risiko für Atherothrombose besser zu identifizieren und möglicherweise neue therapeutische Ansatzpunkte zu definieren.
Collapse
|
39
|
Degirmenci SE, Zobairi F, Berger A, Meyer G, Burban M, Mostefai HA, Levy B, Toti F, Boisramé-Helms J, Delabranche X, Meziani F. Pharmacological modulation of procoagulant microparticles improves haemodynamic dysfunction during septic shock in rats. Thromb Haemost 2017; 111:154-64. [DOI: 10.1160/th13-04-0313] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 09/04/2013] [Indexed: 01/02/2023]
Abstract
SummaryCirculating microparticles play a pro-inflammatory and procoagulant detrimental role in the vascular dysfunction of septic shock. It was the objective of this study to investigate mechanisms by which a pharmacological modulation of microparticles could affect vascular dysfunction in a rat model of septic shock. Septic or sham rats were treated by activated protein C (aPC) and resuscitated during 4 hours. Their microparticles were harvested and inoculated to another set of healthy recipient rats. Haemodynamic parameters were monitored, circulating total procoagulant microparticles assessed by prothrombinase assay, and their cell origin characterised. Mesenteric resistance arteries, aorta and heart were harvested for western blotting analysis. We found that a) the amount and phenotype of circulating microparticles were altered in septic rats with an enhanced endothelial, leucocyte and platelet contribution; b) aPC treatment significantly reduced the generation of leucocyte microparticles and norepinephrine requirements to reach the mean arterial pressure target in septic rats; c) Microparticles from untreated septic rats, but not from aPC-treated ones, significantly reduced the healthy recipients’ mean arterial pressure; d) Microparticle thromboxane content and aPC activity were significantly increased in aPC-treated septic rats. In inoculated naïve recipients, microparticles from aPC-treated septic rats prompted reduced NF-κB and cyclooxygenase-2 arterial activation, blunted the generation of pro-inflammatory iNOS and secondarily increased platelet and endothelial microparticles. In conclusion, in this septic shock model, increased circulating levels of procoagulant microparticles led to negative haemodynamic outcomes. Pharmacological treatment by aPC modified the cell origin and levels of circulating microparticles, thereby limiting vascular inflammation and favouring haemodynamic improvement.
Collapse
|
40
|
Renaud-Picard B, Toussaint J, Leclercq A, Reeb J, Kessler L, Toti F, Kessler R. [Membranous microparticles and respiratory disease]. Rev Mal Respir 2017; 34:1058-1071. [PMID: 29132745 DOI: 10.1016/j.rmr.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/05/2017] [Indexed: 01/30/2023]
Abstract
Microparticles (MP) are plasmic membrane fragments released from cells after physiological stimulation or stress conditions like inflammation or infection. Their production is correlated to the rate of cell apoptosis. All types of cells can produce MP but they are produced mainly by platelets, endothelial cells, and leukocytes. They carry many bio-active molecules on their surface, specific to the parental cell, giving them the ability to be biomarkers and bio-effectors. MP are present in circulating blood, tissues and many biological fluids. Circulating MP levels can change during the course of many diseases. They have been the subject of many studies in the fields of cardiovascular disease and oncology. In the lungs, they are present in circulating blood and in the airways. They seem to have a role in pulmonary homeostasis in physiological situations and also in the expression of several disease processes. In this review of the literature, we were interested in the quantitative and qualitative variations in MP and their impact in airway diseases like chronic obstructive pulmonary disease (COPD) and asthma, pulmonary fibrosis and pulmonary hypertension.
Collapse
Affiliation(s)
- B Renaud-Picard
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France.
| | - J Toussaint
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - A Leclercq
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - J Reeb
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - L Kessler
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - F Toti
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - R Kessler
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
41
|
Jayachandran M, Garovic VD, Mielke MM, Bailey KR, Lahr BD, Miller VM. Characterization of intravascular cellular activation in relationship to subclinical atherosclerosis in postmenopausal women. PLoS One 2017; 12:e0183159. [PMID: 28910282 PMCID: PMC5598935 DOI: 10.1371/journal.pone.0183159] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/31/2017] [Indexed: 02/02/2023] Open
Abstract
Objective Mechanisms and interactions among intravascular cells contributing to development of subclinical atherosclerosis are poorly understood. In women, both menopausal status and pregnancy history influence progression of atherosclerosis. This study examined activation and interactions among blood elements with subclinical atherosclerosis in menopausal women with known pregnancy histories. Methods Carotid intima-media thickness (CIMT), as a marker of subclinical atherosclerosis, was measured using B-mode ultrasound in age- and parity-matched women [40 with and 40 without a history of preeclampsia] 35 years after the index pregnancy. Interactions among intravascular cells (38 parameters) were measured by flow cytometry in venous blood. Data analysis was by principal component which retained 7 independent dimensions accounting for 63% of the variability among 38 parameters. Results CIMT was significantly greater in women with a history of preeclampsia (P = 0.004). Platelet aggregation and platelet interactions with granulocytes and monocytes positively associated with CIMT in postmenopausal women independent of their pregnancy history (ρ = 0.258, P< 0.05). However, the association of the number of platelets, platelet activation and monocyte-platelet interactions with CIMT differed significantly depending upon pregnancy history (test for interaction, P<0.001). Conclusion Interactions among actived intravascular cells and their association with subclinical atherosclerosis differ in women depending upon their pregnancy histories.
Collapse
Affiliation(s)
- Muthuvel Jayachandran
- Departments of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Vesna D. Garovic
- General Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michelle M. Mielke
- Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, United States of America
- Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kent R. Bailey
- Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, United States of America
- Health Science Research, Division of Biostatistics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brian D. Lahr
- Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, United States of America
- Health Science Research, Division of Biostatistics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Virginia M. Miller
- Departments of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
42
|
Aharon A, Sabbah A, Ben-Shaul S, Berkovich H, Loven D, Brenner B, Bar-Sela G. Chemotherapy administration to breast cancer patients affects extracellular vesicles thrombogenicity and function. Oncotarget 2017; 8:63265-63280. [PMID: 28968987 PMCID: PMC5609919 DOI: 10.18632/oncotarget.18792] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) is the most prevalent type of malignancy in women. Extracellular vesicles (EVs) are subcellular membrane blebs that include exosomes and microparticles.
Collapse
Affiliation(s)
- Anat Aharon
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Anni Sabbah
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Shahar Ben-Shaul
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Hila Berkovich
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - David Loven
- Department of Oncology, Ha'emek Medical Center, Afula, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gil Bar-Sela
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Oncology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
43
|
Cannito S, Morello E, Bocca C, Foglia B, Benetti E, Novo E, Chiazza F, Rogazzo M, Fantozzi R, Povero D, Sutti S, Bugianesi E, Feldstein AE, Albano E, Collino M, Parola M. Microvesicles released from fat-laden cells promote activation of hepatocellular NLRP3 inflammasome: A pro-inflammatory link between lipotoxicity and non-alcoholic steatohepatitis. PLoS One 2017; 12:e0172575. [PMID: 28249038 PMCID: PMC5331985 DOI: 10.1371/journal.pone.0172575] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is a major form of chronic liver disease in the general population in relation to its high prevalence among overweight/obese individuals and patients with diabetes type II or metabolic syndrome. NAFLD can progress to steatohepatitis (NASH), fibrosis and cirrhosis and end-stage of liver disease but mechanisms involved are still incompletely characterized. Within the mechanisms proposed to mediate the progression of NAFLD, lipotoxicity is believed to play a major role. In the present study we provide data suggesting that microvesicles (MVs) released by fat-laden cells undergoing lipotoxicity can activate NLRP3 inflammasome following internalization by either cells of hepatocellular origin or macrophages. Inflammasome activation involves NF-kB-mediated up-regulation of NLRP3, pro-caspase-1 and pro-Interleukin-1, then inflammasome complex formation and Caspase-1 activation leading finally to an increased release of IL-1β. Since the release of MVs from lipotoxic cells and the activation of NLRP3 inflammasome have been reported to occur in vivo in either clinical or experimental NASH, these data suggest a novel rational link between lipotoxicity and increased inflammatory response.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisa Benetti
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Erica Novo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Fausto Chiazza
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Mara Rogazzo
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Roberto Fantozzi
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Davide Povero
- Department of Pediatrics, University of California San Diego (UCSD), La Jolla, CA, United States of America
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University “Amedeo Avogadro” of East Piedmont, Novara, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, Torino, Italy
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California San Diego (UCSD), La Jolla, CA, United States of America
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University “Amedeo Avogadro” of East Piedmont, Novara, Italy
| | - Massimo Collino
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
44
|
Johnson BL, Midura EF, Prakash PS, Rice TC, Kunz N, Kalies K, Caldwell CC. Neutrophil derived microparticles increase mortality and the counter-inflammatory response in a murine model of sepsis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2554-2563. [PMID: 28108420 DOI: 10.1016/j.bbadis.2017.01.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/04/2017] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Abstract
Although advances in medical care have significantly improved sepsis survival, sepsis remains the leading cause of death in the ICU. This is likely due to a lack of complete understanding of the pathophysiologic mechanisms that lead to dysfunctional immunity. Neutrophil derived microparticles (NDMPs) have been shown to be the predominant microparticle present at infectious and inflamed foci in human models, however their effect on the immune response to inflammation and infection is sepsis has not been fully elucidated. As NDMPs may be a potential diagnostic and therapeutic target, we sought to determine the impact NDMPs on the immune response to a murine polymicrobial sepsis. We found that peritoneal neutrophil numbers, bacterial loads, and NDMPs were increased in our abdominal sepsis model. When NDMPs were injected into septic mice, we observed increased bacterial load, decreased neutrophil recruitment, increased expression of IL-10 and worsened mortality. Furthermore, the NDMPs express phosphatidylserine and are ingested by F4/80 macrophages via a Tim-4 and MFG-E8 dependent mechanism. Finally, upon treatment, NDMPs decrease macrophage activation, increase IL-10 release and decrease macrophage numbers. Altogether, these data suggest that NDMPs enhance immune dysfunction in sepsis by blunting the function of neutrophils and macrophages, two key cell populations involved in the early immune response to infection. This article is part of a Special Issue entitled: Immune and Metabolic Alterations in Trauma and Sepsis edited by Dr. Raghavan Raju.
Collapse
Affiliation(s)
- Bobby L Johnson
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Emily F Midura
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Priya S Prakash
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Teresa C Rice
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Natalia Kunz
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States; Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Kathrin Kalies
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States; Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Charles C Caldwell
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States.
| |
Collapse
|
45
|
Liu Y, Zhang R, Qu H, Wu J, Li L, Tang Y. Endothelial microparticles activate endothelial cells to facilitate the inflammatory response. Mol Med Rep 2017; 15:1291-1296. [DOI: 10.3892/mmr.2017.6113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/21/2016] [Indexed: 11/05/2022] Open
|
46
|
Sparrow RL, Simpson RJ, Greening DW. A Protocol for the Preparation of Cryoprecipitate and Cryo-depleted Plasma for Proteomic Studies. Methods Mol Biol 2017; 1619:23-30. [PMID: 28674874 DOI: 10.1007/978-1-4939-7057-5_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cryoprecipitate is a concentrate of high-molecular-weight plasma proteins that precipitate when frozen plasma is slowly thawed at 1-6 °C. The concentrate contains factor VIII (antihemophilic factor), von Willebrand factor (vWF), fibrinogen, factor XIII, fibronectin, and small amounts of other plasma proteins. Clinical grade preparations of cryoprecipitate are mainly used to treat fibrinogen deficiency caused by acute bleeding or functional abnormalities of the fibrinogen protein. In the past, cryoprecipitate was used to treat von Willebrand disease and hemophilia A (factor VIII deficiency), but the availability of more highly purified coagulation factor concentrates or recombinant protein preparations has superseded the use of cryoprecipitate for these coagulopathies. Cryo-depleted plasma ("cryosupernatant") is the plasma supernatant remaining following removal of the cryoprecipitate from frozen-thawed plasma. It contains all the other plasma proteins and clotting factors present in plasma that remain soluble during cold-temperature thawing of the plasma. This protocol describes the clinical-scale preparation of cryoprecipitate and cryo-depleted plasma for proteomic studies.
Collapse
Affiliation(s)
- Rosemary L Sparrow
- Transfusion Science, Melbourne, VIC, Australia.
- Department of Immunology and Pathology, Monash University, AMREP, 89 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
47
|
Puhm F, Binder CJ. Characterization of Natural IgM Antibodies Recognizing Oxidation-Specific Epitopes on Circulating Microvesicles. Methods Mol Biol 2017; 1643:147-154. [PMID: 28667535 DOI: 10.1007/978-1-4939-7180-0_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Natural IgM antibodies specific for oxidation-specific epitopes (OSEs) have been found to bind to circulating microvesicles (MVs), also known as microparticles. This chapter describes how endogenous natural IgM antibodies bound to circulating MV subsets can be characterized on the one hand, and how monoclonal natural IgM antibodies can be used to characterize subsets of circulating MVs on the other hand.
Collapse
Affiliation(s)
- Florian Puhm
- Department of Laboratory Medicine, Medical University of Vienna, Lazarettgasse 14, AKH BT 25.2/6, 1090, Vienna, Austria.,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Lazarettgasse 14, AKH BT 25.2/6, 1090, Vienna, Austria. .,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, 1090, Vienna, Austria.
| |
Collapse
|
48
|
Affiliation(s)
- Leonard C. Edelstein
- Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
49
|
Żmigrodzka M, Guzera M, Miśkiewicz A, Jagielski D, Winnicka A. The biology of extracellular vesicles with focus on platelet microparticles and their role in cancer development and progression. Tumour Biol 2016; 37:14391-14401. [PMID: 27629289 PMCID: PMC5126185 DOI: 10.1007/s13277-016-5358-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of structures which can be classified into smaller in size and relatively homogenous exosomes (EXSMs)—spherical fragments of lipid bilayers from inner cell compartments—and bigger in size ectosomes (ECSMs)—a direct consequence of cell-membrane blebbing. EVs can be found in body fluids of healthy individuals. Their number increases in cancer and other pathological conditions. EVs can originate from various cell types, including leukocytes, erythrocytes, thrombocytes, and neoplastic cells. Platelet microparticles (PMPs) are the most abundant population of EVs in blood. It is well documented that PMPs, being a crucial element of EVs signaling, are involved in tumor growth, metastasis, and angiogenesis and may participate in the development of multidrug resistance by tumor cells. The aim of this review is to present the role of PMPs in carcinogenesis. The biology and functions of PMPs with a particular emphasis on the most recent scientific reports on EV properties are also characterized.
Collapse
Affiliation(s)
- M Żmigrodzka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| | - M Guzera
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - A Miśkiewicz
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland.
| | - D Jagielski
- Veterinary Clinic BIALOBRZESKA, Częstochowska 20, Warsaw, Poland
| | - A Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| |
Collapse
|
50
|
Antonelou MH, Seghatchian J. Update on extracellular vesicles inside red blood cell storage units: Adjust the sails closer to the new wind. Transfus Apher Sci 2016; 55:92-104. [DOI: 10.1016/j.transci.2016.07.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|