1
|
Pan K, Zhu Y, Chen P, Yang K, Chen Y, Wang Y, Dai Z, Huang Z, Zhong P, Zhao X, Fan S, Ning L, Zhang J, Chen P. Biological functions and biomedical applications of extracellular vesicles derived from blood cells. Free Radic Biol Med 2024; 222:43-61. [PMID: 38848784 DOI: 10.1016/j.freeradbiomed.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
There is a growing interest in using extracellular vesicles (EVs) for therapeutic applications. EVs are composed of cytoplasmic proteins and nucleic acids and an external lipid bilayer containing transmembrane proteins on their surfaces. EVs can alter the state of the target cells by interacting with the receptor ligand of the target cell or by being internalised by the target cell. Blood cells are the primary source of EVs, and 1 μL of plasma contains approximately 1.5 × 107 EVs. Owing to their easy acquisition and the avoidance of cell amplification in vitro, using blood cells as a source of therapeutic EVs has promising clinical application prospects. This review summarises the characteristics and biological functions of EVs derived from different blood cell types (platelets, erythrocytes, and leukocytes) and analyses the prospects and challenges of using them for clinical therapeutic applications. In summary, blood cell-derived EVs can regulate different cell types such as immune cells (macrophages, T cells, and dendritic cells), stem cells, and somatic cells, and play a role in intercellular communication, immune regulation, and cell proliferation. Overall, blood cell-derived EVs have the potential for use in vascular diseases, inflammatory diseases, degenerative diseases, and injuries. To promote the clinical translation of blood cell-derived EVs, researchers need to perform further studies on EVs in terms of scalable and reproducible isolation technology, quality control, safety, stability and storage, regulatory issues, cost-effectiveness, and long-term efficacy.
Collapse
Affiliation(s)
- Kaifeng Pan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yiwei Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Ke Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yiyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yongcheng Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325088, China
| | - Zhenxiang Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Peiyu Zhong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Xing Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China.
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China.
| | - Lei Ning
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China.
| | - Jianfeng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China.
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
2
|
Marshall JN, Klein MN, Karki P, Promnares K, Setua S, Fan X, Buehler PW, Birukov KG, Vasta GR, Fontaine MJ. Aberrant GPA expression and regulatory function of red blood cells in sickle cell disease. Blood Adv 2024; 8:1687-1697. [PMID: 38231087 PMCID: PMC11006809 DOI: 10.1182/bloodadvances.2023011611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 01/18/2024] Open
Abstract
ABSTRACT Glycophorin A (GPA), a red blood cell (RBC) surface glycoprotein, can maintain peripheral blood leukocyte quiescence through interaction with a sialic acid-binding Ig-like lectin (Siglec-9). Under inflammatory conditions such as sickle cell disease (SCD), the GPA of RBCs undergo structural changes that affect this interaction. Peripheral blood samples from patients with SCD before and after RBC transfusions were probed for neutrophil and monocyte activation markers and analyzed by fluorescence-activated cell sorting (FACS). RBCs were purified and tested by FACS for Siglec-9 binding and GPA expression, and incubated with cultured endothelial cells to evaluate their effect on barrier function. Activated leukocytes from healthy subjects (HS) were coincubated with healthy RBCs (RBCH), GPA-altered RBCs, or GPA-overexpressing (OE) cells and analyzed using FACS. Monocyte CD63 and neutrophil CD66b from patients with SCD at baseline were increased 47% and 27%, respectively, as compared with HS (P = .0017, P = .0162). After transfusion, these markers were suppressed by 22% and 17% (P = .0084, P = .0633). GPA expression in RBCSCD was 38% higher (P = .0291) with decreased Siglec-9 binding compared with RBCH (0.0266). Monocyte CD63 and neutrophil CD66b were suppressed after incubation with RBCH and GPA-OE cells, but not with GPA-altered RBCs. Endothelial barrier dysfunction after lipopolysaccharide challenge was restored fully with exposure to RBCH, but not with RBCSCD, from patients in pain crisis, or with RBCH with altered GPA. Pretransfusion RBCSCD do not effectively maintain the quiescence of leukocytes and endothelium, but quiescence is restored through RBC transfusion, likely by reestablished GPA-Siglec-9 interactions.
Collapse
Affiliation(s)
- Juliana N. Marshall
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Matthew N. Klein
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Pratap Karki
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD
| | - Saini Setua
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Xiaoxuan Fan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD
| | - Paul W. Buehler
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD
| | - Gerardo R. Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD
- The Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD
| | - Magali J. Fontaine
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
3
|
Ebeyer-Masotta M, Eichhorn T, Fischer MB, Weber V. Impact of production methods and storage conditions on extracellular vesicles in packed red blood cells and platelet concentrates. Transfus Apher Sci 2024; 63:103891. [PMID: 38336556 DOI: 10.1016/j.transci.2024.103891] [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] [Indexed: 02/12/2024]
Abstract
The use of blood and blood products can be life-saving, but there are also certain risks associated with their administration and use. Packed red blood cells (pRBCs) and platelet concentrates are the most commonly used blood products in transfusion medicine to treat anemia or acute and chronic bleeding disorders, respectively. During the production and storage of blood products, red blood cells and platelets release extracellular vesicles (EVs) as a result of the storage lesion, which may affect product quality. EVs are subcellular structures enclosed by a lipid bilayer and originate from the endosomal system or from the plasma membrane. They play a pivotal role in intercellular communication and are emerging as important regulators of inflammation and coagulation. Their cargo and their functional characteristics depend on the cell type from which they originate, as well as on their microenvironment, influencing their capacity to promote coagulation and inflammatory responses. Hence, the potential involvement of EVs in transfusion-related adverse events is increasingly recognized and studied. Here, we review the knowledge regarding the effect of production and storage conditions of pRBCs and platelet concentrates on the release of EVs. In this context, the mode of processing and anticoagulation, the influence of additive solutions and leukoreduction, as well as the storage duration will be addressed, and we discuss potential implications of EVs for the clinical outcome of transfusion.
Collapse
Affiliation(s)
- Marie Ebeyer-Masotta
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria
| | - Tanja Eichhorn
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria
| | - Michael B Fischer
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria; Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Viktoria Weber
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria.
| |
Collapse
|
4
|
Zhang E, Phan P, Zhao Z. Cellular nanovesicles for therapeutic immunomodulation: A perspective on engineering strategies and new advances. Acta Pharm Sin B 2023; 13:1789-1827. [PMID: 37250173 PMCID: PMC10213819 DOI: 10.1016/j.apsb.2022.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular nanovesicles which are referred to as cell-derived, nanosized lipid bilayer structures, have emerged as a promising platform for regulating immune responses. Owing to their outstanding advantages such as high biocompatibility, prominent structural stability, and high loading capacity, cellular nanovesicles are suitable for delivering various immunomodulatory molecules, such as small molecules, nucleic acids, peptides, and proteins. Immunomodulation induced by cellular nanovesicles has been exploited to modulate immune cell behaviors, which is considered as a novel cell-free immunotherapeutic strategy for the prevention and treatment of diverse diseases. Here we review emerging concepts and new advances in leveraging cellular nanovesicles to activate or suppress immune responses, with the aim to explicate their applications for immunomodulation. We overview the general considerations and principles for the design of engineered cellular nanovesicles with tailored immunomodulatory activities. We also discuss new advances in engineering cellular nanovesicles as immunotherapies for treating major diseases.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
- Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
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
|
Red Blood Cell Inspired Strategies for Drug Delivery: Emerging Concepts and New Advances. Pharm Res 2022; 39:2673-2698. [PMID: 35794397 DOI: 10.1007/s11095-022-03328-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/29/2022] [Indexed: 12/09/2022]
Abstract
In the past five decades, red blood cells (RBCs) have been extensively explored as drug delivery systems due to their distinguishing potential in modulating the pharmacokinetic, pharmacodynamics, and biological activity of carried payloads. The extensive interests in RBC-mediated drug delivery technologies are in part derived from RBCs' unique biological features such as long circulation time, wide access to many tissues in the body, and low immunogenicity. Owing to these outstanding properties, a large body of efforts have led to the development of various RBC-inspired strategies to enable precise drug delivery with enhanced therapeutic efficacy and reduced off-target toxicity. In this review, we discuss emerging concepts and new advances in such RBC-inspired strategies, including native RBCs, ghost RBCs, RBC-mimetic nanoparticles, and RBC-derived extracellular vesicles, for drug delivery.
Collapse
|
7
|
Su WL, Chan CY, Cheng CF, Shui HA, Ku HC. Erythrocyte degradation, metabolism, secretion, and communication with immune cells in the blood during sepsis: A review. Tzu Chi Med J 2022; 34:125-133. [PMID: 35465286 PMCID: PMC9020243 DOI: 10.4103/tcmj.tcmj_58_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 05/26/2021] [Indexed: 11/04/2022] Open
Abstract
Sepsis is a health issue that affects millions of people worldwide. It was assumed that erythrocytes were affected by sepsis. However, in recent years, a number of studies have shown that erythrocytes affect sepsis as well. When a pathogen invades the human body, it infects the blood and organs, causing infection and sepsis-related symptoms. Pathogens change the internal environment, increasing the levels of reactive oxygen species, influencing erythrocyte morphology, and causing erythrocyte death, i.e., eryptosis. Characteristics of eryptosis include cell shrinkage, membrane blebbing, and surface exposure of phosphatidylserine (PS). Eryptotic erythrocytes increase immune cell proliferation, and through PS, attract macrophages that remove the infected erythrocytes. Erythrocyte-degraded hemoglobin derivatives and heme deteriorate infection; however, they could also be metabolized to a series of derivatives. The result that erythrocytes play an anti-infection role during sepsis provides new perspectives for treatment. This review focuses on erythrocytes during pathogenic infection and sepsis.
Collapse
|
8
|
Ma X, Liu Y, Han Q, Han Y, Wang J, Zhang H. Transfusion‑related immunomodulation in patients with cancer: Focus on the impact of extracellular vesicles from stored red blood cells (Review). Int J Oncol 2021; 59:108. [PMID: 34841441 DOI: 10.3892/ijo.2021.5288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/05/2021] [Indexed: 01/28/2023] Open
Abstract
Red blood cell (RBC) transfusions may have a negative impact on the prognosis of patients with cancer, where transfusion‑related immunomodulation (TRIM) may be a significant contributing factor. A number of components have been indicated to be associated with TRIM. Among these, the impact of extracellular vesicles (EVs) has been garnering increasing attention from researchers. EVs are defined as nano‑scale, cell‑derived vesicles that carry a variety of bioactive molecules, including proteins, nucleic acids and lipids, to mediate cell‑to‑cell communication and exert immunoregulatory functions. RBCs in storage constitutively secrete EVs, which serve an important role in TRIM in patients with cancer receiving a blood transfusion. Therefore, the present review aimed to first summarize the available information on the biogenesis and characterization of EVs. Subsequently, the possible mechanisms of TRIM in patients with cancer and the impact of EVs on TRIM were discussed, aiming to provide an outlook for future studies, specifically for formulating recommendations for managing patients with cancer receiving RBC transfusions.
Collapse
Affiliation(s)
- Xingyu Ma
- Class 2018 Medical Inspection Technology, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yanxi Liu
- Class 2018 Medical Inspection Technology, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qianlan Han
- Class 2018 Medical Inspection Technology, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jing Wang
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Hongwei Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
9
|
Flatman LK, Fergusson DA, Lacroix J, Ducruet T, Papenburg J, Fontela PS. Association between the length of storage of transfused leukoreduced red blood cell units and hospital-acquired infections in critically ill children: A secondary analysis of the TRIPICU study. Transfus Med 2021; 31:467-473. [PMID: 34585466 DOI: 10.1111/tme.12824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Evaluate the association between leukoreduced red blood cell (RBC) storage length and hospital-acquired infection (HAI) incidence rate in critically ill children. BACKGROUND RBC transfusions are common in critically ill children. Despite their benefits, observational studies suggest an association between them and HAIs. One possible mechanism for increased HAI is transfusion-related immunomodulation due to bioactive substances' release as transfused blood ages. METHODS In this secondary analysis of the 'Transfusion Requirement in Paediatric Intensive Care Units' (TRIPICU) study, we analysed a subset of 257 participants that received only one pre-storage leukoreduced RBC transfusion. RBC storage length was classified as 1) transfusion of 'fresh' RBCs (≤10 days), 2) transfusion of 'stored' RBCs (21-34 days), and 3) transfusion of 'long-stored' RBCs (≥35 days). All were compared to a 'golden' period (11-20 days), representing the time between 'fresh' and 'stored'. We used quasi-Poisson multivariable regression models to estimate the HAI incidence rate ratio (IRR) and corresponding 95% confidence interval (CI). RESULTS We found that the association between the length of storage time of leukoreduced RBCs and HAIs was not significant in the 'fresh' group (IRR 1.23; 95% CI 0.55, 2.78) and the 'stored' group (IRR 1.61; 95% CI 0.63, 4.13) when compared to the 'golden' period. However, we observed a statistically significant association between the 'long-stored' group and an increase in the HAI incidence rate (IRR 3.66; 95% CI 1.22, 10.98). CONCLUSION Transfusion of leukoreduced RBC units stored for ≥35 days is associated with increased HAI incidence rate in haemodynamically stable, critically ill children.
Collapse
Affiliation(s)
- Leah K Flatman
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jacques Lacroix
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Thierry Ducruet
- Unité de recherche clinique appliquée (URCA), Université de Montréal, Centre de Recherche, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Jesse Papenburg
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada.,Division of Pediatric Infectious Diseases, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Patricia S Fontela
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada.,Division of Pediatric Critical Care Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Fang X, Li J, Hao X, Zhang W, Zhong J, Zhu T, Liao R. Exosomes From Packed Red Cells Induce Human Mast Cell Activation and the Production of Multiple Inflammatory Mediators. Front Immunol 2021; 12:677905. [PMID: 34025676 PMCID: PMC8135094 DOI: 10.3389/fimmu.2021.677905] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/23/2021] [Indexed: 02/05/2023] Open
Abstract
Most blood transfusion-related adverse reactions involve the immunologic responses of recipients to exogenous blood components. Extracellular vesicles isolated from packed red cells can affect the recipient’s immune system. Mast cells are traditionally known as effector cells for allergic transfusion reactions. However, growing evidence supports the notion that activated mast cells might disturb host innate immunologic responses. Exosomes are a type of extracellular vesicle. To determine the effect of exosomes on mast cells, we enriched exosomes derived from volunteer plasma (EXs-nor) and packed red cells (EXs-RBCs) using ultracentrifugation and incubated them with a human mast cell line (HMC-1). We found that EXs-RBC exposure increased the expression of tryptase-1 and prostaglandin D2, the production of multiple inflammatory mediators, and the levels of Toll-like receptor-3 (TLR-3) and phospho-mitogen-activated protein kinase (MAPK) in HMC-1 cells. MAPK inhibitors (SB203580, PD98059, and SP600125) and a TLR-3/dsRNA complex inhibitor reduced the EXs-RBC-stimulated production of inflammatory mediators in HMC-1 cells, whereas the TLR-3 agonist [poly (A:U)] elevated the production of these mediators. These results indicate that EXs-RBCs activate HMC-1 cells and elicit the production of multiple inflammatory mediators, partly via the TLR-3 and MAPK pathways. Mast cells activated by EXs-RBCs exhibit complex inflammatory properties and might play a potential role in transfusion-related adverse reactions.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| | - Jingyi Li
- Department of Dermatovenereology, West China Hospital of Sichuan University, Chengdu, China
| | - Xuechao Hao
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| | - Weiyi Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| | - Jie Zhong
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| | - Ren Liao
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, China
| |
Collapse
|
11
|
Thangaraju K, Neerukonda SN, Katneni U, Buehler PW. Extracellular Vesicles from Red Blood Cells and Their Evolving Roles in Health, Coagulopathy and Therapy. Int J Mol Sci 2020; 22:E153. [PMID: 33375718 PMCID: PMC7796437 DOI: 10.3390/ijms22010153] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Red blood cells (RBCs) release extracellular vesicles (EVs) including both endosome-derived exosomes and plasma-membrane-derived microvesicles (MVs). RBC-derived EVs (RBCEVs) are secreted during erythropoiesis, physiological cellular aging, disease conditions, and in response to environmental stressors. RBCEVs are enriched in various bioactive molecules that facilitate cell to cell communication and can act as markers of disease. RBCEVs contribute towards physiological adaptive responses to hypoxia as well as pathophysiological progression of diabetes and genetic non-malignant hematologic disease. Moreover, a considerable number of studies focus on the role of EVs from stored RBCs and have evaluated post transfusion consequences associated with their exposure. Interestingly, RBCEVs are important contributors toward coagulopathy in hematological disorders, thus representing a unique evolving area of study that can provide insights into molecular mechanisms that contribute toward dysregulated hemostasis associated with several disease conditions. Relevant work to this point provides a foundation on which to build further studies focused on unraveling the potential roles of RBCEVs in health and disease. In this review, we provide an analysis and summary of RBCEVs biogenesis, composition, and their biological function with a special emphasis on RBCEV pathophysiological contribution to coagulopathy. Further, we consider potential therapeutic applications of RBCEVs.
Collapse
Affiliation(s)
- Kiruphagaran Thangaraju
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
| | - Sabari Nath Neerukonda
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Upendra Katneni
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
| | - Paul W. Buehler
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Zhao Y, Wei W, Liu ML. Extracellular vesicles and lupus nephritis - New insights into pathophysiology and clinical implications. J Autoimmun 2020; 115:102540. [PMID: 32893081 PMCID: PMC9107953 DOI: 10.1016/j.jaut.2020.102540] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022]
Abstract
Lupus nephritis (LN) is a major cause for overall morbidity and mortality in patients with systemic lupus erythematosus (SLE), while its pathogenic mechanisms are still not well understood. Extracellular vesicles (EVs) are membrane vesicles that are released from almost all cell types. EVs can be subdivided into exosomes, microvesicles, and apoptotic bodies. Latest studies have shown that EVs can be released during several cellular events, including cell activation, autophagy, and several types of programed cell death, i.e. apoptosis, necroptosis, pyroptosis, and NETosis. Emerging evidence demonstrates that EVs harbor different bioactive molecules, including nucleic acids, proteins, lipids, cytokines, immune complexes (ICs), complements, and other molecules, some of which may contribute to pathogenesis of autoimmune diseases. EVs can serve as novel information shuttle to mediate local autocrine or paracrine signals to nearby cells, and distant endocrine signals to cells located far away. In LN, EVs may have pathogenic effects by transportation of autoantigens or complements, promotion of IC deposition or complement activation, and stimulation of inflammatory responses, renal tissue injury, or microthrombus formation. Additionally, EVs released from kidney cells may serve as specific biomarkers for diagnosis or monitoring of disease activity and therapeutic efficacy. In this review, we will summarize the latest progress about EV generation from basic research, their potential pathologic effects on LN, and their clinical implications. The cutting-edge knowledge about EV research provides insights into novel therapeutic strategy, new tools for diagnosis or prognosis, and evaluation approaches for treatment effectiveness in LN.
Collapse
Affiliation(s)
- Yin Zhao
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, 300020, China
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, 300020, China.
| | - Ming-Lin Liu
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Corporal Michael J. Crescenz VA Medical Center (Philadelphia), Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Papadopoulos C, Panopoulou M, Anagnostopoulos K, Tentes I. Immune and Metabolic Interactions of Human Erythrocytes: A Molecular Perspective. Endocr Metab Immune Disord Drug Targets 2020; 21:843-853. [PMID: 33148159 DOI: 10.2174/1871530320666201104115016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Apart from their main function as oxygen carriers in vertebrates, erythrocytes are also involved in immune regulation. By circulating throughout the body, the erythrocytes are exposed and interact with tissues that are damaged as a result of a disease. In this study, we summarize the literature regarding the contribution of erythrocytes to immune regulation and metabolism. Under the circumstances of a disease state, the erythrocytes may lose their antioxidant capacity and release Damage Associated Molecular Patterns, resulting in the regulation of innate and adaptive immunity. In addition, the erythrocytes scavenge and affect the levels of chemokines, circulating cell-free mtDNA, and C3b attached immune complexes. Furthermore, through surface molecules, erythrocytes control the function of T lymphocytes, macrophages, and dendritic cells. Through an array of enzymes, red blood cells contribute to the pool of blood's bioactive lipids. Finally, the erythrocytes contribute to reverse cholesterol transport through various mechanisms. Our study is highlighting overlooked molecular interactions between erythrocytes and immunity and metabolism, which could lead to the discovery of potent therapeutic targets for immunometabolic diseases.
Collapse
Affiliation(s)
| | - Maria Panopoulou
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Ioannis Tentes
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
14
|
Abstract
The components of the immune system may be present in early stages of embryonic and then fetal, then they reach maturity at different stages of pregnancy. Just as the growth and development of the components of the embryonic and then fetal immune system progressively mature, functions are acquired sequentially during the course of pregnancy, both the ability to mount a cell-mediated or antibody-mediated immune response and the tolerance towards a certain group of antigens. The fetus is immunocompetent because during this development, it acquires the ability to generate an immune response. As development takes place, the fetus also generates specific tolerance as it is exposed to genetically foreign and non-inherited maternal antigens. Nonetheless, the fetal immune system does not attack nor harm maternal tissues. At birth, the immune system, although developed, is not mature enough yet. Furthermore, passive transfer of maternal antibodies creates a unique scenario of compatibility that cannot be seen in children or adults. Recent advances in knowledge of fetal and neonatal immunology make it possible to recognize the risks associated with transfusion and how to resolve them.
Collapse
|
15
|
Gao Y, Jin H, Tan H, Wang Y, Wu J, Wang Y, Zhang J, Yang Y, Tian W, Hou R. The role of extracellular vesicles from stored RBC units in B lymphocyte survival and plasma cell differentiation. J Leukoc Biol 2020; 108:1765-1776. [PMID: 32421907 DOI: 10.1002/jlb.1a0220-666r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/22/2020] [Accepted: 03/28/2020] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are small, double-membrane vesicles derived from erythrocytes, leukocytes, platelets, and cells of multiple tissues under physiologic or pathologic conditions. The role of EVs in stored RBC units is of great interest with respect to transfusion-related immunomodulation. The current study focuses on the quantity of EVs isolated from stored RBC units and their action on B cell-mediated immune responses. The in vitro experiment demonstrated that EVs exhibited a negative role in B cell survival, plasmacytic differentiation, and class switch recombination under LPS stimulation. Furthermore, LPS-induced antibody production was significantly decreased after EVs injection in vivo. Biochemical analysis revealed that EVs hampered the expression of Blimp-1 and IRF4 and the activation of NF-κB pathway in LPS-primed B cells. Overall, these data imply a vital role for EVs isolated from RBC units in B cell-mediated immune responses.
Collapse
Affiliation(s)
- Yuhan Gao
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Hui Tan
- Guangdong Innovation Platform of Translational Research for Cerebrovascular Diseases, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yan Wang
- Department of Immunology, and Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, China
| | - Jia Wu
- Department of Immunology, and Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, China
| | - Yuqing Wang
- Department of Immunology, and Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, China
| | - Jianhua Zhang
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| | - Ying Yang
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| | - Wenqin Tian
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| | - Ruiqin Hou
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| |
Collapse
|
16
|
Bhatti G, Romero R, Rice GE, Fitzgerald W, Pacora P, Gomez-Lopez N, Kavdia M, Tarca AL, Margolis L. Compartmentalized profiling of amniotic fluid cytokines in women with preterm labor. PLoS One 2020; 15:e0227881. [PMID: 31945128 PMCID: PMC6964819 DOI: 10.1371/journal.pone.0227881] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Amniotic fluid cytokines have been implicated in the mechanisms of preterm labor and birth. Cytokines can be packaged within or on the surface of extracellular vesicles. The main aim of this study was to test whether the protein abundance internal to and on the surface of extracellular vesicles changes in the presence of sterile intra-amniotic inflammation and proven intra-amniotic infection in women with preterm labor as compared to the women with preterm labor without either intra-amniotic inflammation or proven intra-amniotic infection. Study design Women who had an episode of preterm labor and underwent an amniocentesis for the diagnosis of intra-amniotic infection or intra-amniotic inflammation were classified into three groups: 1) preterm labor without either intra-amniotic inflammation or proven intra-amniotic infection, 2) preterm labor with sterile intra-amniotic inflammation, and 3) preterm labor with intra-amniotic infection. The concentrations of 38 proteins were determined on the extracellular vesicle surface, within the vesicles, and in the soluble fraction of amniotic fluid. Results 1) Intra-amniotic inflammation, regardless of detected microbes, was associated with an increased abundance of amniotic fluid cytokines on the extracellular vesicle surface, within vesicles, and in the soluble fraction. These changes were most prominent in women with proven intra-amniotic infection. 2) Cytokine changes on the surface of extracellular vesicles were correlated with those determined in the soluble fraction; yet the magnitude of the increase was significantly different between these compartments. 3) The performance of prediction models of early preterm delivery based on measurements on the extracellular vesicle surface was equivalent to those based on the soluble fraction. Conclusions Differential packaging of amniotic fluid cytokines in extracellular vesicles during preterm labor with sterile intra-amniotic inflammation or proven intra-amniotic infection is reported herein for the first time. The current study provides insights into the biology of the intra-amniotic fluid ad may aid in the development of biomarkers for obstetrical disease.
Collapse
Affiliation(s)
- Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit Michigan, United States of America
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, Michigan, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit Michigan, United States of America
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Detroit Medical Center, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, United States of America
- * E-mail: (RR); (GER); (ALT)
| | - Gregory Edward Rice
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
- * E-mail: (RR); (GER); (ALT)
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, United States of America
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, Michigan, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, United States of America
- * E-mail: (RR); (GER); (ALT)
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, United States of America
| |
Collapse
|
17
|
Kaphan E, Laurin D, Lafeuillade B, Drillat P, Park S. Impact of transfusion on survival in patients with myelodysplastic syndromes: Current knowledge, new insights and transfusion clinical practice. Blood Rev 2019; 41:100649. [PMID: 31918886 DOI: 10.1016/j.blre.2019.100649] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/25/2019] [Accepted: 12/05/2019] [Indexed: 02/01/2023]
Abstract
Red Blood Cell (RBC) transfusion dependence is a prevalent consequence of anaemia in patients with lower risk Myelodysplastic Syndromes (MDS). These patients have shorter survival compared to patients responding to Erythropoiesis-stimulating agents (ESA), raising the question of potential negative effects of chronic RBC transfusions on MDS prognosis, independently of IPSS-R. Besides commonly identified complications of transfusions like iron toxicity or cardiac events, oxidative stress could be a risk factor for ineffective haematopoiesis. Recently, physicochemical changes of RBC during storage have been described. These changes called storage lesions could play a role in immunomodulation in vivo. We review the currently identified sources of potential impact on transfusion-associated effects in MDS patients and we discuss the unexplored potential role of erythrocyte-derived-extracellular vesicles. They could amplify impairment of haematopoiesis in addition to the negative intrinsic effects underlying the pathology in MDS. Thus, chronic RBC transfusions appear to potentially impact the outcome of MDS.
Collapse
Affiliation(s)
- Eléonore Kaphan
- Service d'Hématologie, CHU de Grenoble, CS 10 217, Grenoble Cedex 09 38043, France.
| | - David Laurin
- Département scientifique, Etablissement Français du Sang Auvergne Rhône-Alpes, La Tronche, France; Institute for Advanced Biosciences, Equipe Pathologie Moléculaire des Cancers et Biomarqueurs, Université Grenoble Alpes, INSERM U1209 & CNRS UMR 5309, France
| | - Bruno Lafeuillade
- Service d'Hématologie, CHU de Grenoble, CS 10 217, Grenoble Cedex 09 38043, France
| | - Philippe Drillat
- Service d'Hématologie, CHU de Grenoble, CS 10 217, Grenoble Cedex 09 38043, France; Département scientifique, Etablissement Français du Sang Auvergne Rhône-Alpes, La Tronche, France
| | - Sophie Park
- Service d'Hématologie, CHU de Grenoble, CS 10 217, Grenoble Cedex 09 38043, France; Institute for Advanced Biosciences, Equipe Pathologie Moléculaire des Cancers et Biomarqueurs, Université Grenoble Alpes, INSERM U1209 & CNRS UMR 5309, France.
| |
Collapse
|
18
|
Blood manufacturing methods affect red blood cell product characteristics and immunomodulatory activity. Blood Adv 2019; 2:2296-2306. [PMID: 30217795 DOI: 10.1182/bloodadvances.2018021931] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022] Open
Abstract
Transfusion of red cell concentrates (RCCs) is associated with increased risk of adverse outcomes that may be affected by different blood manufacturing methods and the presence of extracellular vesicles (EVs). We investigated the effect of different manufacturing methods on hemolysis, residual cells, cell-derived EVs, and immunomodulatory effects on monocyte activity. Thirty-two RCC units produced using whole blood filtration (WBF), red cell filtration (RCF), apheresis-derived (AD), and whole blood-derived (WBD) methods were examined (n = 8 per method). Residual platelet and white blood cells (WBCs) and the concentration, cell of origin, and characterization of EVs in RCC supernatants were assessed in fresh and stored supernatants. Immunomodulatory activity of RCC supernatants was assessed by quantifying monocyte cytokine production capacity in an in vitro transfusion model. RCF units yielded the lowest number of platelet and WBC-derived EVs, whereas the highest number of platelet EVs was in AD (day 5) and in WBD (day 42). The number of small EVs (<200 nm) was greater than large EVs (≥200 nm) in all tested supernatants, and the highest level of small EVs were in AD units. Immunomodulatory activity was mixed, with evidence of both inflammatory and immunosuppressive effects. Monocytes produced more inflammatory interleukin-8 after exposure to fresh WBF or expired WBD supernatants. Exposure to supernatants from AD and WBD RCC suppressed monocyte lipopolysaccharide-induced cytokine production. Manufacturing methods significantly affect RCC unit EV characteristics and are associated with an immunomodulatory effect of RCC supernatants, which may affect the quality and safety of RCCs.
Collapse
|
19
|
Stolla M, Zhang F, Meyer MR, Zhang J, Dong JF. Current state of transfusion in traumatic brain injury and associated coagulopathy. Transfusion 2019; 59:1522-1528. [PMID: 30980753 DOI: 10.1111/trf.15169] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/10/2018] [Accepted: 11/17/2018] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury (TBI)-induced coagulopathy has long been recognized as a significant risk for poor outcomes in patients with TBI, but its pathogenesis remains poorly understood. As a result, current treatment options for the condition are limited and ineffective. The lack of information is most significant for the impact of blood transfusions on patients with isolated TBI and in the absence of confounding influences from trauma to the body and limbs and the resultant hemorrhagic shock. Here we discuss recent progress in understanding the pathogenesis of TBI-induced coagulopathy and the current state of blood transfusions for patients with TBI and associated coagulopathy.
Collapse
Affiliation(s)
- Moritz Stolla
- Bloodworks Research Institute, Seattle, Washington.,Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, Washington
| | - Fangyi Zhang
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Michael R Meyer
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Jianning Zhang
- Tianjin Institute of Neurology, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, Washington.,Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, Washington
| |
Collapse
|
20
|
Menocha S, Muszynski JA. Transfusion-related immune modulation: functional consequence of extracellular vesicles? Transfusion 2019; 59:3553-3555. [PMID: 31322730 DOI: 10.1111/trf.15461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are small, subcellular vesicles that are released from a variety of cells and play important roles in cell-to-cell communication. Transfused blood products, including red blood cell, platelet, and plasma products contain EVs that are capable of interacting with and altering immune cell function. The extent to which EVs may contribute to clinically meaningful immunomodulatory effects of transfusion remains unclear and deserving of further study.
Collapse
Affiliation(s)
- Somaang Menocha
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Jennifer A Muszynski
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
21
|
Abstract
BACKGROUND Restoration of a balanced innate immune response is paramount to recovery from critical injury. Plasma transfusion may modulate innate immune responses; however, little is known about the immunomodulatory potential of various plasma products. We conducted in vitro experiments to determine the effects of fresh frozen plasma, thawed plasma, solvent/detergent plasma, and an investigational spray-dried solvent/detergent plasma product on monocyte function. METHODS Monocytes were isolated from healthy adult volunteers and cocultured with aliquots of autologous plasma (control), fresh frozen plasma, thawed plasma, solvent/detergent treated plasma, or spray-dried solvent/detergent plasma. Monocyte function was assessed by cytokine production with and without lipopolysaccharide (LPS) stimulation, and flow cytometric assessment of HLA-DR cell surface expression. RESULTS Monocyte cytokine production was not significantly altered after exposure to fresh frozen plasma or thawed plasma. In the absence of LPS, spray-dried solvent/detergent plasma exposure resulted in markedly increased IL-8 production compared to other plasma groups and controls (p = 0.01, analysis of variance [ANOVA]). Likewise, spray-dried SD plasma exposure resulted in higher LPS-induced IL-8, TNFα, and IL-1β production compared with autologous plasma controls (p < 0.0001; p < 0.0001, p = 0.002, respectively; ANOVA). LPS-induced IL-8 and TNFα production was lowest after exposure to solvent/detergent plasma (p < 0.0001, ANOVA). CONCLUSION Exposure to spray-dried solvent/detergent plasma resulted in marked augmentation of monocyte inflammatory cytokine production. Solvent/detergent plasma exposure resulted in the lowest cytokine production, suggesting lower immunomodulatory potential. Further work is needed to determine how these in vitro findings may translate to the bedside.
Collapse
|
22
|
Ng MSY, Suen JY, Tung JP, Fraser JF. Endothelialized flow models for blood transfusion research. Haematologica 2019; 104:428-434. [PMID: 30765473 PMCID: PMC6395319 DOI: 10.3324/haematol.2018.205203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/15/2019] [Indexed: 01/21/2023] Open
Affiliation(s)
- Monica S Y Ng
- Critical Care Research Group, Faculty of Medicine, University of Queensland, Brisbane
- Research and Development, Australian Red Cross Blood Service, Brisbane, Australia
| | - Jacky Y Suen
- Critical Care Research Group, Faculty of Medicine, University of Queensland, Brisbane
| | - John-Paul Tung
- Critical Care Research Group, Faculty of Medicine, University of Queensland, Brisbane
- Research and Development, Australian Red Cross Blood Service, Brisbane, Australia
| | - John F Fraser
- Critical Care Research Group, Faculty of Medicine, University of Queensland, Brisbane
| |
Collapse
|
23
|
Słomka A, Urban SK, Lukacs-Kornek V, Żekanowska E, Kornek M. Large Extracellular Vesicles: Have We Found the Holy Grail of Inflammation? Front Immunol 2018; 9:2723. [PMID: 30619239 PMCID: PMC6300519 DOI: 10.3389/fimmu.2018.02723] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
The terms microparticles (MPs) and microvesicles (MVs) refer to large extracellular vesicles (EVs) generated from a broad spectrum of cells upon its activation or death by apoptosis. The unique surface antigens of MPs/MVs allow for the identification of their cellular origin as well as its functional characterization. Two basic aspects of MP/MV functions in physiology and pathological conditions are widely considered. Firstly, it has become evident that large EVs have strong procoagulant properties. Secondly, experimental and clinical studies have shown that MPs/MVs play a crucial role in the pathophysiology of inflammation-associated disorders. A cardinal feature of these disorders is an enhanced generation of platelets-, endothelial-, and leukocyte-derived EVs. Nevertheless, anti-inflammatory effects of miscellaneous EV types have also been described, which provided important new insights into the large EV-inflammation axis. Advances in understanding the biology of MPs/MVs have led to the preparation of this review article aimed at discussing the association between large EVs and inflammation, depending on their cellular origin.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Sabine Katharina Urban
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Ewa Żekanowska
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Miroslaw Kornek
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
24
|
Schonewille H, van Rood JJ, Verduin EP, van de Watering LMG, Haasnoot GW, Claas FHJ, Oepkes D, Lopriore E, Brand A. Exposure to non-inherited maternal antigens by breastfeeding affects antibody responsiveness. Haematologica 2018; 104:263-268. [PMID: 30213833 PMCID: PMC6355501 DOI: 10.3324/haematol.2018.199406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/11/2018] [Indexed: 12/02/2022] Open
Abstract
The observation, by Ray Owen and colleagues in 1954, that D-negative women were less likely to form anti-D antibodies against their D-positive fetus if their mother possessed the D-antigen, was not found in all later studies. We hypothesized that breastfeeding, received by the mother, may affect her immunity against non-inherited maternal red blood cell antigens. We studied a cohort of 125 grandmother-mother-child combinations, from a follow-up study of mothers after intrauterine transfusion of the fetus for alloimmune hemolytic disease. For mismatched red blood cell antigens the mother was exposed to, whether or not antibodies were formed, we determined whether her mother, the grandmother, carried these antigens. The duration for which the mothers were breastfed was estimated by way of a questionnaire. Using multivariate logistic regression analyses, the interaction term (non-inherited maternal antigen exposure by categorized breastfeeding period) showed that a longer breastfeeding period was associated with decreased alloimmunization against non-inherited maternal antigens (adjusted odds ratio 0.66; 95% confidence interval 0.48–0.93). Sensitivity analysis with dichotomized (shorter versus longer) breastfeeding periods showed that this lower risk was reached after two months (aOR 0.22; 95% CI 0.07–0.71) and longer duration of breastfeeding did not seem to provide additional protection. These data suggest that oral neonatal exposure to non-inherited maternal red blood cell antigens through breastfeeding for at least two months diminishes the risk of alloimmunization against these antigens when encountered later in life.
Collapse
Affiliation(s)
- Henk Schonewille
- Center for Clinical Transfusion Research, Sanquin Research, Leiden .,Jon J van Rood Center for Clinical Transfusion Research, Sanquin-Leiden University Medical Center, Amsterdam
| | - Jon J van Rood
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - Esther P Verduin
- Center for Clinical Transfusion Research, Sanquin Research, Leiden.,Jon J van Rood Center for Clinical Transfusion Research, Sanquin-Leiden University Medical Center, Amsterdam.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - Leo M G van de Watering
- Center for Clinical Transfusion Research, Sanquin Research, Leiden.,Jon J van Rood Center for Clinical Transfusion Research, Sanquin-Leiden University Medical Center, Amsterdam
| | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - Dick Oepkes
- Division of Fetal Medicine, Department of Obstetrics, Leiden University Medical Center
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, the Netherlands
| | - Anneke Brand
- Center for Clinical Transfusion Research, Sanquin Research, Leiden.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center
| |
Collapse
|
25
|
Leal JKF, Adjobo-Hermans MJW, Bosman GJCGM. Red Blood Cell Homeostasis: Mechanisms and Effects of Microvesicle Generation in Health and Disease. Front Physiol 2018; 9:703. [PMID: 29937736 PMCID: PMC6002509 DOI: 10.3389/fphys.2018.00703] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022] Open
Abstract
Red blood cells (RBCs) generate microvesicles to remove damaged cell constituents such as oxidized hemoglobin and damaged membrane constituents, and thereby prolong their lifespan. Damage to hemoglobin, in combination with altered phosphorylation of membrane proteins such as band 3, lead to a weakening of the binding between the lipid bilayer and the cytoskeleton, and thereby to membrane budding and microparticle shedding. Microvesicle generation is disturbed in patients with RBC-centered diseases, such as sickle cell disease, glucose 6-phosphate dehydrogenase deficiency, spherocytosis or malaria. A disturbance of the membrane-cytoskeleton interaction is likely to be the main underlying mechanism, as is supported by data obtained from RBCs stored in blood bank conditions. A detailed proteomic, lipidomic and immunogenic comparison of microvesicles derived from different sources is essential in the identification of the processes that trigger vesicle generation. The contribution of RBC-derived microvesicles to inflammation, thrombosis and autoimmune reactions emphasizes the need for a better understanding of the mechanisms and consequences of microvesicle generation.
Collapse
Affiliation(s)
- Joames K F Leal
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Giel J C G M Bosman
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
26
|
Danesh A, Inglis HC, Abdel-Mohsen M, Deng X, Adelman A, Schechtman KB, Heitman JW, Vilardi R, Shah A, Keating SM, Cohen MJ, Jacobs ES, Pillai SK, Lacroix J, Spinella PC, Norris PJ. Granulocyte-Derived Extracellular Vesicles Activate Monocytes and Are Associated With Mortality in Intensive Care Unit Patients. Front Immunol 2018; 9:956. [PMID: 29867942 PMCID: PMC5951932 DOI: 10.3389/fimmu.2018.00956] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/17/2018] [Indexed: 11/13/2022] Open
Abstract
To understand how extracellular vesicle (EV) subtypes differentially activate monocytes, a series of in vitro studies were performed. We found that plasma-EVs biased monocytes toward an M1 profile. Culturing monocytes with granulocyte-, monocyte-, and endothelial-EVs induced several pro-inflammatory cytokines. By contrast, platelet-EVs induced TGF-β and GM-CSF, and red blood cell (RBC)-EVs did not activate monocytes in vitro. The scavenger receptor CD36 was important for binding of RBC-EVs to monocytes, while blockade of CD36, CD163, CD206, TLR1, TLR2, and TLR4 did not affect binding of plasma-EVs to monocytes in vitro. To identify mortality risk factors, multiple soluble factors and EV subtypes were measured in patients' plasma at intensive care unit admission. Of 43 coagulation factors and cytokines measured, two were significantly associated with mortality, tissue plasminogen activator and cystatin C. Of 14 cellular markers quantified on EVs, 4 were early predictors of mortality, including the granulocyte marker CD66b. In conclusion, granulocyte-EVs have potent pro-inflammatory effects on monocytes in vitro. Furthermore, correlation of early granulocyte-EV levels with mortality in critically ill patients provides a potential target for intervention in management of the pro-inflammatory cascade associated with critical illness.
Collapse
Affiliation(s)
- Ali Danesh
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Heather C Inglis
- Blood Systems Research Institute, San Francisco, CA, United States
| | - Mohamed Abdel-Mohsen
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Xutao Deng
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Avril Adelman
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Kenneth B Schechtman
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, United States.,Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - John W Heitman
- Blood Systems Research Institute, San Francisco, CA, United States
| | - Ryan Vilardi
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Avani Shah
- Blood Systems Research Institute, San Francisco, CA, United States
| | - Sheila M Keating
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mitchell J Cohen
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Evan S Jacobs
- Blood Systems Research Institute, San Francisco, CA, United States
| | - Satish K Pillai
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jacques Lacroix
- Centre Hospitalier Universitaire (CHU) Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Philip C Spinella
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Philip J Norris
- Blood Systems Research Institute, San Francisco, CA, United States.,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States.,Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
27
|
Bal SH, Heper Y, Kumaş LT, Guvenc F, Budak F, Göral G, Oral HB. Effect of storage period of red blood cell suspensions on helper T-cell subpopulations. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2018; 16:262-272. [PMID: 28488961 PMCID: PMC5919838 DOI: 10.2450/2017.0238-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND The aim of this study was to investigate the immunological alterations that occur during the storage of erythrocyte suspensions which may lead to transfusion-related immunomodulation following allogeneic blood transfusion. MATERIALS AND METHODS One part of the erythrocyte suspensions obtained from donors was leucoreduced while the other part was not. The leucoreduced (LR) and non-leucoreduced (NL) erythrocyte suspensions were then further divided into three equal amounts which were stored for 0, 21 or 42 days prior to measurements, by enzyme-linked immunosorbent assays, of cytokine levels in their supernatants. T-helper (Th) lymphocyte subgroups and gene expression were analysed in the NL erythrocyte suspensions by flow cytometry and real-time polymerase chain reaction, respectively. Results were compared to those of storage day 0. RESULTS By day 21, the number of Th2 cells had increased significantly and the numbers of Th1, Th22 and Treg cells had decreased significantly in the NL erythrocyte suspensions. On day 42 the numbers of Th2 and Treg cells in the NL suspensions were significantly increased while the number of Th1 cells was significantly decreased. The levels of transcription factors (TBX21, GATA3, and SPI.1) were significantly decreased on days 21 and 42, and AHR, FOXP3 and RORC2 levels were significantly increased on day 42 in NL erythrocyte suspensions. The decrease in interleukin-22 and increase in transforming growth factor-β levels found in NL erythrocyte suspensions on day 21 were statistically significant. Elevated levels of interleukin-17A were found in both LR and NL erythrocyte suspensions on day 42. DISCUSSION Our results suggest that allogeneic leucocytes and cytokines may play significant roles in the development of transfusion-related immunomodulation.
Collapse
Affiliation(s)
- Salih H Bal
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Microbiology/Immunology, Institute of Health Sciences, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Yasemin Heper
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Levent T Kumaş
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Microbiology/Immunology, Institute of Health Sciences, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Furkan Guvenc
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Ferah Budak
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Güher Göral
- Department of Medical Microbiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Haluk B Oral
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
28
|
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: 16.0] [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
|
29
|
Remy KE, Hall MW, Cholette J, Juffermans NP, Nicol K, Doctor A, Blumberg N, Spinella PC, Norris PJ, Dahmer MK, Muszynski JA. Mechanisms of red blood cell transfusion-related immunomodulation. Transfusion 2018; 58:804-815. [PMID: 29383722 DOI: 10.1111/trf.14488] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/13/2017] [Accepted: 12/10/2017] [Indexed: 01/28/2023]
Abstract
Red blood cell (RBC) transfusion is common in critically ill, postsurgical, and posttrauma patients in whom both systemic inflammation and immune suppression are associated with adverse outcomes. RBC products contain a multitude of immunomodulatory mediators that interact with and alter immune cell function. These interactions can lead to both proinflammatory and immunosuppressive effects. Defining clinical outcomes related to immunomodulatory effects of RBCs in transfused patients remains a challenge, likely due to complex interactions between individual blood product characteristics and patient-specific risk factors. Unpacking these complexities requires an in-depth understanding of the mechanisms of immunomodulatory effects of RBC products. In this review, we outline and classify potential mediators of RBC transfusion-related immunomodulation and provide suggestions for future research directions.
Collapse
Affiliation(s)
- Kenneth E Remy
- Department of Pediatrics, Division of Pediatric Critical Care, Washington University School of Medicine, St Louis, Missouri
| | - Mark W Hall
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio.,The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Jill Cholette
- Pediatric Critical Care and Cardiology, University of Rochester, Rochester, New York
| | - Nicole P Juffermans
- Department of Intensive Care Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Kathleen Nicol
- Department of Pathology, Nationwide Children's Hospital, Columbus, Ohio
| | - Allan Doctor
- Department of Pediatrics, Division of Pediatric Critical Care, Washington University School of Medicine, St Louis, Missouri
| | - Neil Blumberg
- Transfusion Medicine/Blood Bank and Clinical Laboratories, Departments of Pathology and Laboratory Medicine, University of Rochester, Rochester, New York
| | - Philip C Spinella
- Department of Pediatrics, Division of Pediatric Critical Care, Washington University School of Medicine, St Louis, Missouri
| | - Philip J Norris
- Blood Systems Research Institute, San Francisco, California.,Departments of Laboratory Medicine and Medicine, University of California at San Francisco, San Francisco, California
| | - Mary K Dahmer
- Department of Pediatrics, Division of Pediatric Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Jennifer A Muszynski
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio.,The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | | |
Collapse
|
30
|
Said AS, Rogers SC, Doctor A. Physiologic Impact of Circulating RBC Microparticles upon Blood-Vascular Interactions. Front Physiol 2018; 8:1120. [PMID: 29379445 PMCID: PMC5770796 DOI: 10.3389/fphys.2017.01120] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Here, we review current data elucidating the role of red blood cell derived microparticles (RMPs) in normal vascular physiology and disease progression. Microparticles (MPs) are submicron-size, membrane-encapsulated vesicles derived from various parent cell types. MPs are produced in response to numerous stimuli that promote a sequence of cytoskeletal and membrane phospholipid changes and resulting MP genesis. MPs were originally considered as potential biomarkers for multiple disease processes and more recently are recognized to have pleiotropic biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in initiating apoptosis. RMPs, specifically, form normally during RBC maturation in response to injury during circulation, and are copiously produced during processing and storage for transfusion. Notably, several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs markedly from that of intact RBCs and the nature/composition of RMP components are affected by the specific circumstances of RMP genesis. Described RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion as well as influence upon vasoregulation via influence upon nitric oxide (NO) bioavailability. Of particular relevance, RMPs scavenge NO more avidly than do intact RBCs; this physiology has been proposed to contribute to the impaired oxygen delivery homeostasis that may be observed following transfusion. In summary, RMPs are submicron particles released from RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in normal and patho-physiology and in transfusion recipients is an area of continued investigation.
Collapse
Affiliation(s)
- Ahmed S Said
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Stephen C Rogers
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Allan Doctor
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States.,Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
31
|
Vostal JG, Buehler PW, Gelderman MP, Alayash AI, Doctor A, Zimring JC, Glynn SA, Hess JR, Klein H, Acker JP, Spinella PC, D'Alessandro A, Palsson B, Raife TJ, Busch MP, McMahon TJ, Intaglietta M, Swartz HM, Dubick MA, Cardin S, Patel RP, Natanson C, Weisel JW, Muszynski JA, Norris PJ, Ness PM. Proceedings of the Food and Drug Administration's public workshop on new red blood cell product regulatory science 2016. Transfusion 2017; 58:255-266. [PMID: 29243830 DOI: 10.1111/trf.14435] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
Abstract
The US Food and Drug Administration (FDA) held a workshop on red blood cell (RBC) product regulatory science on October 6 and 7, 2016, at the Natcher Conference Center on the National Institutes of Health (NIH) Campus in Bethesda, Maryland. The workshop was supported by the National Heart, Lung, and Blood Institute, NIH; the Department of Defense; the Office of the Assistant Secretary for Health, Department of Health and Human Services; and the Center for Biologics Evaluation and Research, FDA. The workshop reviewed the status and scientific basis of the current regulatory framework and the available scientific tools to expand it to evaluate innovative and future RBC transfusion products. A full record of the proceedings is available on the FDA website (http://www.fda.gov/BiologicsBloodVaccines/NewsEvents/WorkshopsMeetingsConferences/ucm507890.htm). The contents of the summary are the authors' opinions and do not represent agency policy.
Collapse
Affiliation(s)
- Jaroslav G Vostal
- Division of Blood Components and Devices, OBRR, CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Paul W Buehler
- Division of Blood Components and Devices, OBRR, CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Monique P Gelderman
- Division of Blood Components and Devices, OBRR, CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Abdu I Alayash
- Division of Blood Components and Devices, OBRR, CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Alan Doctor
- Department of Pediatric Critical Care, St Louis Children's Hospital, St Louis, Missouri
| | | | - Simone A Glynn
- Division of Blood Diseases and Resources, NHLBI, NIH, Bethesda, Maryland
| | - John R Hess
- Department of Laboratory Medicine and Hematology, University of Washington, School of Medicine, Seattle, Washington
| | - Harvey Klein
- Department of Transfusion Medicine, National Institutes of Health, Clinical Center, Bethesda, Maryland
| | - Jason P Acker
- Department of Research & Development, Canadian Blood Services, Edmonton, Alberta, Canada
| | - Philip C Spinella
- Department of Pediatric Critical Care, Washington University School of Medicine, St Louis, Missouri
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Denver, Colorado
| | - Bernhard Palsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | - Thomas J Raife
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Timothy J McMahon
- Department of Medicine, Pulmonary, Allergy, & Critical Care Medicine, Duke University Medical Center, and the Durham VA Medical Center, Durham, North Carolina
| | - Marcos Intaglietta
- Department of Bioengineering, University of California at San Diego, San Diego, California
| | - Harold M Swartz
- Department of Radiology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire
| | | | - Sylvain Cardin
- Naval Medical Research Unit-San Antonio, San Antonio, Texas
| | - Rakesh P Patel
- Center for Free Radical Biology and Translational and Molecular Sciences Certificate Program, University of Alabama, Birmingham, Alabama
| | | | - John W Weisel
- Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer A Muszynski
- Division of Critical Care Medicine, The Ohio State University College of Medicine, Columbus, Ohio
| | - Philip J Norris
- Blood Systems Research Institute, Blood Systems, Inc., San Francisco, California
| | - Paul M Ness
- Division of Transfusion Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Barteneva NS, Baiken Y, Fasler-Kan E, Alibek K, Wang S, Maltsev N, Ponomarev ED, Sautbayeva Z, Kauanova S, Moore A, Beglinger C, Vorobjev IA. Extracellular vesicles in gastrointestinal cancer in conjunction with microbiota: On the border of Kingdoms. Biochim Biophys Acta Rev Cancer 2017; 1868:372-393. [DOI: 10.1016/j.bbcan.2017.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/26/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022]
|
33
|
Laurén E, Tigistu-Sahle F, Valkonen S, Westberg M, Valkeajärvi A, Eronen J, Siljander P, Pettilä V, Käkelä R, Laitinen S, Kerkelä E. Phospholipid composition of packed red blood cells and that of extracellular vesicles show a high resemblance and stability during storage. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:1-8. [PMID: 28965917 DOI: 10.1016/j.bbalip.2017.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 09/07/2017] [Accepted: 09/24/2017] [Indexed: 01/25/2023]
Abstract
Red blood cells (RBCs) are stored up to 35-42days at 2-6°C in blood banks. During storage, the RBC membrane is challenged by energy depletion, decreasing pH, altered cation homeostasis, and oxidative stress, leading to several biochemical and morphological changes in RBCs and to shedding of extracellular vesicles (EVs) into the storage medium. These changes are collectively known as RBC storage lesions. EVs accumulate in stored RBC concentrates and are, thus, transfused into patients. The potency of EVs as bioactive effectors is largely acknowledged, and EVs in RBC concentrates are suspected to mediate some adverse effects of transfusion. Several studies have shown accumulation of lipid raft-associated proteins in RBC EVs during storage, whereas a comprehensive phospholipidomic study on RBCs and corresponding EVs during the clinical storage period is lacking. Our mass spectrometric and chromatographic study shows that RBCs maintain their major phospholipid (PL) content well during storage despite abundant vesiculation. The phospholipidomes were largely similar between RBCs and EVs. No accumulation of raft lipids in EVs was seen, suggesting that the primary mechanism of RBC vesiculation during storage might not be raft -based. Nonetheless, a slight tendency of EV PLs for shorter acyl chains was observed.
Collapse
Affiliation(s)
- Eva Laurén
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland; Department of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Feven Tigistu-Sahle
- University of Helsinki, Department of Biosciences, Division of Physiology and Neuroscience, Helsinki, Finland
| | - Sami Valkonen
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland; University of Helsinki, Department of Biosciences, Division of Biochemistry and Biotechnology, Helsinki, Finland; University of Helsinki, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Helsinki, Finland
| | - Melissa Westberg
- University of Helsinki, Department of Biosciences, Division of Physiology and Neuroscience, Helsinki, Finland
| | - Anne Valkeajärvi
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland
| | - Juha Eronen
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland
| | - Pia Siljander
- University of Helsinki, Department of Biosciences, Division of Biochemistry and Biotechnology, Helsinki, Finland; University of Helsinki, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Helsinki, Finland
| | - Ville Pettilä
- Department of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Reijo Käkelä
- University of Helsinki, Department of Biosciences, Division of Physiology and Neuroscience, Helsinki, Finland
| | - Saara Laitinen
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland.
| |
Collapse
|
34
|
Influence of red blood cell-derived microparticles upon vasoregulation. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2017; 15:522-534. [PMID: 28686154 DOI: 10.2450/2017.0353-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022]
Abstract
Here we review recent data and the evolving understanding of the role of red blood cell-derived microparticles (RMPs) in normal physiology and in disease progression. Microparticles (MPs) are small membrane vesicles derived from various parent cell types. MPs are produced in response to a variety of stimuli through several cytoskeletal and membrane phospholipid changes. MPs have been investigated as potential biomarkers for multiple disease processes and are thought to have biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in apoptosis. Specifically, RMPs are produced normally during RBC maturation and their production is accelerated during processing and storage for transfusion. Several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs from that of intact RBCs, and the nature and composition of RMP components are affected by both storage duration and the character of storage solutions. Recognised RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion, as well as influence upon vasoregulation via nitric oxide (NO) scavenging. Of particular relevance, RMPs are more avid NO scavengers than intact RBCs and this feature has been proposed as a mechanism for the impaired oxygen delivery homeostasis that has been observed following transfusion. Preliminary human studies demonstrate that circulating RMP abundance increases with RBC transfusion and is associated with altered plasma vasoactivity and abnormal vasoregulation. In summary, RMPs are submicron particles released from stored RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in transfusion recipients is an area of continued investigation.
Collapse
|
35
|
Foster BP, Balassa T, Benen TD, Dominovic M, Elmadjian GK, Florova V, Fransolet MD, Kestlerova A, Kmiecik G, Kostadinova IA, Kyvelidou C, Meggyes M, Mincheva MN, Moro L, Pastuschek J, Spoldi V, Wandernoth P, Weber M, Toth B, Markert UR. Extracellular vesicles in blood, milk and body fluids of the female and male urogenital tract and with special regard to reproduction. Crit Rev Clin Lab Sci 2016; 53:379-95. [PMID: 27191915 DOI: 10.1080/10408363.2016.1190682] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are released from almost all cells and tissues. They are able to transport substances (e.g. proteins, RNA or DNA) at higher concentrations than in their environment and may adhere in a receptor-controlled manner to specific cells or tissues in order to release their content into the respective target structure. Blood contains high concentrations of EVs mainly derived from platelets, and, at a smaller amount, from erythrocytes. The female and male reproductive tracts produce EVs which may be associated with fertility or infertility and are released into body fluids and mucosas of the urogenital organs. In this review, the currently relevant detection methods are presented and critically compared. During pregnancy, placenta-derived EVs are dynamically detectable in peripheral blood with changing profiles depending upon progress of pregnancy and different pregnancy-associated pathologies, such as preeclampsia. EVs offer novel non-invasive diagnostic tools which may reflect the situation of the placenta and the foetus. EVs in urine have the potential of reflecting urogenital diseases including cancers of the neighbouring organs. Several methods for detection, quantification and phenotyping of EVs have been established, which include electron microscopy, flow cytometry, ELISA-like methods, Western blotting and analyses based on Brownian motion. This review article summarises the current knowledge about EVs in blood and cord blood, in the different compartments of the male and female reproductive tracts, in trophoblast cells from normal and pre-eclamptic pregnancies, in placenta ex vivo perfusate, in the amniotic fluid, and in breast milk, as well as their potential effects on natural killer cells as possible targets.
Collapse
Affiliation(s)
- B P Foster
- a Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, and Manchester Academic Health Sciences Centre, University Research , Manchester , UK
| | - T Balassa
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - T D Benen
- c Microtrac GmbH , Krefeld , Germany
| | - M Dominovic
- d Department of Physiology and Immunology , Medical Faculty, University of Rijeka , Rijeka , Croatia
| | - G K Elmadjian
- e Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - V Florova
- f Department of Obstetrics , Gynecology and Perinatology, First Moscow State Medical University , Moscow , Russia
| | - M D Fransolet
- g Laboratory of Tumor and Development Biology , GIGA-R, University of Liège , Liège , Belgium
| | - A Kestlerova
- h Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine , Charles University Prague , Czech Republic
- i Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - G Kmiecik
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - I A Kostadinova
- k Department of Immunoneuroendocrinology , Institute of Biology and Immunology of Reproduction , Sofia , Bulgaria
| | - C Kyvelidou
- l Department of Biology , University of Crete , Crete , Greece
| | - M Meggyes
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - M N Mincheva
- m Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - L Moro
- n ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic- Universitat de Barcelona , Barcelona , Spain
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - J Pastuschek
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - V Spoldi
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - P Wandernoth
- p Institute of Anatomy, University Hospital, University Duisburg-Essen , Essen , Germany
| | - M Weber
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - B Toth
- q Department of Gynecological Endocrinology and Fertility Disorders , Ruprecht-Karls University of Heidelberg , Heidelberg , Germany
| | - U R Markert
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| |
Collapse
|
36
|
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: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Sadallah S, Schmied L, Eken C, Charoudeh HN, Amicarella F, Schifferli JA. Platelet-Derived Ectosomes Reduce NK Cell Function. THE JOURNAL OF IMMUNOLOGY 2016; 197:1663-71. [DOI: 10.4049/jimmunol.1502658] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/21/2016] [Indexed: 11/19/2022]
|
38
|
Speth JM, Bourdonnay E, Penke LRK, Mancuso P, Moore BB, Weinberg JB, Peters-Golden M. Alveolar Epithelial Cell-Derived Prostaglandin E2 Serves as a Request Signal for Macrophage Secretion of Suppressor of Cytokine Signaling 3 during Innate Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 196:5112-20. [PMID: 27183597 DOI: 10.4049/jimmunol.1502153] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/15/2016] [Indexed: 01/22/2023]
Abstract
Preservation of gas exchange mandates that the pulmonary alveolar surface restrain unnecessarily harmful inflammatory responses to the many challenges to which it is exposed. These responses reflect the cross-talk between alveolar epithelial cells (AECs) and resident alveolar macrophages (AMs). We recently determined that AMs can secrete suppressor of cytokine signaling (SOCS) proteins within microparticles. Uptake of these SOCS-containing vesicles by epithelial cells inhibits cytokine-induced STAT activation. However, the ability of epithelial cells to direct AM release of SOCS-containing vesicles in response to inflammatory insults has not been studied. In this study, we report that SOCS3 protein was elevated in bronchoalveolar lavage fluid of both virus- and bacteria-infected mice, as well as in an in vivo LPS model of acute inflammation. In vitro studies revealed that AEC-conditioned medium (AEC-CM) enhanced AM SOCS3 secretion above basal levels. Increased amounts of PGE2 were present in AEC-CM after LPS challenge, and both pharmacologic inhibition of PGE2 synthesis in AECs and neutralization of PGE2 in AEC-CM implicated this prostanoid as the major AEC-derived factor mediating enhanced AM SOCS3 secretion. Moreover, pharmacologic blockade of PGE2 synthesis or genetic deletion of a PGE2 synthase similarly attenuated the increase in bronchoalveolar lavage fluid SOCS3 noted in lungs of mice challenged with LPS in vivo. These results demonstrate a novel tunable form of cross-talk in which AECs use PGE2 as a signal to request SOCS3 from AMs to dampen their endogenous inflammatory responses during infection.
Collapse
Affiliation(s)
- Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Emilie Bourdonnay
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Loka Raghu Kumar Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jason B Weinberg
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and Division of Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109;
| |
Collapse
|
39
|
All plasma products are not created equal: Characterizing differences between plasma products. J Trauma Acute Care Surg 2015; 78:S18-25. [PMID: 26002258 DOI: 10.1097/ta.0000000000000629] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Plasma can be manufactured by multiple methods. Few studies have compared quality parameters between plasma products that may affect efficacy and safety. METHODS Four different plasma products were analyzed to include fresh frozen plasma (FFP), liquid plasma (LP), solvent detergent plasma (SDP), and a spray-dried, solvent detergent-treated plasma (SD-SDP) at multiple time points of storage. Parameters measured included red blood cell, platelet, and white blood cell counts; microparticle phenotypes; thrombin generation; and thrombelastography. These parameters were compared in 10 samples of each product. RESULTS SDP and SD-SDP contained the smallest number of residual cells compared with FFP and LP. Platelets were the most common residual cell in all products and were highest in LP. FFP contained the greatest number of residual red blood cells. Total microparticle counts were elevated in LP and FFP compared with SDP and SD-SDP. Cell-derived microparticles in both LP and FFP were mostly platelet in origin. Microparticle counts in SDP and SD-SDP were negligible. Thrombelastography results demonstrated similar thrombin, fibrinogen, and platelet function on Day 28 LP compared with Day 5 thawed FFP. Thrombin generation assays revealed that the total, lag time to, and peak thrombin formation were higher in SDP and SD-SDP compared with FFP and LP. All parameters in FFP and LP products were characterized by a large degree of variability. CONCLUSION The differences in cellular, microparticle, and functional hemostatic parameters measured between plasma products have the potential to affect efficacy and safety. Further study is needed to elucidate the potential immune effects of the cellular and microparticle differences noted as well as the clinical implications of altered thrombin generation kinetics in SD products.
Collapse
|
40
|
Smith SA, Ngwenyama TR, O'Brien M, Herring JM, Corsi R, Galligan A, Beloshapka AN, Deng P, Swanson KS, McMichael M. Procoagulant phospholipid concentration in canine erythrocyte concentrates stored with or without prestorage leukoreduction. Am J Vet Res 2015; 76:35-41. [PMID: 25535659 DOI: 10.2460/ajvr.76.1.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate canine erythrocyte concentrates (ECs) for the presence of procoagulant phospholipid (PPL), determine whether PPL concentration changes during the course of storage of ECs, and ascertain whether prestorage leukoreduction (removal of leukocytes via gravity filtration) reduces the development of PPL. SAMPLE 10 whole blood units (420 g each) collected from 10 random-source, clinically normal dogs (1 U/dog). PROCEDURES The dogs were randomized to 1 of 2 groups. Of the 10 whole blood units collected, 5 were processed through a standard method, and 5 underwent leukoreduction. Whole blood units were processed to generate ECs, from which aliquots were aseptically collected from each unit weekly for 5 weeks. Supernatants from the concentrates were evaluated for procoagulant activity, which was converted to PPL concentration, by use of an automated assay and by measurement of real-time thrombin generation. RESULTS Supernatants from stored canine ECs contained procoagulant activity as measured by both assays. In general, the PPL concentration gradually increased during the storage period, but leukoreduction reduced the development of increased procoagulant activity over time. CONCLUSIONS AND CLINICAL RELEVANCE The presence of PPL in canine ECs may be associated with procoagulant and proinflammatory effects in vivo, which could have adverse consequences for dogs treated with ECs.
Collapse
Affiliation(s)
- Stephanie A Smith
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois, Urbana, IL 61801
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mittag D, Sran A, Chan KS, Boland MP, Bandala-Sanchez E, Huet O, Xu W, Sparrow RL. Stored red blood cell susceptibility to in vitro transfusion-associated stress conditions is higher after longer storage and increased by storage in saline-adenine-glucose-mannitol compared to AS-1. Transfusion 2015; 55:2197-206. [PMID: 25968419 DOI: 10.1111/trf.13138] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/23/2015] [Accepted: 02/28/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Biochemical changes induced in red blood cells (RBCs) during storage may impair their function upon transfusion. Transfusion-associated stresses may further amplify storage lesion effects including increased phosphatidylserine (PS) exposure at the RBC membrane, microparticle (MP) release, and adhesion to endothelial cells (ECs). RBC stress susceptibility in vitro was investigated in relation to storage time and additive solution. STUDY DESIGN AND METHODS Leukoreduced whole blood donations (n = 18) were paired, mixed, and resplit before separating the RBCs for storage in saline-adenine-glucose-mannitol (SAGM) or AS-1. Samples were taken after 3, 21, or 35 days. For oxidative stress treatment, RBCs were exposed to 0.5 mmol/L tert-butylhydroperoxide. Transfusion-associated stress was simulated by overnight culture at 37 °C with plasma containing inflammatory mediators. PS exposure and MPs were measured by flow cytometry and adhesion to ECs was tested under flow conditions. PS specificity of adhesion was tested by blocking with PS-containing lipid vesicles. RESULTS Oxidative stress induced significantly higher PS exposure and adhesion to ECs in RBCs stored for 35 days compared to 3 days (p < 0.04). PS-containing vesicles blocked RBC-EC adhesion. After overnight culture with or without plasma, PS exposure and EC adhesion were significantly increased (p < 0.05). MP numbers increased with longer RBC storage and after RBC culture with plasma. Culture conditions influenced MP numbers from Day 35 RBCs. RBCs stored in SAGM had significantly higher PS exposure after stress treatment than AS-1 RBCs (p < 0.02). CONCLUSION Storage for 35 days significantly increased RBC susceptibility to oxidative and in vitro transfusion-associated stresses and was higher for RBCs stored in SAGM compared to AS-1.
Collapse
Affiliation(s)
- Diana Mittag
- formerly Research and Development, Australian Red Cross Blood Service, West Melbourne, Victoria, Australia
| | - Amrita Sran
- formerly Research and Development, Australian Red Cross Blood Service, West Melbourne, Victoria, Australia
| | - Kasey S Chan
- formerly Research and Development, Australian Red Cross Blood Service, West Melbourne, Victoria, Australia
| | - Martin P Boland
- School of Psychological and Clinical Sciences, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Esther Bandala-Sanchez
- Department of Medical Biology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Olivier Huet
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria
| | - William Xu
- formerly Research and Development, Australian Red Cross Blood Service, West Melbourne, Victoria, Australia
| | - Rosemary L Sparrow
- formerly Research and Development, Australian Red Cross Blood Service, West Melbourne, Victoria, Australia.,Department of Immunology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
Bourdonnay E, Zasłona Z, Penke LRK, Speth JM, Schneider DJ, Przybranowski S, Swanson JA, Mancuso P, Freeman CM, Curtis JL, Peters-Golden M. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. ACTA ACUST UNITED AC 2015; 212:729-42. [PMID: 25847945 PMCID: PMC4419346 DOI: 10.1084/jem.20141675] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 03/17/2015] [Indexed: 12/20/2022]
Abstract
SOCS1 and -3 proteins are released by alveolar macrophages into exosomes and microparticles, respectively, which are then taken up by alveolar epithelial cells, resulting in inhibition of STAT signaling. This process was dampened by exposure to cigarette smoke and may thus be important in suppressing airway inflammation. JAK-STAT signaling mediates the actions of numerous cytokines and growth factors, and its endogenous brake is the family of SOCS proteins. Consistent with their intracellular roles, SOCS proteins have never been identified in the extracellular space. Here we report that alveolar macrophages can secrete SOCS1 and -3 in exosomes and microparticles, respectively, for uptake by alveolar epithelial cells and subsequent inhibition of STAT activation. Secretion is tunable and occurs both in vitro and in vivo. SOCS secretion into lung lining fluid was diminished by cigarette smoking in humans and mice. Secretion and transcellular delivery of vesicular SOCS proteins thus represent a new model for the control of inflammatory signaling, which is subject to dysregulation during states of inflammation.
Collapse
Affiliation(s)
- Emilie Bourdonnay
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Zbigniew Zasłona
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Loka Raghu Kumar Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Daniel J Schneider
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Sally Przybranowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Joel A Swanson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Peter Mancuso
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| | - Christine M Freeman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109 Research Services and Medical Services, Department of Veterans Affairs Health Care System, Ann Arbor, MI 48105
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109 Research Services and Medical Services, Department of Veterans Affairs Health Care System, Ann Arbor, MI 48105
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School; and Department of Environmental Health Sciences, School of Public Health; University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
43
|
Muszynski JA, Bale J, Nateri J, Nicol K, Wang Y, Wright V, Marsh CB, Gavrilin MA, Sarkar A, Wewers MD, Hall MW. Supernatants from stored red blood cell (RBC) units, but not RBC-derived microvesicles, suppress monocyte function in vitro. Transfusion 2015; 55:1937-45. [PMID: 25819532 DOI: 10.1111/trf.13084] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND We have previously shown that critically ill children transfused with red blood cells (RBCs) of longer storage durations have more suppressed monocyte function after transfusion compared to children transfused with fresher RBCs and that older stored RBCs directly suppress monocyte function in vitro, through unknown mechanisms. We hypothesized that RBC-derived microvesicles (MVs) were responsible for monocyte suppression. STUDY DESIGN AND METHODS To determine the role of stored RBC unit-derived MVs, we cocultured monocytes with supernatants, isolated MVs, or supernatants that had been depleted of MVs from prestorage leukoreduced RBCs that had been stored for either 7 or 30 days. Isolated MVs were characterized by electron microscopy and flow cytometry. Monocyte function after coculture experiments was measured by cytokine production after stimulation with lipopolysaccharide (LPS). RESULTS Monocyte function was suppressed after exposure to supernatants from 30-day RBC units compared to monocytes cultured in medium alone (LPS-induced tumor necrosis factor-α production, 17,611 ± 3,426 vs. 37,486 ± 5,598 pg/mL; p = 0.02). Monocyte function was not suppressed after exposure to MV fractions. RBC supernatants that had been depleted of MVs remained immunosuppressive. Treating RBC supernatants with heat followed by RNase (to degrade protein-bound RNA) prevented RBC supernatant-induced monocyte suppression. CONCLUSION Our findings implicate soluble mediators of stored RBC-induced monocyte suppression outside of MV fractions and suggest that extracellular protein-bound RNAs (such as microRNA) may play a role in transfusion-related immunomodulation.
Collapse
Affiliation(s)
- Jennifer A Muszynski
- Division of Critical Care Medicine.,The Research Institute.,Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | - Yijie Wang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Valerie Wright
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Clay B Marsh
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Mikhail A Gavrilin
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Anasuya Sarkar
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Mark D Wewers
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Mark W Hall
- Division of Critical Care Medicine.,The Research Institute.,Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
44
|
Crosstalk between red blood cells and the immune system and its impact on atherosclerosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:616834. [PMID: 25722984 PMCID: PMC4334626 DOI: 10.1155/2015/616834] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/16/2015] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is a chronic multifactorial disease of the arterial wall characterized by inflammation, oxidative stress, and immune system activation. Evidence exists on a pathogenic role of oxidized red blood cells (RBCs) accumulated in the lesion after intraplaque hemorrhage. This review reports current knowledge on the impact of oxidative stress in RBC modifications with the surface appearance of senescent signals characterized by reduced expression of CD47 and glycophorin A and higher externalization of phosphatidylserine. The review summarizes findings indicating that oxidized, senescent, or stored RBCs, due to surface antigen modification and release of prooxidant and proinflammatory molecules, exert an impaired modulatory activity on innate and adaptive immune cells and how this activity contributes to atherosclerotic disease. In particular RBCs from patients with atherosclerosis, unlike those from healthy subjects, fail to control lipopolysaccharide-induced DC maturation and T lymphocyte apoptosis. Stored RBCs, accompanied by shedding of extracellular vesicles, stimulate peripheral blood mononuclear cells to release proinflammatory cytokines, augment mitogen-driven T cell proliferation, and polarize macrophages toward the proinflammatory M1 activation pathway. Collectively, literature data suggest that the crosstalk between RBCs with immune cells represents a novel mechanism by which oxidative stress can contribute to atherosclerotic disease progression and may be exploited for therapeutic interventions.
Collapse
|
45
|
Red blood cell storage duration and trauma. Transfus Med Rev 2014; 29:120-6. [PMID: 25573415 DOI: 10.1016/j.tmrv.2014.09.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/01/2014] [Accepted: 09/25/2014] [Indexed: 12/28/2022]
Abstract
Numerous retrospective clinical studies suggest that transfusion of longer stored red blood cells (RBCs) is associated with an independent risk of poorer outcomes for certain groups of patients, including trauma, intensive care, and cardiac surgery patients. Large multicenter randomized controlled trials are currently underway to address the concern about RBC storage duration. However, none of these randomized controlled trials focus specifically on trauma patients with hemorrhage. Major trauma, particularly due to road accidents, is the leading cause of critical injury in the younger-than-40-year-old age group. Severe bleeding associated with major trauma induces hemodynamic dysregulation that increases the risk of hypoxia, coagulopathy, and potentially multiorgan failure, which can be fatal. In major trauma, a multitude of stress-associated changes occur to the patient's RBCs, including morphological changes that increase cell rigidity and thereby alter blood flow hemodynamics, particularly in the microvascular vessels, and reduce RBC survival. Initial inflammatory responses induce deleterious cellular interactions, including endothelial activation, RBC adhesion, and erythrophagocytosis that are quickly followed by profound immunosuppressive responses. Stored RBCs exhibit similar biophysical characteristics to those of trauma-stressed RBCs. Whether transfusion of RBCs that exhibit storage lesion changes exacerbates the hemodynamic perturbations already active in the trauma patient is not known. This article reviews findings from several recent nonrandomized studies examining RBC storage duration and clinical outcomes in trauma patients. The rationale for further research on RBC storage duration in the trauma setting is provided.
Collapse
|
46
|
Microparticles provide a novel biomarker to predict severe clinical outcomes of dengue virus infection. J Virol 2014; 89:1587-607. [PMID: 25410854 DOI: 10.1128/jvi.02207-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Shedding of microparticles (MPs) is a consequence of apoptotic cell death and cellular activation. Low levels of circulating MPs in blood help maintain homeostasis, whereas increased MP generation is linked to many pathological conditions. Herein, we investigated the role of MPs in dengue virus (DENV) infection. Infection of various susceptible cells by DENV led to apoptotic death and MP release. These MPs harbored a viral envelope protein and a nonstructural protein 1 (NS1) on their surfaces. Ex vivo analysis of clinical specimens from patients with infections of different degrees of severity at multiple time points revealed that MPs generated from erythrocytes and platelets are two major MP populations in the circulation of DENV-infected patients. Elevated levels of red blood cell-derived MPs (RMPs) directly correlated with DENV disease severity, whereas a significant decrease in platelet-derived MPs was associated with a bleeding tendency. Removal by mononuclear cells of complement-opsonized NS1-anti-NS1 immune complexes bound to erythrocytes via complement receptor type 1 triggered MP shedding in vitro, a process that could explain the increased levels of RMPs in severe dengue. These findings point to the multiple roles of MPs in dengue pathogenesis. They offer a potential novel biomarker candidate capable of differentiating dengue fever from the more serious dengue hemorrhagic fever. IMPORTANCE Dengue is the most important mosquito-transmitted viral disease in the world. No vaccines or specific treatments are available. Rapid diagnosis and immediate treatment are the keys to achieve a positive outcome. Dengue virus (DENV) infection, like some other medical conditions, changes the level and composition of microparticles (MPs), tiny bag-like structures which are normally present at low levels in the blood of healthy individuals. This study investigated how MPs in culture and patients' blood are changed in response to DENV infection. Infection of cells led to programmed cell death and MP release. In patients' blood, the majority of MPs originated from red blood cells and platelets. Decreased platelet-derived MPs were associated with a bleeding tendency, while increased levels of red blood cell-derived MPs (RMPs) correlated with more severe disease. Importantly, the level of RMPs during the early acute phase could serve as a biomarker to identify patients with potentially severe disease who require immediate care.
Collapse
|
47
|
Sadallah S, Amicarella F, Eken C, Iezzi G, Schifferli JA. Ectosomes released by platelets induce differentiation of CD4+T cells into T regulatory cells. Thromb Haemost 2014; 112:1219-29. [PMID: 25209750 DOI: 10.1160/th14-03-0281] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/14/2014] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests an immune-modulatory role for platelets (PLT) and PLT-derived microvesicles. In particular, ectosomes, i.e. vesicles budding from PLT surface, have been shown to exert immunosuppressive activities on phagocytes. Here we investigated the effects mediated by PLT-derived ectosomes (PLT-Ecto) on CD4+ T cells. Exposure of activated CD4+ T cells to PLT-Ecto decreased their release of IFNγ, TNFα and IL-6, and increased the production of TGF-β1. Concomitantly, PLT-Ecto-exposed CD4+ T cells displayed increased frequencies of CD25high Foxp3+ cells. These phenomena were dose-dependent and PLT-Ecto specific, since they were not observed in the presence of polymorphonuclear- and erythrocyte-derived ectosomes. Analysis of specific T cell subsets revealed that PLT-Ecto induced differentiation of naïve T cells into Foxp3+ cells, but had no effect on pre-differentiated Foxp3+ regulatory T cells (Tregs). Importantly, PLT-Ecto-induced Foxp3+ cells were as effective as peripheral blood Tregs in suppressing CD8+ T cell proliferation. PLT-Ecto-mediated effects were partly dependent on PLT-derived TGF-β1, as they were to some extent inhibited by PLT-Ecto pretreatment with TGF-β1-neutralising antibodies. Interestingly, ectosome-derived TGF-β1 levels correlated with Foxp3+ T cell frequencies in blood of healthy donors. In conclusion, PLT-Ecto induce differentiation of CD4+ T cells towards functional Tregs. This may represent a mechanism by which PLT-Ecto enhance peripheral tolerance.
Collapse
Affiliation(s)
- S Sadallah
- Salima Sadallah, Immunonephrology Laboratory, Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland, Tel.: +41 61 265 32 62, Fax: +41 61 265 23 50, E-mail:
| | | | | | | | | |
Collapse
|
48
|
Cunningham M, Marks N, Barnado A, Wirth JR, Gilkeson G, Markiewicz M. Are microparticles the missing link between thrombosis and autoimmune diseases? Involvement in selected rheumatologic diseases. Semin Thromb Hemost 2014; 40:675-81. [PMID: 25173498 DOI: 10.1055/s-0034-1387924] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microparticles (MPs) are membrane-bound vesicles with important physiologic effects. MPs exchange information intercellularly, with each kind of MP carrying antigens and receptors of the cells from which they originated. They are biologic effectors in inflammation, angiogenesis, vascular injury, and thrombosis. Thrombosis is generally caused by abnormalities in blood flow, blood composition, and/or properties of the vessel wall. Thrombosis is a well-described feature of cardiovascular disease and cerebrovascular disease. Accumulating evidence suggests that increased risk of thrombosis is also characteristic of autoimmune disorders and immune-mediated diseases affecting all age groups, although the older adults are most vulnerable. Current research has also implicated MPs as a source of autoantigenic nuclear material that can form immune complexes, activate the innate immune system, and may lead to autoimmunity. This review focuses on the contribution of MPs to both the pathogenesis of autoimmune diseases and, as the immune and coagulation systems are tightly linked, their role in hypercoagulability in the setting of autoimmunity in an aging population.
Collapse
Affiliation(s)
- Melissa Cunningham
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Natalia Marks
- Department of Radiology, Maimonides Medical Center, Brooklyn, New York
| | - April Barnado
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Jena R Wirth
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Gary Gilkeson
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Margaret Markiewicz
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
49
|
Flatt JF, Bawazir WM, Bruce LJ. The involvement of cation leaks in the storage lesion of red blood cells. Front Physiol 2014; 5:214. [PMID: 24987374 PMCID: PMC4060409 DOI: 10.3389/fphys.2014.00214] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/19/2014] [Indexed: 12/12/2022] Open
Abstract
Stored blood components are a critical life-saving tool provided to patients by health services worldwide. Red cells may be stored for up to 42 days, allowing for efficient blood bank inventory management, but with prolonged storage comes an unwanted side-effect known as the "storage lesion", which has been implicated in poorer patient outcomes. This lesion is comprised of a number of processes that are inter-dependent. Metabolic changes include a reduction in glycolysis and ATP production after the first week of storage. This leads to an accumulation of lactate and drop in pH. Longer term damage may be done by the consequent reduction in anti-oxidant enzymes, which contributes to protein and lipid oxidation via reactive oxygen species. The oxidative damage to the cytoskeleton and membrane is involved in increased vesiculation and loss of cation gradients across the membrane. The irreversible damage caused by extensive membrane loss via vesiculation alongside dehydration is likely to result in immediate splenic sequestration of these dense, spherocytic cells. Although often overlooked in the literature, the loss of the cation gradient in stored cells will be considered in more depth in this review as well as the possible effects it may have on other elements of the storage lesion. It has now become clear that blood donors can exhibit quite large variations in the properties of their red cells, including microvesicle production and the rate of cation leak. The implications for the quality of stored red cells from such donors is discussed.
Collapse
Affiliation(s)
- Joanna F Flatt
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant Bristol, UK
| | - Waleed M Bawazir
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant Bristol, UK ; School of Biochemistry, University of Bristol Bristol, UK
| | - Lesley J Bruce
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant Bristol, UK
| |
Collapse
|
50
|
Kent MW, Kelher MR, West FB, Silliman CC. The pro-inflammatory potential of microparticles in red blood cell units. Transfus Med 2014; 24:176-81. [PMID: 24786047 DOI: 10.1111/tme.12123] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/31/2014] [Accepted: 04/05/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Microparticles (MPs) are submicron size cell fragments that are released from cells. OBJECTIVES We hypothesise that MPs increase during red blood cell (RBC) storage and are part of the pro-inflammatory activity, which accumulates in the RBC supernatant. METHODS/MATERIALS RBC units were separated from whole blood of eight healthy donors: 5 U were split, with 50% undergoing leucoreduction (LR) and the remaining left as unmodified controls. The remaining 3 U were leucoreduced. Samples were obtained at days (D) 1 and 42 and cell-free supernatants separated and stored. The supernatants were centrifuged at 17 000 × g (60 min) or 100 000 × g (120 min) into microparticle-rich (MPR) and microparticle-poor (MPP) portions, resuspended in albumin, incubated with antibodies to CD235 (RBCs), CD45 [white blood cells (WBCs)] and CD41a [platelets (Plts)], and analysed by flow cytometry. Isolated neutrophils were incubated with these samples, and priming activity measured. RESULTS Total MPs increased during storage; however, MPs that marked for precursor cell types did not. Significant priming accumulated in the MPP fraction during storage with some activity present in the MPR fraction from D1 and D42 LR-RBCs. CONCLUSION Most of the pro-inflammatory priming activity from stored RBCs resides in the MPP supernatant, although the MPR fraction from D42 LR-RBCs does contain some priming activity.
Collapse
Affiliation(s)
- M W Kent
- Research Laboratory, Bonfils Blood Center, Denver, Colorado, USA; Department of Pediatrics, University of Colorado, Denver, Colorado, USA
| | | | | | | |
Collapse
|