1
|
Rajput S, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Cardiovascular disease and thrombosis: Intersections with the immune system, inflammation, and the coagulation system. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:228-250. [PMID: 39159826 DOI: 10.1016/j.pharma.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The coagulation and immune system, both essential physiological systems in the human body, are intricately interconnected and play a critical role in determining the overall health of patients. These systems collaborate via various shared regulatory pathways, such as the Tissue Factor (TF) Pathway. Immunological cells that express TF and generate pro-inflammatory cytokines have the ability to affect coagulation. Conversely, coagulation factors and processes have a reciprocal effect on immunological responses by stimulating immune cells and regulating their functions. These interconnected pathways play a role in both preserving well-being and contributing to a range of pathological disorders. The close relationship between blood clotting and inflammation in the development of vascular disease has become a central focus of clinical study. This research specifically examines the crucial elements of this interaction within the contexts of cardiovascular disease and acute coronary syndrome. Tissue factor, the primary trigger of the extrinsic coagulation pathway, has a crucial function by inducing a proinflammatory reaction through the activation of coagulation factors. This, in turn, initiates coagulation and subsequent cellular signalling pathways. Protease-activated receptors establish the molecular connection between coagulation and inflammation by interacting with activated clotting factors II, X, and VII. Thrombosis, a condition characterised by the formation of blood clots, is the most dreaded consequence of cardiovascular disorders and a leading cause of death globally. Consequently, it poses a significant challenge to healthcare systems. Antithrombotic treatments efficiently target platelets and the coagulation cascade, but they come with the inherent danger of causing bleeding. Furthermore, antithrombotics are unable to fully eliminate thrombotic events, highlighting a treatment deficiency caused by a third mechanism that has not yet been sufficiently addressed, namely inflammation. Understanding these connections may aid in the development of novel approaches to mitigate the harmful mutual exacerbation of inflammation and coagulation. Gaining a comprehensive understanding of the intricate interaction among these systems is crucial for the management of diseases and the creation of efficacious remedies. Through the examination of these prevalent regulatory systems, we can discover novel therapeutic approaches that specifically target these complex illnesses. This paper provides a thorough examination of the reciprocal relationship between the coagulation and immune systems, emphasising its importance in maintaining health and understanding disease processes. This review examines the interplay between inflammation and thrombosis and its role in the development of thrombotic disorders.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India.
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
2
|
Deng G, Zhang Y, Song J, Zhang Y, Zheng Q, Luo Y, Fei X, Yang Y, Kuai L, Li B, Luo Y. The role and therapeutic strategies for tissue-resident memory T cells, central memory T cells, and effector memory T cells in psoriasis. Immunology 2024; 173:470-480. [PMID: 39136109 DOI: 10.1111/imm.13843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/19/2024] [Indexed: 10/04/2024] Open
Abstract
Psoriasis is a skin disease that is inflammatory and persistent, causing a high rate of recurrence, poor quality of life, and significant socioeconomic burden. Its main pathological manifestations are abnormal activation and infiltration of T cells and excessive proliferation of keratinocytes (KCs). The great majority of patients with psoriasis will relapse after remission. It usually lasts a lifetime and necessitates long-term treatment strategies. During periods of activity and remission, one of the main cell types in psoriasis is memory T cells, which include tissue-resident memory T (TRM) cells, central memory T (TCM) cells, and effector memory T (TEM) cells. They work by releasing inflammatory factors, cytotoxic particles, or altering cell subpopulations, leading to increased inflammation or recurrence. This review summarizes the role of memory T cells in the pathology and treatment of psoriasis, with a view to potential novel therapies and therapeutic targets.
Collapse
Affiliation(s)
- Guoshu Deng
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulin Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiankun Song
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zhang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Qi Zheng
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yue Luo
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoya Fei
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yang Yang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Li
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Jian X, Yang D, Wang L, Wang H. CREB1 Silencing Protects Against Inflammatory Response in Rats with Deep Vein Thrombosis Through Reducing RPL9 Expression and Blocking NF-κB Signaling. J Cardiovasc Transl Res 2024; 17:570-584. [PMID: 37891366 DOI: 10.1007/s12265-023-10450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Apoptosis and inflammation of vascular endothelial cells (VECs) are the most important causes of deep vein thrombosis (DVT). cAMP response element binding protein 1 (CREB1) encodes a transcription factor that binds as a homodimer to the cAMP-responsive element and can promote inflammation. CREB1 is found to be upregulated in the plasma of patients with venous thromboembolism. However, the biological functions of CREB1 in DVT remain unknown. We evaluated the effect of CREB1 in a rat model of inferior vena cava (IVA) stenosis-induced DVT. IVC stenosis resulted in stable thrombus, inflammatory response and CREB1 upregulation, whereas CREB1 knockdown inhibited thrombus and inflammation in DVT rats. In vitro analysis showed that CREB1 knockdown inhibited VEC apoptosis. Mechanistically, CREB1 knockdown reduced Ribosomal protein L9 (RPL9) expression and blocked the NF-κB pathway. Therefore, CREB1 may become a potential therapeutic target of DVT prevention.
Collapse
Affiliation(s)
- Xiaorong Jian
- Department of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26, Shengli Street, Jiang'an District, Wuhan, 430014, Hubei, China.
| | - Dehua Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Wang
- Department of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26, Shengli Street, Jiang'an District, Wuhan, 430014, Hubei, China
| | - Hongxiang Wang
- Department of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26, Shengli Street, Jiang'an District, Wuhan, 430014, Hubei, China.
| |
Collapse
|
4
|
Jacobs JW, Adkins BD, Bibb LA, Woo JS, Allen ES, Stephens LD, Binns TC, Bloch EM, Booth GS. The potential association between extracorporeal photopheresis and thrombosis. Bone Marrow Transplant 2024; 59:270-273. [PMID: 37996561 PMCID: PMC11423869 DOI: 10.1038/s41409-023-02153-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Affiliation(s)
- Jeremy W Jacobs
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Brian D Adkins
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lorin A Bibb
- Department of Dermatology, Mayo Clinic, Rocester, MN, USA
| | - Jennifer S Woo
- Department of Pathology, City of Hope National Medical Center, Irvine, CA, USA
| | - Elizabeth S Allen
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Laura D Stephens
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Thomas C Binns
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Garrett S Booth
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
5
|
Huang SL, Xin HY, Wang XY, Feng GG, Wu FQ, Feng ZP, Xing Z, Zhang XH, Xin HW, Luo WY. Recent Advances on the Molecular Mechanism and Clinical Trials of Venous Thromboembolism. J Inflamm Res 2023; 16:6167-6178. [PMID: 38111686 PMCID: PMC10726951 DOI: 10.2147/jir.s439205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
Venous thromboembolism is a condition that includes deep vein thrombosis and pulmonary embolism. It is the third most common cardiovascular disease behind acute coronary heart disease and stroke. Over the past few years, growing research suggests that venous thrombosis is also related to the immune system and inflammatory factors have been confirmed to be involved in venous thrombosis. The role of inflammation and inflammation-related biomarkers in cerebrovascular thrombotic disease is the subject of ongoing debate. P-selectin leads to platelet-monocyte aggregation and stimulates vascular inflammation and thrombosis. The dysregulation of miRNAs has also been reported in venous thrombosis, suggesting the involvement of miRNAs in the progression of venous thrombosis. Plasminogen activator inhibitor-1 (PAI-1) is a crucial component of the plasminogen-plasmin system, and elevated levels of PAI-1 in conjunction with advanced age are significant risk factors for thrombosis. In addition, it has been showed that one of the ways that neutrophils promote venous thrombosis is the formation of neutrophil extracellular traps (NETs). In recent years, the role of extracellular vesicles (EVs) in the occurrence and development of VTE has been continuously revealed. With the advancement of research technology, the complex regulatory role of EVs on the coagulation process has been gradually discovered. However, our understanding of the causes and consequences of these changes in venous thrombosis is still limited. Therefore, we review our current understanding the molecular mechanisms of venous thrombosis and the related clinical trials, which is crucial for the future treatment of venous thrombosis.
Collapse
Affiliation(s)
- Shao-Li Huang
- Medical Laboratory Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524400, People’s Republic of China
- First Clinical College, Guangdong Medical University, Guangdong, 524400, People’s Republic of China
- Clinical laboratory, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Hong-Yi Xin
- Doctoral Scientific Research Center, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
- Guangdong Medical University Affiliated Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Xiao-Yan Wang
- Doctoral Scientific Research Center, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
- Guangdong Medical University Affiliated Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Guang-Gui Feng
- Clinical laboratory, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Fu-Qing Wu
- Clinical laboratory, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Zhi-Peng Feng
- Department of Gastroenterology, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang, Hunan, 414000, People’s Republic of China
| | - Zhou Xing
- First Clinical College, Guangdong Medical University, Guangdong, 524400, People’s Republic of China
| | - Xi-He Zhang
- Doctoral Scientific Research Center, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
- Guangdong Medical University Affiliated Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
| | - Hong-Wu Xin
- Doctoral Scientific Research Center, Lianjiang People’s Hospital, Guangdong, 524400, People’s Republic of China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, Hubei, 434023, People’s Republic of China
- Research Centre of Molecular Medicine, Medical College of Chifeng University, Chifeng, Inner Mongolian Autonomous Region, 024000, People’s Republic of China
| | - Wen-Ying Luo
- Medical Laboratory Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524400, People’s Republic of China
| |
Collapse
|
6
|
Chooklin S, Chuklin S. PATHOPHYSIOLOGICAL MECHANISMS OF DEEP VEIN THROMBOSIS. FIZIOLOHICHNYĬ ZHURNAL 2023; 69:133-144. [DOI: 10.15407/fz69.06.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Deep venous thrombosis is a frequent multifactorial disease and most of the time is triggered by the interaction between acquired risk factors, particularly immobility, and hereditary risk factors such as thrombophilias. The mechanisms underlying deep venous thrombosis are not fully elucidated; however, in recent years the role of venous flow, endothelium, platelets, leukocytes, and the interaction between inflammation and hemostasis has been determined. Alteration of venous blood flow produces endothelial activation, favoring the adhesion of platelets and leukocytes, which, through tissue factor expression and neutrophil extracellular traps formation, contribute to the activation of coagulation, trapping more cells, such as red blood cells, monocytes, eosinophils, lymphocytes. The coagulation factor XI-driven propagation phase of blood coagulation plays a major role in venous thrombus growth, but a minor role in hemostasis. In this work, the main mechanisms involved in the pathophysiology of deep vein thrombosis are described.
Collapse
|
7
|
Henke PK, Nicklas JM, Obi A. Immune cell-mediated venous thrombus resolution. Res Pract Thromb Haemost 2023; 7:102268. [PMID: 38193054 PMCID: PMC10772895 DOI: 10.1016/j.rpth.2023.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 01/10/2024] Open
Abstract
Herein, we review the current processes that govern experimental deep vein thrombus (DVT) resolution. How the human DVT resolves at the molecular and cellular level is not well known due to limited specimen availability. Experimentally, the thrombus resolution resembles wound healing, with early neutrophil-mediated actions followed by monocyte/macrophage-mediated events, including neovascularization, fibrinolysis, and eventually collagen replacement. Potential therapeutic targets are described, and coupling with site-directed approaches to mitigate off-target effects is the long-term goal. Similarly, timing of adjunctive agents to accelerate DVT resolution is an area that is only starting to be considered. There is much critical research that is needed in this area.
Collapse
Affiliation(s)
- Peter K. Henke
- Department of Surgery, University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| | - John M. Nicklas
- Department of Medicine, Brown University Medical School, Providence, Rhode Island, USA
| | - Andrea Obi
- Department of Surgery, University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Lao P, Chen J, Tang L, Zhang J, Chen Y, Fang Y, Fan X. Regulatory T cells in lung disease and transplantation. Biosci Rep 2023; 43:BSR20231331. [PMID: 37795866 PMCID: PMC10611924 DOI: 10.1042/bsr20231331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/06/2023] Open
Abstract
Pulmonary disease can refer to the disease of the lung itself or the pulmonary manifestations of systemic diseases, which are often connected to the malfunction of the immune system. Regulatory T (Treg) cells have been shown to be important in maintaining immune homeostasis and preventing inflammatory damage, including lung diseases. Given the increasing amount of evidence linking Treg cells to various pulmonary conditions, Treg cells might serve as a therapeutic strategy for the treatment of lung diseases and potentially promote lung transplant tolerance. The most potent and well-defined Treg cells are Foxp3-expressing CD4+ Treg cells, which contribute to the prevention of autoimmune lung diseases and the promotion of lung transplant rejection. The protective mechanisms of Treg cells in lung disease and transplantation involve multiple immune suppression mechanisms. This review summarizes the development, phenotype and function of CD4+Foxp3+ Treg cells. Then, we focus on the therapeutic potential of Treg cells in preventing lung disease and limiting lung transplant rejection. Furthermore, we discussed the possibility of Treg cell utilization in clinical applications. This will provide an overview of current research advances in Treg cells and their relevant application in clinics.
Collapse
Affiliation(s)
- Peizhen Lao
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jingyi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Longqian Tang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jiwen Zhang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuxi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuyin Fang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Xingliang Fan
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| |
Collapse
|
9
|
Wilhelm G, Mertowska P, Mertowski S, Przysucha A, Strużyna J, Grywalska E, Torres K. The Crossroads of the Coagulation System and the Immune System: Interactions and Connections. Int J Mol Sci 2023; 24:12563. [PMID: 37628744 PMCID: PMC10454528 DOI: 10.3390/ijms241612563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The coagulation and immune systems, two vital systems in the human body, share intimate connections that fundamentally determine patient health. These systems work together through several common regulatory pathways, including the Tissue Factor (TF) Pathway. Immune cells expressing TF and producing pro-inflammatory cytokines can influence coagulation, while coagulation factors and processes reciprocally impact immune responses by activating immune cells and controlling their functions. These shared pathways contribute to maintaining health and are also involved in various pathological conditions. Dysregulated coagulation, triggered by infection, inflammation, or tissue damage, can result in conditions such as disseminated intravascular coagulation (DIC). Concurrently, immune dysregulation may lead to coagulation disorders and thrombotic complications. This review elucidates these intricate interactions, emphasizing their roles in the pathogenesis of autoimmune diseases and cancer. Understanding the complex interplay between these systems is critical for disease management and the development of effective treatments. By exploring these common regulatory mechanisms, we can uncover innovative therapeutic strategies targeting these intricate disorders. Thus, this paper presents a comprehensive overview of the mutual interaction between the coagulation and immune systems, highlighting its significance in health maintenance and disease pathology.
Collapse
Affiliation(s)
- Grzegorz Wilhelm
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Anna Przysucha
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Jerzy Strużyna
- East Center of Burns Treatment and Reconstructive Surgery, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Kamil Torres
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| |
Collapse
|
10
|
Solomou EE, Delaporta P, Mantzou A, Tzannoudaki M, Diamantopoulos P, Salamaliki C, Kontandreopoulou CN, Vyniou NA, Perganti F, Papassotiriou I, Kattamis A. Evidence that platelets from transfusion-dependent β-thalassemia patients induce T cell activation. Clin Immunol 2023:109653. [PMID: 37269985 DOI: 10.1016/j.clim.2023.109653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
A hypercoagulable state leading to increased risk for thrombotic events represents one of the most common complications observed in transfusion-dependent β-thalassemia (TDT) patients. TDT patients have increased frequencies of circulating activated platelets. However, there is no information so far if platelets from TDT patients can activate T cells. In the present study we showed that T cells treated with platelets from TDT patients showed significant increased surface expression of CD69 compared to the T cells treated with platelets from healthy individuals. Patients with splenectomy showed increased T cell activation compared to patients with intact spleen. No T cell activation was observed following incubation with plasma alone, nor with platelets from healthy subjects. The percentages of regulatory T cells (Tregs) were also examined. TDT patients showed statistically significant increased percentages of Tregs compared to healthy controls. Additionally, we observed a positive statistically significant correlation between the percentages of Tregs and the platelet-induced activated T cells in patients who were not treated with aspirin. TDT patients showed increased levels of sP-selectin, suPAR and GDF-15, molecules implicated in platelet activation. We show that platelets from TDT patients can activate T cells in vitro. This activation correlates with markers of platelet activation and increased numbers of Tregs, perhaps in an effort to eliminate immune dysregulation, conceivably secondary to platelet activation.
Collapse
Affiliation(s)
- Elena E Solomou
- University of Patras Medical School, Department of Internal Medicine, Rio 26500, Greece.
| | - Polyxeni Delaporta
- First Department of Pediatrics, Thalassemia Unit, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Aimilia Mantzou
- First Department of Pediatrics, Thalassemia Unit, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Marianna Tzannoudaki
- Department of Immunology-Histocompatibility, Specialized Center and Referral Center for Primary Immunodeficiencies-Pediatric Immunology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Panagiotis Diamantopoulos
- First Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Christina Salamaliki
- University of Patras Medical School, Department of Internal Medicine, Rio 26500, Greece
| | | | - Nora-Athina Vyniou
- First Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Foteini Perganti
- First Department of Pediatrics, Thalassemia Unit, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Ioannis Papassotiriou
- First Department of Pediatrics, Thalassemia Unit, National and Kapodistrian University of Athens, School of Medicine, Greece
| | - Antonis Kattamis
- First Department of Pediatrics, Thalassemia Unit, National and Kapodistrian University of Athens, School of Medicine, Greece
| |
Collapse
|
11
|
Deng Y, Zhou F, Li Q, Guo J, Cai B, Li G, Liu J, Li L, Zheng Q, Chang D. Associations between neutrophil-lymphocyte ratio and monocyte to high-density lipoprotein ratio with left atrial spontaneous echo contrast or thrombus in patients with non-valvular atrial fibrillation. BMC Cardiovasc Disord 2023; 23:234. [PMID: 37142962 PMCID: PMC10157900 DOI: 10.1186/s12872-023-03270-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/27/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND The importance of inflammation in thrombosis is increasingly appreciated. Neutrophil-lymphocyte ratio (NLR) and monocyte to high-density lipoprotein ratio (MHR) are important indicators of systemic inflammation. This study aimed to investigate the associations between NLR and MHR with left atrial appendage thrombus (LAAT) and spontaneous echo contrast (SEC) in patients with non-valvular atrial fibrillation. METHODS This retrospective, cross-sectional study enrolled 569 consecutive patients with non-valvular atrial fibrillation. Multivariable logistic regression analysis was used to investigate independent risk factors of LAAT/SEC. Receiver operating characteristic (ROC) curves were used to evaluate the specificity and sensitivity of NLR and MHR in predicting LAAT/SEC. Subgroup and Pearson correlation analyses were used to assess the correlations between NLR and MHR with the CHA2DS2-VASc score. RESULTS Multivariate logistic regression analysis showed that NLR (OR: 1.49; 95%CI: 1.173-1.892) and MHR (OR: 2.951; 95%CI: 1.045-8.336) were independent risk factors for LAAT/SEC. The area under the ROC curve of NLR (0.639) and MHR (0.626) was similar to that of the CHADS2 score (0.660) and CHA2DS2-VASc score (0.637). Subgroup and Pearson correlation analyses showed significant but very weak associations between NLR (r = 0.139, P < 0.05) and MHR (r = 0.095, P < 0.05) with the CHA2DS2-VASc score. CONCLUSION Generally, NLR and MHR are independent risk factors for predicting LAAT/SEC in patients with non-valvular atrial fibrillation.
Collapse
Affiliation(s)
- Yingjian Deng
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Faguang Zhou
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Qiang Li
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Jincun Guo
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Binni Cai
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Guiyang Li
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Jianghai Liu
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Linlin Li
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Qi Zheng
- Department of Psychology, Xiamen Xianyue Hospital, Xiamen, China.
| | - Dong Chang
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiamen, China.
| |
Collapse
|
12
|
Van Bruggen S, Martinod K. The coming of age of neutrophil extracellular traps in thrombosis: Where are we now and where are we headed? Immunol Rev 2022; 314:376-398. [PMID: 36560865 DOI: 10.1111/imr.13179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombosis remains a major problem in our society, manifesting across multiple demographic groups and with high associated morbidity and mortality. Thrombus development is the result of a complex mechanism in which multiple cell types and soluble factors play a crucial role. One cell that has gained the most attention in recent years is the neutrophil. This key member of the innate immune system can form neutrophil extracellular traps (NETs) in response to activating stimuli in circulation. NETs form a scaffold for thrombus formation, both initiating the process and stabilizing the final product. As the first responders of the host immune system, neutrophils have the flexibility to recognize a variety of molecules and can quickly interact with a range of different cell types. This trait makes them sensitive to exogenous stimuli. NET formation in response to pathogens is well established, leading to immune-mediated thrombus formation or immunothrombosis. NETs can also be formed during sterile inflammation through the activation of neutrophils by fellow immune cells including platelets, or activated endothelium. In chronic inflammatory settings, NETs can ultimately promote the development of tissue fibrosis, with organ failure as an end-stage outcome. In this review, we discuss the different pathways through which neutrophils can be activated toward NET formation and how these processes can result in a shared outcome: thrombus formation. Finally, we evaluate these different interactions and mechanisms for their potential as therapeutic targets, with neutrophil-targeted therapies providing a future approach to treating thrombosis. In contrast to current practices, such treatment could result in reduced pathogenic blood clot formation without increasing the risk of bleeding.
Collapse
Affiliation(s)
- Stijn Van Bruggen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Dysregulated haemostasis in thrombo-inflammatory disease. Clin Sci (Lond) 2022; 136:1809-1829. [PMID: 36524413 PMCID: PMC9760580 DOI: 10.1042/cs20220208] [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: 09/03/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Inflammatory disease is often associated with an increased incidence of venous thromboembolism in affected patients, although in most instances, the mechanistic basis for this increased thrombogenicity remains poorly understood. Acute infection, as exemplified by sepsis, malaria and most recently, COVID-19, drives 'immunothrombosis', where the immune defence response to capture and neutralise invading pathogens causes concurrent activation of deleterious prothrombotic cellular and biological responses. Moreover, dysregulated innate and adaptive immune responses in patients with chronic inflammatory conditions, such as inflammatory bowel disease, allergies, and neurodegenerative disorders, are now recognised to occur in parallel with activation of coagulation. In this review, we describe the detailed cellular and biochemical mechanisms that cause inflammation-driven haemostatic dysregulation, including aberrant contact pathway activation, increased tissue factor activity and release, innate immune cell activation and programmed cell death, and T cell-mediated changes in thrombus resolution. In addition, we consider how lifestyle changes increasingly associated with modern life, such as circadian rhythm disruption, chronic stress and old age, are increasingly implicated in unbalancing haemostasis. Finally, we describe the emergence of potential therapies with broad-ranging immunothrombotic functions, and how drug development in this area is challenged by our nascent understanding of the key molecular and cellular parameters that control the shared nodes of proinflammatory and procoagulant pathways. Despite the increasing recognition and understanding of the prothrombotic nature of inflammatory disease, significant challenges remain in effectively managing affected patients, and new therapeutic approaches to curtail the key pathogenic steps in immune response-driven thrombosis are urgently required.
Collapse
|
14
|
B lymphocyte-deficiency in mice promotes venous thrombosis. Heliyon 2022; 8:e11740. [DOI: 10.1016/j.heliyon.2022.e11740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/30/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
|
15
|
Zhang L, Fang Y, Xing J, Cheng H, Sun X, Yuan Z, Xu Y, Hao J. The Efficacy of the Systemic Immune-Inflammation Index and Prognosis Nutritional Index for the Diagnosis of Venous Thromboembolism in Gastrointestinal Cancers. J Inflamm Res 2022; 15:4649-4661. [PMID: 35996687 PMCID: PMC9391990 DOI: 10.2147/jir.s376601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aimed to analyze the association between venous thromboembolism (VTE) and inflammatory markers like systemic immune-inflammation index (SII) and prognosis nutritional index (PNI), and to evaluate their efficacy for the diagnosis of VTE in patients with gastrointestinal malignancies. Patients and Methods A total of 1326 patients with the initial diagnosis of gastrointestinal cancer in the First Affiliated Hospital of Anhui Medical University (AHMU) were enrolled in the training cohort. Univariate and multivariate analysis was used to pinpoint independent predictors of VTE, which were eventually visualized as the nomogram models. The Akaike Information Criterion (AIC) was used to screen the best model. The receiver operating characteristic curve (ROC) and the clinical decision curve analysis (DCA) were utilized to evaluate the models’ predictive performance in the training queue and another external sample of 250 patients at the Second Affiliated Hospital of AHMU. Results A total of 476 patients were complicated with VTE in the training cohort. Multifactorial analysis of clinical characteristics and inflammatory markers showed that PNI, SII, age, tumor location, and therapy were independent risk factors of VTE, visualized as model A. Another model B was constructed by adding coagulation markers to the previous analysis. Model B was the best prediction model with the minimum AIC value, followed by model A with an AUC of 0.806 (95% CI 0.782~0.830) which was similar to model B’s 0.832 (95% CI 0.810~0.855) but significantly higher than the currently widely used Khorana score’s 0.592 (95% CI 0.562~0.621) and the CATS score’s 0.682 (95% CI 0.653~0.712). The external verification yielded similar findings, with the AUC being 0.792 (95% CI 0.734~0.851), 0.834 (95% CI 0.778~0.890), 0.655 (95% CI 0.582~0.729), and 0.774 (95% CI 0.699~0.849) respectively. The DCA curves demonstrated that new models had excellent usefulness in screening patients with a high VTE risk. Conclusion The SII and PNI were simple and viable inflammatory markers associated with VTE, and the nomogram based on them and clinical features had a meaningful clinical utility for VTE in patients with gastrointestinal malignancies.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yue Fang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Jianghao Xing
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Hao Cheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Xiaonan Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Zhichao Yuan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yidan Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Jiqing Hao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
16
|
Ding J, Song B, Xie X, Li X, Chen Z, Wang Z, Pan L, Lan D, Meng R. Inflammation in Cerebral Venous Thrombosis. Front Immunol 2022; 13:833490. [PMID: 35444662 PMCID: PMC9013750 DOI: 10.3389/fimmu.2022.833490] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/28/2022] [Indexed: 01/04/2023] Open
Abstract
Cerebral venous thrombosis (CVT) is a rare form of cerebrovascular disease that impairs people's wellbeing and quality of life. Inflammation is considered to play an important role in CVT initiation and progression. Several studies have reported the important role of leukocytes, proinflammatory cytokines, and adherence molecules in the CVT-related inflammatory process. Moreover, inflammatory factors exacerbate CVT-induced brain tissue injury leading to poor prognosis. Based on clinical observations, emerging evidence shows that peripheral blood inflammatory biomarkers-especially neutrophil-to-lymphocyte ratio (NLR) and lymphocyte count-are correlated with CVT [mean difference (MD) (95%CI), 0.74 (0.11, 1.38), p = 0.02 and -0.29 (-0.51, -0.06), p = 0.01, respectively]. Moreover, increased NLR and systemic immune-inflammation index (SII) portend poor patient outcomes. Evidence accumulated since the outbreak of coronavirus disease-19 (COVID-19) indicates that COVID-19 infection and COVID-19 vaccine can induce CVT through inflammatory reactions. Given the poor understanding of the association between inflammation and CVT, many conundrums remain unsolved. Further investigations are needed to elucidate the exact relationship between inflammation and CVT in the future.
Collapse
Affiliation(s)
- Jiayue Ding
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoying Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Xiran Xie
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Xaingyu Li
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhiying Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongao Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Duo Lan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
17
|
Shahneh F, Christian Probst H, Wiesmann SC, A-Gonzalez N, Ruf W, Steinbrink K, Raker VK, Becker C. Inflammatory Monocyte Counts Determine Venous Blood Clot Formation and Resolution. Arterioscler Thromb Vasc Biol 2022; 42:145-155. [PMID: 34911360 DOI: 10.1161/atvbaha.121.317176] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Monocytes are thought to be involved in venous thrombosis but the role of individual monocyte subpopulations on thrombus formation, clot inflammation, and degradation is an important unresolved issue. We investigate the role of inflammatory Ly6Chi monocytes in deep vein thrombosis and their potential therapeutic impact. METHODS Frequencies and compositions of blood monocytes were analyzed by flow cytometry in CCR2-/- (C-C chemokine receptor type 2) and wild-type mice of different ages and after treatment with the NR4A1 (nuclear receptor group 4 family A member 1, Nur77) agonist CnsB (cytosporone B). TF (tissue factor) sufficient and deficient Ly6Chi monocytes were adoptively transferred into aged CCR2-/- mice. Thrombus formation and size were followed by ultrasound over a 3-week period after surgical reduction of blood flow (stenosis) in the inferior vena cava. RESULTS Reduced numbers of peripheral monocytes in aged (>30 w) CCR2-/- mice are accompanied by reduced thrombus formation after inferior vena cava ligation. Reducing the number of inflammatory Ly6Chi monocytes in wild-type mice by CsnB treatment before ligation, similarly suspends clotting, while later treatment (d1 or d4) reduces thrombus growth and accelerates resolution. We describe how changes in inflammatory monocyte numbers affect the gradual differentiation of monocytes in thrombi and show that only tissue factor-competent Ly6Chi monocytes restore thrombosis in aged CCR2-/- mice. CONCLUSIONS We conclude that the number of inflammatory Ly6Chi monocytes controls deep vein thrombosis formation, growth, and resolution and can be therapeutically manipulated with a NR4A1 agonist at all disease stages.
Collapse
Affiliation(s)
- Fatemeh Shahneh
- Department of Dermatology (F.S.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
- Center for Thrombosis and Hemostasis (F.S., W.R.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Hans Christian Probst
- Institute for Immunology (H.C.P.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Sabine C Wiesmann
- Institute of Immunology (S.C.W., N.A.-G) and Westfälische Wilhelms-University Münster, Germany
| | - Noelia A-Gonzalez
- Institute of Immunology (S.C.W., N.A.-G) and Westfälische Wilhelms-University Münster, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (F.S., W.R.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| | - Verena K Raker
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| | - Christian Becker
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| |
Collapse
|
18
|
Fu C, Wang G, Yang W. Vascular Thrombosis and Anti-PD-1 Therapy: A Series of Cases. Cancer Manag Res 2021; 13:8849-8853. [PMID: 34858058 PMCID: PMC8629915 DOI: 10.2147/cmar.s338023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/05/2021] [Indexed: 11/23/2022] Open
Abstract
Immune checkpoint inhibitors may cause unique immune-related adverse events. Vascular thrombosis, especially arterial thrombosis, is rare but life-threatening, and little is known about its relevance to immunotherapy. Here, we reported two cases of vascular thrombosis, including venous and arterial thrombosis in cancer patients receiving anti-PD-1 antibody in combination with chemotherapy. Systemic corticosteroids and anticoagulant treatment were administered immediately in two cases. In case 1, anti-PD-1 antibody was permanently discontinued, and recurrence of vascular thrombosis was not observed during the follow-up. In case 2, the patient continued anti-PD-1 immunotherapy and unfortunately died of cerebral infarction 2 months later. This case report provides a strong evidence for the association between PD-1 blockade and vascular thrombosis and offers some general guidelines on the management of the immune-related vascular thrombosis events induced by anti-PD-1 therapy.
Collapse
Affiliation(s)
- Changfang Fu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, People’s Republic of China
| | - Gang Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, People’s Republic of China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People’s Republic of China
| |
Collapse
|
19
|
|
20
|
Grosse GM, Werlein C, Blume N, Abu-Fares O, Götz F, Gabriel MM, Ernst J, Leotescu A, Worthmann H, Kühnel MP, Jonigk DD, Falk CS, Weissenborn K, Schuppner R. Circulating Cytokines and Growth Factors in Acute Cerebral Large Vessel Occlusion-Association with Success of Endovascular Treatment. Thromb Haemost 2021; 122:623-632. [PMID: 34225367 PMCID: PMC9142215 DOI: 10.1055/a-1544-5431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mechanical thrombectomy (MT) is a highly efficient treatment in patients with acute ischemic stroke due to large vessel occlusion (LVO). However, in a relevant proportion of LVO, no sufficient recanalization can be achieved. The composition of cerebral thrombi is highly heterogeneous and may constitute a relevant factor for insufficient reperfusion. We hypothesized that circulating cytokines and growth factors involved in thromboinflammation and platelet activation may be associated with reperfusion status and thrombus composition in patients undergoing MT. An according biomarker panel was measured in plasma specimens taken prior to MT and at a 7-day follow-up. The reperfusion status was categorized into sufficient or insufficient. The composition of retrieved thrombi was histologically analyzed. Differences of baseline biomarker concentrations between insufficient and sufficient reperfusions were highest for interferon (IFN)-γ, epidermal growth factor, platelet-derived growth factor (PDGF)-AB/BB, and IFN-γ-induced protein 10 (IP-10/CXCL10). After applying correction for multiple comparisons and logistic regression analysis adjusting for stroke etiology, intravenous thrombolysis, and vascular risk factors, PDGF-AB/BB was identified as an independent predictor of reperfusion status (odds ratio: 0.403; 95% confidence interval: 0.199-0.819). Histological analysis revealed that the majority of thrombi had a mixed composition. In conclusion, this study provides the first evidence that cytokines and growth factors are potential effectors in patients undergoing MT for the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Gerrit M. Grosse
- Department of Neurology, Hannover Medical School, Hannover, Germany,Address for correspondence Gerrit M. Grosse, MD Department of Neurology, Hannover Medical SchoolCarl-Neuberg-Str. 1, 30625 HannoverGermany
| | | | - Nicole Blume
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Omar Abu-Fares
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Friedrich Götz
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Maria M. Gabriel
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Johanna Ernst
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Andrei Leotescu
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Mark P. Kühnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany,Member of the German Center for Lung Research (DZL), Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Danny D. Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany,Member of the German Center for Lung Research (DZL), Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | | | - Ramona Schuppner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
21
|
Abstract
The association between inflammation, infection, and venous thrombosis has long been recognized; yet, only in the last decades have we begun to understand the mechanisms through which the immune and coagulation systems interact and reciprocally regulate one another. These interconnected networks mount an effective response to injury and pathogen invasion, but if unregulated can result in pathological thrombosis and organ damage. Neutrophils, monocytes, and platelets interact with each other and the endothelium in host defense and also play critical roles in the formation of venous thromboembolism. This knowledge has advanced our understanding of both human physiology and pathophysiology, as well as identified mechanisms of anticoagulant resistance and novel therapeutic targets for the prevention and treatment of thrombosis. In this review, we discuss the contributions of inflammation and infection to venous thromboembolism.
Collapse
Affiliation(s)
- Meaghan E. Colling
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Benjamin E. Tourdot
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Henke PK, Obi AT. Advances in understanding the interplay between adaptive and innate immunity in experimental venous thrombus resolution. J Thromb Haemost 2021; 19:1387-1389. [PMID: 33595180 DOI: 10.1111/jth.15249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Peter K Henke
- University of Michigan Health System, Ann Arbor, MI, USA
| | - Andrea T Obi
- University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Shahneh F, Grill A, Klein M, Frauhammer F, Bopp T, Schäfer K, Raker VK, Becker C. Specialized regulatory T cells control venous blood clot resolution through SPARC. Blood 2021; 137:1517-1526. [PMID: 32932520 DOI: 10.1182/blood.2020005407] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
The cells and mechanisms involved in blood clot resorption are only partially known. We show that regulatory T cells (Tregs) accumulate in venous blood clots and regulate thrombolysis by controlling the recruitment, differentiation and matrix metalloproteinase (MMP) activity of monocytes. We describe a clot Treg population that forms the matricellular acid- and cysteine-rich protein SPARC (secreted protein acidic and rich in cysteine) and show that SPARC enhances monocyte MMP activity and that SPARC+ Tregs are crucial for blood clot resorption. By comparing different treatment times, we define a therapeutic window of Treg expansion that accelerates clot resorption.
Collapse
Affiliation(s)
- Fatemeh Shahneh
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| | | | - Matthias Klein
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany; and
| | - Felix Frauhammer
- Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany; and
| | | | - Verena K Raker
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| | - Christian Becker
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| |
Collapse
|
24
|
Kotyla PJ, Engelmann M, Giemza-Stokłosa J, Wnuk B, Islam MA. Thromboembolic Adverse Drug Reactions in Janus Kinase (JAK) Inhibitors: Does the Inhibitor Specificity Play a Role? Int J Mol Sci 2021; 22:2449. [PMID: 33671049 PMCID: PMC7957632 DOI: 10.3390/ijms22052449] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Recent advances in immunology enabled the characterization of several signal transmitting pathways responsible for proper cytokine and chemokine signaling. Among them, Janus kinases (JAKs) are essential components of receptor activation systems. The discovery of JAK kinases enabled the synthesis of JAK kinase inhibitors (JAKi or Jakinibs), which have proven to be efficacious in the treatment of hematologic malignancies and several rheumatological disorders and continue to be investigated in many clinical indications. Blocking multiple cytokines belonging to several cytokine families with a single small molecule may, however, create a potential risk for the patients. Recently, a higher risk of thromboembolic complications, namely, deep vein thrombosis and pulmonary embolism, has been recognized as the main concern during treatment with Jakinibs. At present, it is not entirely clear whether this increased risk is related to direct cytokine blockade, the presence of concomitant diseases in treated patients or other unknown circumstances that work together to increase the risk of this side effect. In this review, we discuss data on the risk of thromboembolic side effects, with special emphasis on the mechanism that may be responsible for this increased risk. Many indirect data indicate that higher thromboembolic risk may be related to the specificity of JAK inhibitor action, such that preferentially blocking one signaling pathway upsets the balance between pro and anti-thrombotic activities.
Collapse
Affiliation(s)
- Przemysław J. Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Faculty in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Małgorzata Engelmann
- Department of Physiotherapy in Internal Medicine, Academy of Physical Education in Katowice, 40-065 Katowice, Poland;
| | | | - Bartosz Wnuk
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
25
|
Duan J, Leng X, Han Z, Cai Y, Wang C, Rajah G, Song H, Ding Y, Ji X. Identifying Biomarkers Associated with Venous Infarction in Acute/Subacute Cerebral Venous Thrombosis. Aging Dis 2021; 12:93-101. [PMID: 33532130 PMCID: PMC7801269 DOI: 10.14336/ad.2020.0405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022] Open
Abstract
Among cerebral venous thrombosis (CVT) patients, those with venous infarction have more severe clinical presentations and worse outcomes. Identifying biomarkers associated with venous infarction in CVT may help understand the pathogenesis and provide potentially useful therapeutic markers. Fifty-two CVT patients were prospectively recruited and divided into three groups: acute/subacute CVT with venous infarction (ASVI, n=30), without venous infarction (ASOVI, n=13), and chronic CVT (n=9). Blood brain barrier (BBB) permeability-related proteins, including claudin-5, occludin, matrix metalloproteinase-9, glial fibrillary acidic protein, and S100B, and inflammation-related factor high-sensitivity C-reactive protein (hs-CRP), were tested in serum and/or cerebrospinal fluid upon admission. We compared these biomarkers between the three groups and investigated their associations with venous infarction and clinical symptom severity in acute/subacute CVT patients on admission using the NIH Stroke Scale (NIHSS). Serum hs-CRP was significantly higher in acute/subacute CVT patients than chronic CVT patients. For acute/subacute CVT patients, levels were significantly higher in the ASVI group than the ASOVI group for serum claudin-5 (medians 2.80 vs. 2.50 mg/I, respectively, P = 0.039) and hs-CRP (medians 17.25 vs. 2.27 mg/l, respectively, P = 0.003). Both these biomarkers, analyzed as categorical or continuous variables, were also significantly associated with venous infarction in acute/subacute CVT patients after logistic regression analysis. Additionally, hs-CRP was positively correlated with the NIHSS (r = 0.710, P < 0.001) on admission in acute/subacute CVT patients. In CVT patients, venous infarction was associated with BBB disruption and potentially inflammation. Hs-CRP might serve as a biomarker reflecting the clinical severity of CVT in the acute/subacute stages.
Collapse
Affiliation(s)
- Jiangang Duan
- 1Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China.,7Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinyi Leng
- 2Department of Medicine & Therapeutics, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziping Han
- 3Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,7Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanning Cai
- 4Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chunxiu Wang
- 5Department of Evidence-based Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gary Rajah
- 6Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Haiqing Song
- 7Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- 6Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xunming Ji
- 7Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Hetta HF, Mohamed AAA, Zahran AM, A Mahran S, MY Sayed M, GA Saleh M, Abdelazeem K, Batiha GES, Al-Rejaie S, Waheed Y, Muhammad K, M Hassanien M. Possible Role of Regulatory B Cells in Different Behçet's Disease Phenotypes and Therapies: First Report from Egypt. J Inflamm Res 2021; 14:737-744. [PMID: 33727848 PMCID: PMC7955029 DOI: 10.2147/jir.s279912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIM The data about the role of regulatory B cells (Breg) in Behcet Disease (BD) are scarce. We aimed to evaluate the frequency of total B lymphocytes and Breg cells in different BD phenotypes and therapies attempting to unravel their function. METHODS This cross-sectional study included 35 BD patients and 39 healthy controls (HCs). The demographic data of the study subjects were collected including age and gender. Current medications including disease-modifying anti-rheumatic drugs (DMARDs) were recorded. All patients underwent testing for baseline laboratory investigations including full blood count, liver and kidney function tests, erythrocyte sedimentation rate (ESR) by Westergren blot and C-reactive protein (CRP). Measurement of the total B lymphocytes and their subtypes B regulatory lymphocytes by flow cytometric assay. Assessment of BD activity was done using the revised Behçet's Disease Current Activity Form (BDCAF) 2006 and Behçet's Syndrome Activity Score (BSAS) 1111111111. All participants were assessed for the presence of erectile dysfunction using the International Index of Erectile Function (IIEF-5 score), and for depression using the Beck Depression Inventory. RESULTS A dramatic drop in the number of B cells, total and regulatory, was observed in the patients compared to the HCs. Regulatory cells (Bregs) tend to be upregulated with genital ulcers or vascular disease. Bregs but not B lymphocytes were associated with BSAS and ESR. Neither the total B lymphocytes nor the Bregs correlated with CRP or the sexual function or depression scores. Of all the used medications, low-dose aspirin was seen with markedly high Bregs proportions. CONCLUSION This study supports the role of B cells in BD pathogenesis and strongly suggests a possible role for Bregs in the resolution of different BD manifestations.
Collapse
Affiliation(s)
- Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Correspondence: Helal F Hetta Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt Email
| | - Alaa A A Mohamed
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Asmaa M Zahran
- Clinical Pathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Safaa A Mahran
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwa MY Sayed
- Department of Cardiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mohamed GA Saleh
- Department of Ophthalmology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Khaled Abdelazeem
- Department of Ophthalmology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicines, Damanhour University, Damanhur, 22511, Egypt
| | - Salim Al-Rejaie
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Yasir Waheed
- Foundation University Medical College, Foundation University Islamabad, Islamabad, 44000, Pakistan
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Manal M Hassanien
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
27
|
Henke P, Sharma S, Wakefield T, Myers D, Obi A. Insights from experimental post-thrombotic syndrome and potential for novel therapies. Transl Res 2020; 225:95-104. [PMID: 32442728 PMCID: PMC7487018 DOI: 10.1016/j.trsl.2020.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022]
Abstract
Post-thrombotic syndrome (PTS) is an end stage manifestation of deep vein thrombosis. This is an inherently inflammatory process, with consequent fibrosis. Multiple cellular types are involved, and are likely driven by leukocytes. Herein, we review the current gaps in therapy, and insights from rodent models of venous thrombosis that suggest possible targets to treat and prevent PTS.
Collapse
Affiliation(s)
- Peter Henke
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI.
| | - Sriganesh Sharma
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Thomas Wakefield
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Dan Myers
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Andrea Obi
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| |
Collapse
|
28
|
Dectin-1 and TIM3 Expression in Deep Vein Thrombosis of Lower Limbs (DVTLL). J Clin Med 2020; 9:jcm9113466. [PMID: 33126475 PMCID: PMC7692188 DOI: 10.3390/jcm9113466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/19/2022] Open
Abstract
The pathophysiological mechanisms of venous thromboembolism are venous stasis, endothelial damage, and hypercoagulability, while less attention has been given to the role of both innate and native immunity. In this paper, we investigate the involvement of the activated immune system detected through some indicators such as TIM3 and Dectin-1 expressed by T lymphocytes. TIM3 and Dectin-1, two surface molecules that regulate the fine-tuning of innate and adaptive immune responses, were evaluated in patients affected by deep vein thrombosis of lower limbs (DVTLL). CD3+, CD4+ and CD8+ T lymphocytes obtained from patients affected by DVTLL were analysed using fluorescence-conjugated antibodies for TIM3 and Dectin-1 by an imaging flow cytometer. DVTLL patients showed a higher number of CD4+ and CD8+ T lymphocytes. TIM3 expression in T lymphocytes was very low in both DVTLL patients and controls. On the contrary, an increase in Dectin-1+ cells among CD4+ and CD8+ T lymphocytes from DVTLL patients was observed. Dectin-1 is known to play a role in inflammation and immunity and our result suggests its potential involvement in thrombotic venous disease.
Collapse
|
29
|
Campos J, Brill A. The role of bone marrow-derived cells in venous thromboembolism. Int J Biochem Cell Biol 2020; 128:105850. [PMID: 32950686 PMCID: PMC7607213 DOI: 10.1016/j.biocel.2020.105850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022]
Abstract
Venous thrombosis is a life-threatening condition with high morbidity and mortality. Abnormal functioning of different cells in the blood is an integral part of its pathogenesis. In this review, we describe the contribution of bone marrow-derived cells to the development of this debilitating disease. We present both epidemiological and clinical data demonstrating involvement of various cell types in venous thrombosis, and discuss potential mechanisms underlying these effects. Modern concepts including recently discovered new paradigms in thrombosis, such as neutrophil extracellular traps, mast cells, and polyphosphate, are summarized.
Collapse
Affiliation(s)
- Joana Campos
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
30
|
Abstract
The recent emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing global pandemic has presented a health emergency of unprecedented magnitude. Recent clinical data has highlighted that coronavirus disease 2019 (COVID-19) is associated with a significant risk of thrombotic complications ranging from microvascular thrombosis, venous thromboembolic disease, and stroke. Importantly, thrombotic complications are markers of severe COVID-19 and are associated with multiorgan failure and increased mortality. The evidence to date supports the concept that the thrombotic manifestations of severe COVID-19 are due to the ability of SARS-CoV-2 to invade endothelial cells via ACE-2 (angiotensin-converting enzyme 2), which is expressed on the endothelial cell surface. However, in patients with COVID-19 the subsequent endothelial inflammation, complement activation, thrombin generation, platelet, and leukocyte recruitment, and the initiation of innate and adaptive immune responses culminate in immunothrombosis, ultimately causing (micro)thrombotic complications, such as deep vein thrombosis, pulmonary embolism, and stroke. Accordingly, the activation of coagulation (eg, as measured with plasma D-dimer) and thrombocytopenia have emerged as prognostic markers in COVID-19. Given thrombotic complications are central determinants of the high mortality rate in COVID-19, strategies to prevent thrombosis are of critical importance. Several antithrombotic drugs have been proposed as potential therapies to prevent COVID-19-associated thrombosis, including heparin, FXII inhibitors, fibrinolytic drugs, nafamostat, and dipyridamole, many of which also possess pleiotropic anti-inflammatory or antiviral effects. The growing awareness and mechanistic understanding of the prothrombotic state of COVID-19 patients are driving efforts to more stringent diagnostic screening for thrombotic complications and to the early institution of antithrombotic drugs, for both the prevention and therapy of thrombotic complications. The shifting paradigm of diagnostic and treatment strategies holds significant promise to reduce the burden of thrombotic complications and ultimately improve the prognosis for patients with COVID-19.
Collapse
Affiliation(s)
- James D. McFadyen
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Hannah Stevens
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Karlheinz Peter
- Department of Cardiology (K.P.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| |
Collapse
|
31
|
The choline transporter Slc44a2 controls platelet activation and thrombosis by regulating mitochondrial function. Nat Commun 2020; 11:3479. [PMID: 32661250 PMCID: PMC7359028 DOI: 10.1038/s41467-020-17254-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
Genetic factors contribute to the risk of thrombotic diseases. Recent genome wide association studies have identified genetic loci including SLC44A2 which may regulate thrombosis. Here we show that Slc44a2 controls platelet activation and thrombosis by regulating mitochondrial energetics. We find that Slc44a2 null mice (Slc44a2(KO)) have increased bleeding times and delayed thrombosis compared to wild-type (Slc44a2(WT)) controls. Platelets from Slc44a2(KO) mice have impaired activation in response to thrombin. We discover that Slc44a2 mediates choline transport into mitochondria, where choline metabolism leads to an increase in mitochondrial oxygen consumption and ATP production. Platelets lacking Slc44a2 contain less ATP at rest, release less ATP when activated, and have an activation defect that can be rescued by exogenous ADP. Taken together, our data suggest that mitochondria require choline for maximum function, demonstrate the importance of mitochondrial metabolism to platelet activation, and reveal a mechanism by which Slc44a2 influences thrombosis. Genetic association studies have identified loci including the choline transporter SLC44A2 as a potential regulator of thrombosis. Here the authors report that loss of SLC44A2 impairs platelet activation and thrombosis in mice via a reduction of mitochondrial ATP production.
Collapse
|
32
|
Najem MY, Couturaud F, Lemarié CA. Cytokine and chemokine regulation of venous thromboembolism. J Thromb Haemost 2020; 18:1009-1019. [PMID: 32020753 DOI: 10.1111/jth.14759] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 12/27/2022]
Abstract
Morbidity and mortality from venous thromboembolism (VTE), which refers to deep vein thrombosis and pulmonary embolism, have a substantial effect on the global burden of disease. The field of venous thrombosis research has been dramatically changed over the past 10 years with the improvement of animal models that shed some light on the interaction between inflammation and thrombosis. Important recent advances provided evidence of the implication of the innate immune system in venous thrombosis. In this review, we highlighted the cytokines and chemokines that regulate mechanisms of thrombus formation and resolution. Cytokines are pleiotropic, redundant, and multifunctional endogenous mediators orchestrating the inflammatory responses leading to thrombus formation or resolution. The use of experimental models has revealed the pro-thrombotic activity of some cytokines including interferon-γ, interleukin (IL)-6, chemokine ligand 2, IL-17A, IL-9, IL-1β, and transforming growth factor-β. Other cytokines such as IL-10, tumor necrosis factor-α, and IL-8 appear to promote thrombus resolution in late phase of venous thromboembolism. The purpose of this review is to bring together the current knowledge regarding the cytokines and chemokines that have been involved in thrombosis formation and resolution. We postulate that an imbalance between pro-thrombotic and anti-thrombotic cytokines/chemokines may be involved in the pathophysiology of VTE. However, in-depth basic and clinical research in venous thrombosis is still require to fully understand the precise mechanism of action of these cytokines.
Collapse
Affiliation(s)
- Maria Y Najem
- EA3878 (GETBO), Brest Hospital, Univ Brest, Brest, France
| | | | - Catherine A Lemarié
- EA3878 (GETBO), Brest Hospital, Univ Brest, Brest, France
- INSERM 1078, Brest, France
| |
Collapse
|
33
|
Mahmoudpour SH, Valerio L, Douxfils J, Mahan CE, Jankowski M, Quitzau K, Konstantinides SV, Barco S. Potential Drug Interactions between Recombinant Interleukin-2 and Direct Oral Anticoagulants: Indirect Evidence from In Vivo Animal Studies. Hamostaseologie 2020; 40:679-686. [PMID: 32325520 DOI: 10.1055/a-1120-4064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Recombinant interleukin-2 (rIL-2) is indicated for metastatic renal cell carcinoma and melanoma. Over recent years low-dose rIL-2 has been studied for the treatment of autoimmune diseases and acute coronary syndrome because of its ability to expand and activate T regulatory (Treg) cells. However, several medical conditions potentially benefiting from rIL-2 administrations are characterized by an intrinsic prothrombotic risk, thus requiring concurrent anticoagulation. In our systematic review of the literature, we investigated the potential for drug interactions between oral anticoagulants and rIL-2 by assessing the influence of rIL-2 administration on transporters and cytochromes determining the pharmacokinetics of (direct) oral anticoagulants. We extracted data from 12 studies, consisting of 11 animal studies and one study in humans. Eight studies investigated the pharmacokinetics of P-glycoprotein (P-gp) substrates and reported that the intraperitoneal rIL-2 administration may inhibit intestinal P-gp. Four studies on hepatic cytochrome P450 yielded conflicting results. The only human study included in this systematic review concluded that rIL-2 suppresses the hepatic cytochrome P450, but only if given at higher doses. Based on the results from animal studies, the co-administration of rIL-2 and dabigatran etexilate, a substrate of intestinal P-gp, may lead to higher dabigatran plasma concentrations and bioavailability. Human studies should confirm whether this potential interaction is clinically relevant.
Collapse
Affiliation(s)
- Seyed Hamidreza Mahmoudpour
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany.,Department of Biometry and Bioinformatics, Institute for Medical Biostatistics, Epidemiology, and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Luca Valerio
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Jonathan Douxfils
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Charles E Mahan
- Presbyterian Healthcare Services, University of New Mexico, Albuquerque, United States
| | - Marius Jankowski
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Kurt Quitzau
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Stavros V Konstantinides
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Stefano Barco
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany.,Clinic of Angiology, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Nicklas JM, Gordon AE, Henke PK. Resolution of Deep Venous Thrombosis: Proposed Immune Paradigms. Int J Mol Sci 2020; 21:E2080. [PMID: 32197363 PMCID: PMC7139924 DOI: 10.3390/ijms21062080] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/12/2022] Open
Abstract
Venous thromboembolism (VTE) is a pathology encompassing deep vein thrombosis (DVT) and pulmonary embolism (PE) associated with high morbidity and mortality. Because patients often present after a thrombus has already formed, the mechanisms that drive DVT resolution are being investigated in search of treatment. Herein, we review the current literature, including the molecular mechanisms of fibrinolysis and collagenolysis, as well as the critical cellular roles of macrophages, neutrophils, and endothelial cells. We propose two general models for the operation of the immune system in the context of venous thrombosis. In early thrombus resolution, neutrophil influx stabilizes the tissue through NETosis. Meanwhile, macrophages and intact neutrophils recognize the extracellular DNA by the TLR9 receptor and induce fibrosis, a complimentary stabilization method. At later stages of resolution, pro-inflammatory macrophages police the thrombus for pathogens, a role supported by both T-cells and mast cells. Once they verify sterility, these macrophages transform into their pro-resolving phenotype. Endothelial cells both coat the stabilized thrombus, a necessary early step, and can undergo an endothelial-mesenchymal transition, which impedes DVT resolution. Several of these interactions hold promise for future therapy.
Collapse
Affiliation(s)
| | | | - Peter K. Henke
- School of Medicine, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA; (J.M.N.); (A.E.G.)
| |
Collapse
|
35
|
Depletion of CD4 and CD8 Positive T Cells Impairs Venous Thrombus Resolution in Mice. Int J Mol Sci 2020; 21:ijms21051650. [PMID: 32121269 PMCID: PMC7084232 DOI: 10.3390/ijms21051650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Resolution of deep venous thrombosis involves coordinated inflammatory processes. T cells regulate inflammation in vivo and modulate vascular remodeling in other settings, but their role in venous thrombus resolution remains undefined. To determine the role of T cells in venous thrombus resolution in vivo, stasis induced thrombi were created by vena cava ligation in outbred CD-1 mice. CD4 and CD8 positive T cells, as determined by flow cytometry, were present in thrombi both during thrombus formation and resolution. Depletion of the CD4 and CD8 positive T cells by antibody treatment selectively impaired thrombus resolution compared to animals treated with isotype control antibodies, without an effect on venous thrombus formation. Quantitation of intra-thrombus macrophage numbers, fibrinolytic marker expression, and gelatinolytic activity by zymography revealed that T cell depletion decreased the number of macrophages, reduced the expression of fibrinolytic marker urokinase plasminogen activator (uPA), and decreased the activity of matrix metalloprotinease-9 (MMP-9). These data implicate CD4 and CD8 positive T cells in functionally contributing to venous thrombus resolution, thus representing a potential therapeutic target, but also underscoring potential risks involved in T cell depletion used clinically for solid organ and hematopoietic transplantation procedures.
Collapse
|
36
|
Nosaka M, Ishida Y, Kimura A, Kuninaka Y, Taruya A, Ozaki M, Tanaka A, Mukaida N, Kondo T. Crucial Involvement of IL-6 in Thrombus Resolution in Mice via Macrophage Recruitment and the Induction of Proteolytic Enzymes. Front Immunol 2020; 10:3150. [PMID: 32117207 PMCID: PMC7019028 DOI: 10.3389/fimmu.2019.03150] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/30/2019] [Indexed: 01/08/2023] Open
Abstract
After the ligation of the inferior vena cava (IVC) of wild-type (WT) mice, venous thrombi formed and grew progressively until 5 days and resolved thereafter. Concomitantly, intrathrombotic gene expression of Il6 was enhanced later than 5 days after IVC ligation. IL-6 protein expression was detected mainly in F4/80-positive macrophages in thrombus. When Il6-deficient (Il6−/−) mice were treated in the same manner, thrombus mass was significantly larger than in WT mice. Moreover, the recovery of thrombosed IVC blood flow was markedly delayed in Il6−/− compared with WT mice. F4/80-positive macrophages in thrombus expressed proteolytic enzymes such as matrix metalloproteinase (Mmp) 2, Mmp9, and urokinase-type plasminogen activator (Plau); and their mRNA expression was significantly reduced in Il6−/− mice. Consistently, the administration of anti-IL-6 antibody delayed the thrombus resolution in WT mice, whereas IL-6 administration accelerated thrombus resolution in WT and Il6−/− mice. Moreover, IL-6 in vitro enhanced Mmp2, Mmp9, and Plau mRNA expression in WT-derived peritoneal macrophages in a dose-dependent manner; and the enhancement was abrogated by a specific Stat3 inhibitor, Stattic. Thus, IL-6/Stat3 signaling pathway can promote thrombus resolution by enhancing Mmp2, Mmp9, and Plau expression in macrophages.
Collapse
Affiliation(s)
- Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akira Taruya
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mitsunori Ozaki
- Department of Neurological Surgery, Wakayama Medical University, Wakayama, Japan
| | - Atushi Tanaka
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
37
|
Li D, Zhang X, Zhang H, Li X. Synergic effect of GPIBA and von Willebrand factor in pathogenesis of deep vein thrombosis. Vascular 2020; 28:309-313. [PMID: 31902309 DOI: 10.1177/1708538119896446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES In cardiovascular disease, deep vein thrombosis is one of the vital symptoms causing pulmonary thromboembolism. However, the pathogenesis of deep vein thrombosis is still not clear. One of the critical factors leading to deep vein thrombosis is the platelet aggregation that is mediated by a set of key genes including platelet membrane protein coded by platelet glycoprotein Ib alpha chain (GPIBA). METHODS Deep vein thrombosis model was established according to the previous protocol, and venous blood and thrombi were collected for further analysis. RESULTS The dynamic changes of GPIBA and coagulation factor, von Willebrand factor, were observed in deep vein thrombosis models. Meanwhile, critical proteins participating in adhesion and binding of platelets such as epithelial membrane protein 2 (EMP2), vascular cell adhesion protein 1 (VCAM1), immunoreceptor tyrosine-based activation motif 1 (ITAM1), integrin subunit alpha M (ITGAM), or fibronectin were also differentially expressed in deep vein thrombosis models. CONCLUSIONS Application of heparin could reverse these dynamic changes in deep vein thrombosis models. Thus, we explained the potential synergic role of GPIBA and von Willebrand factor in regulating the occurrence of deep vein thrombosis and provide therapeutic target against cardiovascular disease.
Collapse
Affiliation(s)
- Da Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Soochow University, Suzhou, China.,Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaosong Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Honggang Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Soochow University, Suzhou, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
38
|
Patsouras MD, Vlachoyiannopoulos PG. Evidence of epigenetic alterations in thrombosis and coagulation: A systematic review. J Autoimmun 2019; 104:102347. [PMID: 31607428 DOI: 10.1016/j.jaut.2019.102347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Thrombosis in the context of Cardiovascular disease (CVD) affects mainly the blood vessels supplying the heart, brain and peripheries and it is the leading cause of death worldwide. The pathophysiological thrombotic mechanisms are largely unknown. Heritability contributes to a 30% of the incidence of CVD. The remaining variation can be explained by life style factors such as smoking, dietary and exercise habits, environmental exposure to toxins, and drug usage and other comorbidities. Epigenetic variation can be acquired or inherited and constitutes an interaction between genes and the environment. Epigenetics have been implicated in atherosclerosis, ischemia/reperfusion damage and the cardiovascular response to hypoxia. Epigenetic regulators of gene expression are mainly the methylation of CpG islands, histone post translational modifications (PTMs) and microRNAs (miRNAs). These epigenetic regulators control gene expression either through activation or silencing. Epigenetic control is mostly dynamic and can potentially be manipulated to prevent or reverse the uncontrolled expression of genes, a trait that renders them putative therapeutic targets. In the current review, we systematically studied and present available data on epigenetic alterations implicated in thrombosis derived from human studies. Evidence of epigenetic alterations is observed in several thrombotic diseases such as Coronary Artery Disease and Cerebrovascular Disease, Preeclampsia and Antiphospholipid Syndrome. Differential CpG methylation and specific histone PTMs that control transcription of prothrombotic and proinflammatory genes have also been associated with predisposing factors of thrombosis and CVD, such us smoking, air pollution, hypertriglyceridemia, occupational exposure to particulate matter and comorbidities including cancer, Chronic Obstructive Pulmonary Disease and Chronic Kidney Disease. These clinical observations are further supported by in vitro experiments and indicate that epigenetic regulation affects the pathophysiology of thrombotic disorders with potential diagnostic or therapeutic utility.
Collapse
Affiliation(s)
- M D Patsouras
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - P G Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
39
|
Liu H, Li P, Lin J, Chen W, Guo H, Lin J, Liu J, Lu Z, Yao X, Chen Y, Lin B. Danhong Huayu Koufuye prevents venous thrombosis through antiinflammation via Sirtuin 1/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111975. [PMID: 31141719 DOI: 10.1016/j.jep.2019.111975] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/08/2019] [Accepted: 05/23/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danhong Huayu Koufuye (DHK), a compound traditional Chinese medicine, is composed of Salvia miltiorrhiza radix (Salvia miltiorrhiza Bge.), Angelicae Sinensis radix (Angelicae Sinensis (Oliv.) Diels.), Chuanxiong rhizoma (Ligusticum chuanxiong Hort.), Persicae semen (Prunus persica (L.) Batsch), Carthami flos (Carthamus tinctorius L.), Bupleuri radix (Bupleurum chinense DC.) and Aurantii fructus (Citrus aurantium L.). DHK prevents deep vein thrombosis (DVT) through antiinflammation. However, the antiinflammatory mechanism of DHK is still unknown. OBJECTIVE The aim of this study was to evaluate whether DHK prevented venous thrombosis through antiinflammation via Sirtuin 1 (SIRT1)/NF-κB signaling pathway. METHODS Inferior vena cava (IVC) stenosis-induced DVT rat model was established. Rats were administered with DHK (1.6, 3.2 or 6.4 mL/kg/d, p.o.), heparin (200 U/kg/d, i.v.), clopidogrel (25 mg/kg/d, p.o.), resveratrol (50 mg/kg/d, p.o.) or vehicle (p.o.) once daily for two days. Blood coagulation, blood fibrinolysis, blood viscosity, blood cell counts and platelet activity were evaluated. Serum levels of inflammatory cytokines were analyzed by enzyme-linked immunosorbent assay. Pathological changes were observed by hematoxylin-eosin (HE) staining. Protein expressions in thrombosed IVCs were evaluated by Western blot and/or immunofluorescence analyses. SIRT1 mRNA expression was analyzed by real-time quantitative polymerase chain reaction. Besides, SIRT1-specific inhibitor EX527 was pretreated to confirm the role of SIRT1/NF-κB signaling pathway in the antithrombotic effect of DHK. RESULTS DHK remarkably prevented DVT. DHK had no effects on blood coagulation, blood fibrinolysis, blood viscosity, blood cell counts or platelet activity. But DHK significantly up-regulated protein and mRNA expressions of SIRT1, and reduced leukocytes infiltration into thrombus and vein wall, serum levels of inflammatory cytokines, and protein expressions of acetylated p65 (Ace-p65), phosphorylated p65 (p-p65) and tissue factor (TF). Moreover, the antithrombotic effect of DHK was significantly abolished by EX527. CONCLUSION DHK may prevent DVT by inhibiting inflammation via SIRT1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Han Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Peng Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Juan Lin
- Guangzhou Baiyun Mountain and Hutchison Whampoa Ltd., Guangzhou, Guangdong, 510515, China
| | - Wenpei Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Haibiao Guo
- Guangzhou Baiyun Mountain and Hutchison Whampoa Ltd., Guangzhou, Guangdong, 510515, China
| | - Jianyun Lin
- Guangzhou Baiyun Mountain and Hutchison Whampoa Ltd., Guangzhou, Guangdong, 510515, China
| | - Jin Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ziqi Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xiaolan Yao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yang Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Baoqin Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
40
|
Mukhopadhyay S, Johnson TA, Duru N, Buzza MS, Pawar NR, Sarkar R, Antalis TM. Fibrinolysis and Inflammation in Venous Thrombus Resolution. Front Immunol 2019; 10:1348. [PMID: 31258531 PMCID: PMC6587539 DOI: 10.3389/fimmu.2019.01348] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Clinical observations and accumulating laboratory evidence support a complex interplay between coagulation, inflammation, innate immunity and fibrinolysis in venous thromboembolism (VTE). VTE, which includes deep vein thrombosis (DVT) and pulmonary embolism (PE), and the subsequent complications of post-thrombotic syndrome (PTS), are significant causes of morbidity and mortality in patients. Clinical risk factors for VTE include cancer, major trauma, surgery, sepsis, inflammatory bowel disease, paralysis, prolonged periods of immobility, and aging. Abnormalities in venous blood flow or stasis initiates the activation of endothelial cells, and in concert with platelets, neutrophils and monocytes, propagates VTE in an intact vein. In addition, inflammatory cells play crucial roles in thrombus recanalization and restoration of blood flow via fibrinolysis and vascular remodeling. Faster resolution of the thrombus is key for improved disease prognosis. While in the clinical setting, anticoagulation therapy is successful in preventing propagation of venous thrombi, current therapies are not designed to inhibit inflammation, which can lead to the development of PTS. Animal models of DVT have provided many insights into the molecular and cellular mechanisms involved in the formation, propagation, and resolution of venous thrombi as well as the roles of key components of the fibrinolytic system in these processes. Here, we review the recent advances in our understanding of fibrinolysis and inflammation in the resolution of VTE.
Collapse
Affiliation(s)
- Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tierra A. Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
41
|
Safety of low-dose subcutaneous recombinant interleukin-2: systematic review and meta-analysis of randomized controlled trials. Sci Rep 2019; 9:7145. [PMID: 31073219 PMCID: PMC6509335 DOI: 10.1038/s41598-019-43530-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/26/2019] [Indexed: 02/08/2023] Open
Abstract
Standard-dose intravenous recombinant interleukin-2 (rIL-2) is indicated for the treatment of some subtypes of cancer; however, severe adverse events, including venous thromboembolism (VTE), may complicate its administration. Low-dose subcutaneous rIL-2 is being studied for the management of immune-mediated diseases, since it can modulate the immunological response by specifically targeting T regulatory (Treg) cells; importantly, it is supposed to cause fewer or no complications. In this systematic review and meta-analysis of phase II-III randomized controlled trials (RCTs), we investigated the safety of low-dose (<6 Million International Unit [MIU]/day) and ultra-low-dose (≤1 MIU/day) rIL-2 for severe adverse events (grade III-V) with a focus on VTE. Data of 1,321 patients from 24 RCTs were analysed: 661 patients were randomized to the rIL-2 arm (on top of standard of care) and 660 patients to standard of care alone or placebo. Two studies reported higher rates of thrombocytopenia in the low-dose rIL-2 arm. Ultra-low-dose rIL-2 was reported to be well tolerated in 6 studies with a negligible rate of severe adverse events. Symptomatic VTE events were not reported in any of the study arms (absolute risk difference 0% [95%CI −0.1%; +0.1%]). Our results may facilitate the study and introduction in clinical practice of low-dose rIL-2 for potentially new indications.
Collapse
|
42
|
Bertin FR, Rys RN, Mathieu C, Laurance S, Lemarié CA, Blostein MD. Natural killer cells induce neutrophil extracellular trap formation in venous thrombosis. J Thromb Haemost 2019; 17:403-414. [PMID: 30456926 DOI: 10.1111/jth.14339] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Indexed: 02/04/2023]
Abstract
Essentials Neutrophil extracellular traps (NETs) are generated during deep vein thrombosis (DVT). The role of interferon γ (IFNγ) and natural killer (NK) cells in NET formation was studied. IFNγ promote venous thrombosis through NET formation. NK cell depletion reduces DVT. SUMMARY: Background Neutrophils contribute to venous thrombosis through the release of neutrophil extracellular traps (NETs), but the mechanism triggering their formation remains unclear. In vitro data show that interferon (IFN)-γ induces the formation of NETs. Objectives To determine whether IFN-γ and the transcription factor T-box expressed on T cells (Tbet) promote venous thrombosis through neutrophil activation. Methods Venous thrombosis was induced by flow restriction in the inferior vena cava in IFN-γ-/- , Tbet-/- or wild-type (WT) mice. After 48 h, thrombus size was measured by the use of high-frequency ultrasound. NET formation was determined by immunofluorescence. Results and Conclusions Thrombus formation was reduced in Tbet-/- and IFN-γ-/- mice, suggesting that Tbet/IFN-γ-expressing cells are required for venous thrombosis. The number of NETs formed during thrombosis was significantly lower in Tbet-/- and IFN-γ-/- mice. NET formation was also decreased in WT mice treated with an IFN-γ-blocking antibody. Injection of recombinant IFN-γ into IFN-γ-/- mice rescued the phenotype. Natural killer (NK) cells were specifically depleted prior to venous thrombosis induction. NK cell depletion results in decreased NET formation and smaller thrombi, suggesting that NK cells are required for thrombus development. In depleted mice, adoptive transfer of WT NK cells induced a similar thrombosis burden as in WT mice. In contrast, adoptive transfer of IFN-γ -/- NK cells resulted in thrombi similar in size to those in depleted mice. In vitro, we showed that WT neutrophils released fewer NETs when they were cocultured with IFN-γ-/- NK cells. This study demonstrates that NK cell-dependent IFN-γ production is crucial for thrombus development by promoting the formation of NETs by neutrophils.
Collapse
Affiliation(s)
- F-R Bertin
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - R N Rys
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
| | - C Mathieu
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
| | - S Laurance
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
| | - C A Lemarié
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Québec, Canada
- EA3878 (GETBO), University Hospital of Brest, European University of Occidental Brittany, Brest, France
| | - M D Blostein
- Lady Davis Institute for Medical Research, Montreal, Québec, Canada
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
43
|
Becker C, Reinhardt C. Unexpected role of natural killer cell-derived interferon-γ as a driver of NETosis and DVT. J Thromb Haemost 2019; 17:400-402. [PMID: 30580491 DOI: 10.1111/jth.14368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Christoph Becker
- Dermatology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christian Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany
| |
Collapse
|
44
|
Budnik I, Brill A. Immune Factors in Deep Vein Thrombosis Initiation. Trends Immunol 2018; 39:610-623. [PMID: 29776849 PMCID: PMC6065414 DOI: 10.1016/j.it.2018.04.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
Deep vein thrombosis (DVT) is a major origin of morbidity and mortality. While DVT has long been considered as blood coagulation disorder, several recent lines of evidence demonstrate that immune cells and inflammatory processes are involved in DVT initiation. Here, we discuss these mechanisms, in particular, the role of immune cells in endothelial activation, and the immune cascades leading to expression of adhesion receptors on endothelial cells. We analyze the specific recruitment and functional roles of different immune cells, such as mast cells and leukocytes, in DVT. Importantly, we also speculate how immune modulation could be used for DVT prevention with a lower risk of bleeding complications than conventional therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Brill
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
45
|
Pulmonary Arterial Hypertension and Endothelial Dysfunction Is Linked to NADPH Oxidase-Derived Superoxide Formation in Venous Thrombosis and Pulmonary Embolism in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1860513. [PMID: 29983855 PMCID: PMC6015670 DOI: 10.1155/2018/1860513] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/11/2018] [Accepted: 02/28/2018] [Indexed: 12/30/2022]
Abstract
Pulmonary embolism (PE) results from deep vein thrombosis (DVT) and can lead to chronic thromboembolic pulmonary hypertension (CTEPH) involving vascular dysfunction. Mechanisms are incompletely understood, in part due to lack of mouse models. We induced PE in C57BL/6 mice by intravenous injection of thrombin (166 U/kg BW), confirmed by a sudden bradycardia, bradypnea, and an increase in pulmonary artery (PA) pressure observed by high-frequency ultrasound. While symptoms resolved rapidly after single thrombin application, repeated PEs resulted in sustained PA-pressure increase, increased PA superoxide formation assessed by oxidative fluorescent microtopography, increased PA gp91phox expression, and endothelial dysfunction assessed by isometric tension studies of isolated PA segments after 24 hours. DVT was modeled in C57BL/6 mice by ligation of the inferior vena cava (IVC). Importantly, small pulmonary emboli could be detected along with a mild phenotype of PA endothelial dysfunction and oxidative stress in the absence of PA-pressure elevation. mRNA expression of plasminogen activator inhibitor-1 was increased in PAs of mice with recurrent PE after repetitive thrombin injections and to a lesser extent in DVT mice. In summary, our data suggest that PA endothelial dysfunction, induced by gp91phox-derived ROS, is an early event upon repetitive PE. This phenomenon might help to elucidate the mechanisms of PA dysfunction in the pathogenesis of CTEPH.
Collapse
|
46
|
Agabiti-Rosei C, Trapletti V, Piantoni S, Airò P, Tincani A, De Ciuceis C, Rossini C, Mittempergher F, Titi A, Portolani N, Caletti S, Coschignano MA, Porteri E, Tiberio GAM, Pileri P, Solaini L, Kumar R, Ministrini S, Agabiti Rosei E, Rizzoni D. Decreased circulating T regulatory lymphocytes in obese patients undergoing bariatric surgery. PLoS One 2018; 13:e0197178. [PMID: 29758052 PMCID: PMC5951588 DOI: 10.1371/journal.pone.0197178] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 04/27/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE It has been previously demonstrated that T lymphocytes may be involved in the development of hypertension and microvascular remodeling, and that circulating T effector lymphocytes may be increased in hypertension. In particular, Th1 and Th 17 lymphocytes may contribute to the progression of hypertension and microvascular damage while T-regulatory (Treg) lymphocytes seem to be protective in this regard. However, no data is available about patients with severe obesity, in which pronounced microvascular alterations were observed. DESIGN AND METHODS We have investigated 32 severely obese patients undergoing bariatric surgery, as well as 24 normotensive lean subjects and 12 hypertensive lean subjects undergoing an elective surgical intervention. A peripheral blood sample was obtained before surgery for assessment of CD4+ T lymphocyte subpopulations. Lymphocyte phenotype was evaluated by flow cytometry in order to assess T-effector and Treg lymphocytes. RESULTS A marked reduction of several Treg subpopulations was observed in obese patients compared with controls, together with an increased in CD4+ effector memory T-effector cells. CONCLUSION In severely obese patients, Treg lymphocytes are clearly reduced and CD4+ effector memory cells are increased. It may be hypothesized that they might contribute to the development of marked microvascular alterations previously observed in these patients.
Collapse
Affiliation(s)
- Claudia Agabiti-Rosei
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Valentina Trapletti
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Chair of Rheumatology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Paolo Airò
- Chair of Rheumatology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Angela Tincani
- Chair of Rheumatology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carolina De Ciuceis
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Claudia Rossini
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesco Mittempergher
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Amin Titi
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Nazario Portolani
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Caletti
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Enzo Porteri
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Guido A. M. Tiberio
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Paola Pileri
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Leonardo Solaini
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Rajesh Kumar
- Chair of Rheumatology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Ministrini
- Clinica Chirurgica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Enrico Agabiti Rosei
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Damiano Rizzoni
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Istituto Clinico Città di Brescia, Division of Medicine, Brescia, Italy
| |
Collapse
|
47
|
Kunimasa K, Nishino K, Kimura M, Inoue T, Tamiya M, Kumagai T, Imamura F. Pembrolizumab-induced acute thrombosis: A case report. Medicine (Baltimore) 2018; 97:e10772. [PMID: 29768369 PMCID: PMC5976311 DOI: 10.1097/md.0000000000010772] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
RATIONALE Acute thrombosis has not been reported in the literature so far in lung cancer patients as an immune-related adverse event (irAE) associated with PD-1 pathway inhibitors. PATIENTS CONCERNS Here, we for the first time present two NSCLC (non-small cell lung cancer) patients suffering from acute thrombosis as a pembrolizumab-induced irAE. Immediate treatment with continuous heparin infusion improved their symptoms and enabled them to continue pembrolizumab administration. METHODS Ethical approval was given by the ethics committee of Osaka International Cancer Institute and the informed consents were given by the patients. DIAGNOSIS Serum D-dimer level testing, venous ultrasonography, enhanced computed tomography (CT). INTERVENTIONS Continuous heparin infusion, direct oral anticoagulants (DOAC). OUTCOMES Immediate continuous heparin infusion improved their symptoms and continuing pembrolizumab with direct oral anticoagulant successfully induced tumor shrinkage. LESSONS Reinvigoration of exhausted T cells by pembrolizumab induced systemic inflammation possibly resulting in development of thrombosis. Although acute thrombosis is a rare irAE, it may lead to cessation of treatment and can be lethal.
Collapse
|
48
|
Rys RN, Blostein MD, Lemarié CA. Deep Vein Thrombosis Induced by Stasis in Mice Monitored by High Frequency Ultrasonography. J Vis Exp 2018. [PMID: 29708541 DOI: 10.3791/57392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Venous thrombosis is a common condition affecting 1 - 2% of the population, with an annual incidence of 1 in 500. Venous thrombosis can lead to death through pulmonary embolism or results in the post-thrombotic syndrome, characterized by chronic leg pain, swelling, and ulceration, or in chronic pulmonary hypertension resulting in significant chronic respiratory compromise. This is the most common cardiovascular disease after myocardial infarction and ischemic stroke and is a clinical challenge for all medical disciplines, as it can complicate the course of other disorders such as cancer, systemic disease, surgery, and major trauma. Experimental models are necessary to study these mechanisms. The stasis model induces consistent thrombus size and a quantifiable amount of thrombus. However, it is necessary to systematically ligate side branches of the inferior vena cava to avoid variability in thrombus sizes and any erroneous data interpretation. We have developed a non-invasive technique to measure thrombus size using ultrasonography. Using this technique, we can assess thrombus development and resolution over time in the same animal. This approach limits the number of mice required for quantification of venous thrombosis consistent with the principle of replacement, reduction, and refinement of animals in research. We have demonstrated that thrombus weight and histological analysis of thrombus size correlate with measurement obtained with ultrasonography. Therefore, the current study describes how to induce deep vein thrombosis in mice using the inferior vena cava stasis model and how to monitor it using high frequency ultrasound.
Collapse
Affiliation(s)
- Ryan N Rys
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University
| | - Mark D Blostein
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University; Department of Medicine, Jewish General Hospital, McGill University
| | - Catherine A Lemarié
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University; Department of Medicine, Jewish General Hospital, McGill University;
| |
Collapse
|
49
|
Prochaska JH, Luther N, Brähler M, Schulz A, Hermanns MI, Lackner KJ, Espinola-Klein C, Münzel T, Wild PS, Becker C. Acute deep vein thrombosis suppresses peripheral T cell effector function. Br J Haematol 2018; 184:847-850. [PMID: 29574858 DOI: 10.1111/bjh.15192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jürgen H Prochaska
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Preventive Cardiology and Preventive Medicine - Centre for Cardiology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Natascha Luther
- Department of Dermatology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Melanie Brähler
- Department of Dermatology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine - Centre for Cardiology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - M Iris Hermanns
- Preventive Cardiology and Preventive Medicine - Centre for Cardiology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,School of Chemistry, Biology and Pharmacy, Fresenius University of Applied Sciences, Mainz, Germany
| | - Karl J Lackner
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christine Espinola-Klein
- Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Centre for Cardiology - Cardiology I, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Centre for Cardiology - Cardiology I, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp S Wild
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Preventive Cardiology and Preventive Medicine - Centre for Cardiology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main Mainz, Mainz, Germany.,Centre for Translational Vascular Biology (CTVB), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christian Becker
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Department of Dermatology, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
50
|
Obermayer G, Afonyushkin T, Binder CJ. Oxidized low-density lipoprotein in inflammation-driven thrombosis. J Thromb Haemost 2018; 16:418-428. [PMID: 29316215 DOI: 10.1111/jth.13925] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thrombosis is the defining feature of the most prevalent causes of cardiovascular mortality, such as myocardial infarction, stroke, and pulmonary artery embolism. Although platelet activation and activation of the plasmatic coagulation system are the hallmarks of thrombus formation, inflammatory processes and the cellular responses involved are increasingly being recognized as critical modulators of thrombosis. In the context of many chronic inflammatory diseases that are associated with a high thrombotic risk, oxidized lipoproteins represent a prominent sterile trigger of inflammation. Oxidized low-density lipoprotein and its components play a central role in the initiation and progression of atherosclerotic plaques, but also in other processes that lead to thrombotic events. Moreover, dying cells and microvesicles can be decorated with some of the same oxidized lipid components that are found on oxidized lipoproteins, and thereby similar mechanisms of thromboinflammation may also be active in venous thrombosis. In this review, we summarize the current knowledge on how oxidized lipoproteins and components thereof affect the cells and pathways involved in thrombosis.
Collapse
Affiliation(s)
- G Obermayer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - T Afonyushkin
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - C J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|