1
|
Griem K, Falter T, Hollerbach A, Jurk K, Aboulmaouahib B, Weinmann-Menke J, Müller-Calleja N, Lackner KJ. Association of Specific Antiphospholipid Antibodies to Platelet Count and Thrombocytopenia. Thromb Haemost 2025. [PMID: 39775760 DOI: 10.1055/a-2510-6147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND Thrombocytopenia is one of the most common manifestations of the antiphospholipid syndrome (APS). However, its causes are still poorly defined. We have shown recently that antiphospholipid antibodies (aPL) directed against β2-glycoprotein I (β2GPI) of the IgG isotype induced platelet activation and aggregation while aPL directed against cardiolipin and anti-β2GPI IgM had no effect. Since platelet activation by anti-β2GPI might lead to platelet consumption and lower platelet count or overt thrombocytopenia, we analyzed the association of aPL with platelet count. MATERIAL AND METHODS Data of consecutive patients with test orders for anticardiolipin IgG/IgM and anti-β2GPI IgG/IgM and full blood count in our laboratory from August 2015 to April 2019 were analyzed. RESULTS We identified 2,815 individual patients (mean age 45.7 years; 71.1% women) with complete data on aPL and platelet count, of which 445 individuals (mean age 41.0 years; 75.3% women) showed increased aPL titers. Patients with anti-β2GPI of the IgG isotype had significantly lower platelet count (220 ± 84 versus 264 ± 90 G/L, p < 0.0001) and higher frequency of thrombocytopenia (platelet count <100 G/L; 12.2% versus 2.4%, p < 0.005) than patients negative for all four aPL. These differences could not be explained by comorbidities or medications. Neither anticardiolipin IgG nor aPL of the IgM isotype was associated with lower platelet count or thrombocytopenia. CONCLUSION The exclusive association of anti-β2GPI IgG aPL with low platelet count coincides with its unique ability to activate platelets and induce aggregation in vitro. This supports the hypothesis that anti-β2GPI IgG aPL may induce thrombocytopenia by chronic platelet consumption in vivo.
Collapse
Affiliation(s)
- Katharina Griem
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Tanja Falter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Brahim Aboulmaouahib
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | | | - Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
2
|
Bucci T, Merashli M, Pignatelli P, Pastori D, Delgado-Alves J, Lip GYH, Ames PRJ. Intima media thickness of the carotid artery in primary antiphospholipid syndrome: A systematic review and meta-analysis. Autoimmun Rev 2024; 23:103657. [PMID: 39366515 DOI: 10.1016/j.autrev.2024.103657] [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: 05/30/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Primary antiphospholipid syndrome (PAPS) has been associated with an increase in clinical events associated with atherosclerotic vascular disease. Intima media thickness (IMT) of carotid arteries is a surrogate marker of atherosclerotic vascular disease. OBJECTIVES To conduct a systematic review and meta-analysis of studies evaluating IMT and their clinical correlates in PAPS. METHODS Systematic search of EMBASE and PubMed databases from January 2000 to December 2023; we employed random effect meta-analyses for continuous outcomes and Peto's odds ratio for rare events. RESULTS The meta-analysis included 21 studies (20 case control and 1 cohort) showing that PAPS patients (n = 1103) had thicker IM than controls (n = 832) (p < 0.0001) with wide heterogeneity (I2 = 86.9 %); PAPS patients (n = 782) also had a greater pooled prevalence of carotid plaques than controls (n = 537) (13.1 % vs 2.97 %, p < 0.0001). A sensitivity analysis by meta-regression indicated that mean age, gender, disease duration, lipid profile, blood pressures, smoking and statin use all explained the heterogeneity variance; a sensitivity analysis by subgroups confirmed smoking status and statin use as explanatory variables with the addition of ethnicity. CONCLUSION Atherosclerosis of the carotid artery represents a clinical feature of PAPS in relation to the traditional risk factors and to statin use. Minimising the atherogenic risk with statins could reduce the late arterial atherothrombotic risks of PAPS.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Mira Merashli
- Department of Rheumatology, American University of Beirut, Beirut, Lebanon
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Daniele Pastori
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; IRCCS Neuromed, Localita' Camerelle, Pozzilli, Italy
| | - Jose' Delgado-Alves
- Immune Response & Vascular Disease, iNOVA,4Health, Nova Medical School, Nova University Lisbon, Lisbon, Portugal
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Denmark
| | - Paul R J Ames
- Immune Response & Vascular Disease, iNOVA,4Health, Nova Medical School, Nova University Lisbon, Lisbon, Portugal; Department of Haematology, Dumfries Royal Infirmary, Cargenbridge, Dumfries, UK.
| |
Collapse
|
3
|
Kumar R, Patil G, Dayal S. NLRP3-Induced NETosis: A Potential Therapeutic Target for Ischemic Thrombotic Diseases? Cells 2023; 12:2709. [PMID: 38067137 PMCID: PMC10706381 DOI: 10.3390/cells12232709] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Ischemic thrombotic disease, characterized by the formation of obstructive blood clots within arteries or veins, is a condition associated with life-threatening events, such as stroke, myocardial infarction, deep vein thrombosis, and pulmonary embolism. The conventional therapeutic strategy relies on treatments with anticoagulants that unfortunately pose an inherent risk of bleeding complications. These anticoagulants primarily target clotting factors, often overlooking upstream events, including the release of neutrophil extracellular traps (NETs). Neutrophils are integral components of the innate immune system, traditionally known for their role in combating pathogens through NET formation. Emerging evidence has now revealed that NETs contribute to a prothrombotic milieu by promoting platelet activation, increasing thrombin generation, and providing a scaffold for clot formation. Additionally, NET components enhance clot stability and resistance to fibrinolysis. Clinical and preclinical studies have underscored the mechanistic involvement of NETs in the pathogenesis of thrombotic complications, since the clots obtained from patients and experimental models consistently exhibit the presence of NETs. Given these insights, the inhibition of NETs or NET formation is emerging as a promising therapeutic approach for ischemic thrombotic diseases. Recent investigations also implicate a role for the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome as a mediator of NETosis and thrombosis, suggesting that NLRP3 inhibition may also hold potential for mitigating thrombotic events. Therefore, future preclinical and clinical studies aimed at identifying and validating NLRP3 inhibition as a novel therapeutic intervention for thrombotic disorders are imperative.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Visakhapatnam 530045, India
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
4
|
Bucci T, Ames PRJ, Cammisotto V, Cardamone C, Ciampa A, Mangoni B, Triggiani M, Carnevale R, Lip GYH, Pastori D, Pignatelli P. Low-grade endotoxemia and risk of recurrent thrombosis in primary antiphospholipid syndrome. The multicenter ATHERO-APS study. Thromb Res 2023; 231:76-83. [PMID: 37827070 DOI: 10.1016/j.thromres.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Low-grade endotoxemia is associated with systemic inflammation, enhanced oxidative stress and cardiovascular events in different clinical settings, but its possible role as "second hit" in patients with primary antiphospholipid syndrome (PAPS) has never been investigated. PURPOSE To evaluate the relationship between plasma lipopolysaccharide (LPS) levels, oxidative stress markers and risk of thrombosis in the prospective multicenter ATHERO-APS study. METHODS Baseline LPS, soluble NADPH-oxidase 2-derived peptide (sNOX-dp), H2O2 production, hydrogen peroxide breakdown activity (HBA), and nitric oxide (NO) bioavailability were compared in 97 PAPS, 16 non-thrombotic aPL carriers and 21 controls (CTRL) matched for age and sex. Correlations among laboratory variables were explored by Rho Spearman's correlation (rS). Cox-regression analysis was performed to assess the association between LPS and risk for a composite outcome of cardiovascular death, venous and arterial thromboembolism. RESULTS In the whole cohort (median age 51 years (IQR 43-60), 72 % female), PAPS demonstrated higher levels of LPS, sNOX-dp and H2O2 and lower levels of NO and HBA compared to non-thrombotic aPL carriers and CTRL. LPS levels were inversely correlated with HBA (rS: -0.295, p = 0.001) and NO (rS: -0.322, p < 0.001) and directly correlated with sNOX-dp (rS:0.469, p < 0.001) and H202 (rS:0.282, p < 0.001). PAPS showed higher levels of LPS, sNOX-dp and H2O2 and lower levels of NO and HBA compared to aPL carriers and CTRL. After a 4.7 years follow-up of, 11 composite outcomes were reported in PAPS (2.5 per 100 patient-years) while none was observed in aPL carriers. On Cox-regression analysis, patients with LPS above the median (>23.1 pg/ml) had a 5-fold increased risk of composite outcome compared to those with LPS below the median, after adjustment for sex, age, diabetes, and global antiphospholipid syndrome score. CONCLUSION Low-grade endotoxemia is associated with an increased oxidative stress and a higher risk of thrombosis in PAPS. Its prognostic value in carriers needs to be investigated in larger cohorts.
Collapse
Affiliation(s)
- Tommaso Bucci
- Department of General and Specialized Surgery, Sapienza University of Rome, Italy; Liverpool Centre of Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.
| | - Paul R J Ames
- Immune Response and Vascular Disease Unit, CEDOC, Nova University Lisbon, Rua Camara Pestana, Lisbon, Portugal; Department of Haematology, Dumfries Royal Infirmary, Cargenbridge, Dumfries, United Kingdom
| | - Vittoria Cammisotto
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Chiara Cardamone
- Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| | | | - Bianca Mangoni
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy; IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, IS, Italy
| | - Gregory Y H Lip
- Liverpool Centre of Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Danish Center for Clinical Health Services Research, Department of Clinical Medicine, Aalborg University, Denmark
| | - Daniele Pastori
- Liverpool Centre of Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.
| | - Pasquale Pignatelli
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Abstract
Antiphospholipid syndrome (APS) is a thrombo-inflammatory disease propelled by circulating autoantibodies that recognize cell surface phospholipids and phospholipid binding proteins. The result is an increased risk of thrombotic events, pregnancy morbidity, and various other autoimmune and inflammatory complications. Although antiphospholipid syndrome was first recognized in patients with lupus, the stand alone presentation of antiphospholipid syndrome is at least equally common. Overall, the diagnosis appears to affect at least one in 2000 people. Studies of antiphospholipid syndrome pathogenesis have long focused on logical candidates such as coagulation factors, endothelial cells, and platelets. Recent work has shed light on additional potential therapeutic targets within the innate immune system, including the complement system and neutrophil extracellular traps. Vitamin K antagonists remain the mainstay of treatment for most patients with thrombotic antiphospholipid syndrome and, based on current data, appear superior to the more targeted direct oral anticoagulants. The potential role of immunomodulatory treatments in antiphospholipid syndrome management is receiving increased attention. As for many systemic autoimmune diseases, the most important future direction is to more precisely identify mechanistic drivers of disease heterogeneity in pursuit of unlocking personalized and proactive treatments for patients.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - D Ware Branch
- James R. and Jo Scott Research Chair, Department of Obstetrics and Gynecology, University of Utah Health and Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Thomas L Ortel
- Division of Hematology, Departments of Medicine and Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
6
|
Targeting thromboinflammation in antiphospholipid syndrome. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2022; 21:744-757. [PMID: 36696191 DOI: 10.1016/j.jtha.2022.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/26/2023]
Abstract
Antiphospholipid syndrome (APS) is a systemic autoimmune disease, where persistent presence of antiphospholipid antibodies (aPL) leads to thrombotic and obstetric complications. APS is a paradigmatic thromboinflammatory disease. Thromboinflammation is a pathophysiological mechanism coupling inflammation and thrombosis, which contributes to the pathophysiology of cardiovascular disease. APS can serve as a model to unravel mechanisms of thromboinflammation and the relationship between innate immune cells and thrombosis. Monocytes are activated by aPL into a proinflammatory and procoagulant phenotype, producing proinflammatory cytokines such as tumor necrosis factor α, interleukin 6, as well as tissue factor. Important cellular signaling pathways involved are the NF-κB-pathway, mammalian target of rapamycin (mTOR) signaling, and the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome. All of these may serve as future therapeutic targets. Neutrophils produce neutrophil extracellular traps in response to aPL, and this leads to thrombosis. Thrombosis in APS also stems from increased interaction of neutrophils with endothelial cells through P-selectin glycoprotein ligand-1. NETosis can be targeted not only with several experimental therapeutics, such as DNase, but also through the redirection of current therapies such as defibrotide and the antiplatelet agent dipyridamole. Activation of platelets by aPL leads to a procoagulant phenotype. Platelet-leukocyte interactions are increased, possibly mediated by increased levels of soluble P-selectin and soluble CD40-ligand. Platelet-directed future treatment options involve the inhibition of several platelet receptors activated by aPL, as well as mTOR inhibition. This review discusses mechanisms underlying thromboinflammation in APS that present targetable therapeutic options, some of which may be generalizable to other thromboinflammatory diseases.
Collapse
|
7
|
Antiphospholipid Antibody Syndrome-Associated Increased Surface Expression of VLA4 Integrin on Human Monocytes. Biomedicines 2022; 10:biomedicines10102341. [PMID: 36289603 PMCID: PMC9598317 DOI: 10.3390/biomedicines10102341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Antiphospholipid syndrome (APS) is a systemic autoimmune disorder characterized by thrombosis and/or obstetric complications in the presence of antiphospholipid antibodies (aPL). Catastrophic APS (CAPS) is the most severe form of the disease, in which microvascular thromboses develop rapidly, leading to multiorgan failure. Monocytes, along with endothelial cells, are critical players in the pathogenesis of APS. Recruitment of these cells to the site of injury/inflammation involves a series of events, including capture, rolling, adhesion enhancement, and transmigration, which are controlled by surface adhesion molecules. The aim of our study was to investigate the surface adhesion profile of monocytes from APS patients and monocytes stimulated in vitro with aPL from a CAPS patient. The surface expression of the adhesion molecules LFA1, L-selectin, MAC1, PSGL1, and VLA4 was analyzed by flow cytometry. To our knowledge, this preliminary study was the first to show that VLA4 was significantly increased on the surface of monocytes from APS patients. Moreover, in vitro stimulations mimicking CAPS showed an even greater increase in VLA4. Our data suggest that the surface adhesion profile on monocytes is altered in APS and CAPS and may be involved in the thrombotic pathophysiology of the disease by enhancing monocyte adhesion.
Collapse
|
8
|
Knight JS, Kanthi Y. Mechanisms of immunothrombosis and vasculopathy in antiphospholipid syndrome. Semin Immunopathol 2022; 44:347-362. [PMID: 35122116 PMCID: PMC8816310 DOI: 10.1007/s00281-022-00916-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia propelled by circulating antiphospholipid antibodies that herald vascular thrombosis and obstetrical complications. Antiphospholipid antibodies recognize phospholipids and phospholipid-binding proteins and are not only markers of disease but also key drivers of APS pathophysiology. Thrombotic events in APS can be attributed to various conspirators including activated endothelial cells, platelets, and myeloid-lineage cells, as well as derangements in coagulation and fibrinolytic systems. Furthermore, recent work has especially highlighted the role of neutrophil extracellular traps (NETs) and the complement system in APS thrombosis. Beyond acute thrombosis, patients with APS can also develop an occlusive vasculopathy, a long-term consequence of APS characterized by cell proliferation and infiltration that progressively expands the intima and leads to organ damage. This review will highlight known pathogenic factors in APS and will also briefly discuss similarities between APS and the thrombophilic coagulopathy of COVID-19.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Yogendra Kanthi
- Division of Intramural Research National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| |
Collapse
|
9
|
Inflammatory response in relation to COVID-19 and other prothrombotic phenotypes. REUMATOLOGÍA CLÍNICA (ENGLISH EDITION) 2022; 18:1-4. [PMID: 35090606 PMCID: PMC7561343 DOI: 10.1016/j.reumae.2020.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022]
Abstract
The haemostatic system acts in concert with inflammation, so that after inflammatory response various mediators activate the haemostatic system through endothelial dysfunction, platelet activation and coagulation promoting thrombosis, which is termed thromboinflammation. In this process, the inflammasome acquires special relevance; its stimulation promotes innate and adaptive immune responses. Inflammasome activation plays an important physiopathological role in several disorders with inflammatory and thrombotic phenomena. The role of thromboinflammation has become relevant in the COVID-19 pandemic, in which a cytokine storm has been described as one of the mechanisms responsible.
Collapse
|
10
|
Groarke EM, Dulau-Florea AE, Kanthi Y. Thrombotic manifestations of VEXAS syndrome. Semin Hematol 2021; 58:230-238. [PMID: 34802545 DOI: 10.1053/j.seminhematol.2021.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 11/11/2022]
Abstract
VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome is a recently described autoinflammatory syndrome characterized by diffuse inflammatory manifestations, predisposition to hematological malignancy, and an association with a high rate of thrombosis. VEXAS is attributed to somatic mutations in the UBA1 gene in hematopoietic stem and progenitor cells with myeloid restriction in mature forms. The rate of thrombosis in VEXAS patients is approximately 40% in all reported cases to date. Venous thromboembolism predominates thrombotic events in VEXAS. These are classified as unprovoked in etiology, although systemic and vascular inflammation are implicated. Here, we review the clinical and laboratory characteristics in VEXAS that provide insight into the possible mechanisms leading to thrombosis. We present knowledge gaps in the mechanisms and management of VEXAS-associated thromboinflammation and propose areas for future investigation in the field.
Collapse
Affiliation(s)
- Emma M Groarke
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Alina E Dulau-Florea
- Hematology Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Hollerbach A, Müller-Calleja N, Pedrosa D, Canisius A, Sprinzl MF, Falter T, Rossmann H, Bodenstein M, Werner C, Sagoschen I, Münzel T, Schreiner O, Sivanathan V, Reuter M, Niermann J, Galle PR, Teyton L, Ruf W, Lackner KJ. Pathogenic lipid-binding antiphospholipid antibodies are associated with severity of COVID-19. J Thromb Haemost 2021; 19:2335-2347. [PMID: 34242469 PMCID: PMC8420426 DOI: 10.1111/jth.15455] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Coronavirus disease 19 (COVID-19)-associated coagulopathy is a hallmark of disease severity and poor prognosis. The key manifestations of this prothrombotic syndrome-microvascular thrombosis, stroke, and venous and pulmonary clots-are also observed in severe and catastrophic antiphospholipid syndrome. Antiphospholipid antibodies (aPL) are detectable in COVID-19 patients, but their association with the clinical course of COVID-19 remains unproven. OBJECTIVES To analyze the presence and relevance of lipid-binding aPL in hospitalized COVID-19 patients. METHODS Two cohorts of 53 and 121 patients from a single center hospitalized for PCR-proven severe acute respiratory syndrome-coronavirus 2 infection were analyzed for the presence of aPL and clinical severity of COVID-19. RESULTS We here demonstrate that lipid-binding aPL are common in COVID-19. COVID-19 patients with lipid-binding aPL have higher median concentrations of C-reactive protein and D-dimer, and are more likely to have a critical clinical course and fatal outcome. Lipid-binding aPL isolated from COVID-19 patients target the recently described cell surface complex of lysobisphosphatidic acid (LBPA) with the protein C receptor (EPCR) to induce prothrombotic and inflammatory responses in monocytes and endothelial cells. We show that B1a cells producing lipid-reactive aPL of the IgG isotype circulate in the blood of COVID-19 patients. In vivo, COVID-19 aPL accelerate thrombus formation in an experimental mouse model dependent on the recently delineated signaling pathway involving EPCR-LBPA. CONCLUSIONS COVID-19 patients rapidly expand B1a cells secreting pathogenic lipid-binding aPL with broad thrombotic and inflammatory effects. The association with markers of inflammation and coagulation, clinical severity, and mortality suggests a causal role of aPL in COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Martin F Sprinzl
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Tanja Falter
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Heidi Rossmann
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Marc Bodenstein
- Department of Anesthesiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Christian Werner
- Department of Anesthesiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Ingo Sagoschen
- Department of Cardiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Oliver Schreiner
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Visvakanth Sivanathan
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Michael Reuter
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Johannes Niermann
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| |
Collapse
|
12
|
Goudsmit J, van den Biggelaar AHJ, Koudstaal W, Hofman A, Koff WC, Schenkelberg T, Alter G, Mina MJ, Wu JW. Immune age and biological age as determinants of vaccine responsiveness among elderly populations: the Human Immunomics Initiative research program. Eur J Epidemiol 2021; 36:753-762. [PMID: 34117979 PMCID: PMC8196271 DOI: 10.1007/s10654-021-00767-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/27/2021] [Indexed: 12/17/2022]
Abstract
The Human Immunomics Initiative (HII), a joint project between the Harvard T.H. Chan School of Public Health and the Human Vaccines Project (HVP), focuses on studying immunity and the predictability of immuneresponsiveness to vaccines in aging populations. This paper describes the hypotheses and methodological approaches of this new collaborative initiative. Central to our thinking is the idea that predictors of age-related non-communicable diseases are the same as predictors for infectious diseases like COVID-19 and influenza. Fundamental to our approach is to differentiate between chronological, biological and immune age, and to use existing large-scale population cohorts. The latter provide well-typed phenotypic data on individuals’ health status over time, readouts of routine clinical biochemical biomarkers to determine biological age, and bio-banked plasma samples to deep phenotype humoral immune responses as biomarkers of immune age. The first phase of the program involves 1. the exploration of biological age, humoral biomarkers of immune age, and genetics in a large multigenerational cohort, and 2. the subsequent development of models of immunity in relation to health status in a second, prospective cohort of an aging population. In the second phase, vaccine responses and efficacy of licensed COVID-19 vaccines in the presence and absence of influenza-, pneumococcal- and pertussis vaccines routinely offered to elderly, will be studied in older aged participants of prospective population-based cohorts in different geographical locations who will be selected for representing distinct biological and immune ages. The HII research program is aimed at relating vaccine responsiveness to biological and immune age, and identifying aging-related pathways crucial to enhance vaccine effectiveness in aging populations.
Collapse
Affiliation(s)
- Jaap Goudsmit
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Human Vaccines Project, New York, NY, USA
| | | | | | - Albert Hofman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wayne Chester Koff
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Human Vaccines Project, New York, NY, USA
| | - Theodore Schenkelberg
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Human Vaccines Project, New York, NY, USA
| | - Galit Alter
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Michael Joseph Mina
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Julia Wei Wu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
13
|
McCarty MF, Iloki Assanga SB, Lewis Luján L, O’Keefe JH, DiNicolantonio JJ. Nutraceutical Strategies for Suppressing NLRP3 Inflammasome Activation: Pertinence to the Management of COVID-19 and Beyond. Nutrients 2020; 13:E47. [PMID: 33375692 PMCID: PMC7823562 DOI: 10.3390/nu13010047] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 02/03/2023] Open
Abstract
Inflammasomes are intracellular protein complexes that form in response to a variety of stress signals and that serve to catalyze the proteolytic conversion of pro-interleukin-1β and pro-interleukin-18 to active interleukin-1β and interleukin-18, central mediators of the inflammatory response; inflammasomes can also promote a type of cell death known as pyroptosis. The NLRP3 inflammasome has received the most study and plays an important pathogenic role in a vast range of pathologies associated with inflammation-including atherosclerosis, myocardial infarction, the complications of diabetes, neurological and autoimmune disorders, dry macular degeneration, gout, and the cytokine storm phase of COVID-19. A consideration of the molecular biology underlying inflammasome priming and activation enables the prediction that a range of nutraceuticals may have clinical potential for suppressing inflammasome activity-antioxidants including phycocyanobilin, phase 2 inducers, melatonin, and N-acetylcysteine, the AMPK activator berberine, glucosamine, zinc, and various nutraceuticals that support generation of hydrogen sulfide. Complex nutraceuticals or functional foods featuring a number of these agents may find utility in the prevention and control of a wide range of medical disorders.
Collapse
Affiliation(s)
| | - Simon Bernard Iloki Assanga
- Department of Research and Postgraduate in Food, University of Sonora, Centro 83000, Mexico; (S.B.I.A.); (L.L.L.)
| | - Lidianys Lewis Luján
- Department of Research and Postgraduate in Food, University of Sonora, Centro 83000, Mexico; (S.B.I.A.); (L.L.L.)
| | | | | |
Collapse
|
14
|
Hollerbach A, Müller-Calleja N, Canisius A, Orning C, Lackner KJ. Induction of tissue factor expression by anti-β2-glycoprotein I is mediated by tumor necrosis factor α. J Thromb Thrombolysis 2020; 49:228-234. [PMID: 31612355 DOI: 10.1007/s11239-019-01970-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Antiphospholipid antibodies (aPL) are heterogeneous and there is evidence that binding specificity determines which cellular effects they can trigger. We have therefore hypothesised that the induction of tissue factor (TF) in monocytes and endothelial cells by aPL depends on their binding specificity. To further investigate this, we have analyzed the ability of three human monoclonal aPL with distinctly different binding specificities to induce transcription and cell surface expression of TF in monocytes and endothelial cells. Results with human monoclonal aPL were validated with IgG-fractions obtained from patients with APS. We confirmed previous results that a lipid reactive human monoclonal aPL rapidly induced TF transcription and cell surface expression in monocytes and endothelial cells. A monoclonal aPL reactive against β2 glycoprotein I (β2GPI) induced TF with a delayed time course. This was fully dependent on the induction of tumor necrosis factor alpha (TNFα) secretion as capture of TNFα by adalimumab prevented TF induction. This pattern was confirmed with patient IgG fractions. Both lipid reactive and anti-β2GPI induced TF transcription. Unexpectedly, this activity of anti-β2GPI was mediated fully by TNFα which was secreted in response to incubation with anti-β2GPI. The role of TNFα in mediating TF induction by anti-β2GPI may have wider implications for APS pathogenesis.
Collapse
Affiliation(s)
- Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Carolin Orning
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
15
|
Páramo JA. Inflammatory Response in Relation to COVID-19 and Other Prothrombotic Phenotypes. REUMATOLOGIA CLINICA 2020; 18:S1699-258X(20)30143-1. [PMID: 32600978 PMCID: PMC7298455 DOI: 10.1016/j.reuma.2020.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
The haemostatic system acts in concert with inflammation, so that after inflammatory response various mediators activate the haemostatic system through endothelial dysfunction, platelet activation and coagulation promoting thrombosis, which is termed thromboinflammation. In this process, the inflammasome acquires special relevance; its stimulation promotes innate and adaptive immune responses. Inflammasome activation plays an important physiopathological role in several disorders with inflammatory and thrombotic phenomena. The role of thromboinflammation has become relevant in the COVID-19 pandemic, in which a cytokine storm has been described as one of the responsible mechanisms.
Collapse
Affiliation(s)
- José A Páramo
- Servicio de Hematología, Clínica Universidad de Navarra. IdiSNA. CIBERCV, Pamplona, España.
| |
Collapse
|
16
|
Chanchal S, Mishra A, Singh MK, Ashraf MZ. Understanding Inflammatory Responses in the Manifestation of Prothrombotic Phenotypes. Front Cell Dev Biol 2020; 8:73. [PMID: 32117993 PMCID: PMC7033430 DOI: 10.3389/fcell.2020.00073] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/27/2020] [Indexed: 12/23/2022] Open
Abstract
Inflammasome complex is a multimeric protein comprising of upstream sensor protein of nucleotide-binding oligomerization domain (NOD)-like receptor family. It has an adaptor protein apoptosis-associated speck-like protein and downstream effector cysteine protease procaspase-1. Activation of inflammasome complex is body’s innate response to pathogen attack but its abnormal activation results in many inflammatory and cardiovascular disorders including thrombosis. It has displayed a prominent role in the clot formation advocating an interplay between inflammation and coagulation cascades. Therefore, elucidation of inflammasome and its molecular mechanisms in the manifestation of prothrombotic phenotypes becomes pertinent. Thrombosis is the formation and propagation of blood clot in the arterial or venous system due to several interactions of vascular and immune factors. It is a prevalent pathology underlying disorders like venous thromboembolism, stroke and acute coronary syndrome; thus, making thrombosis, a major contributor to the global disease burden. Recently studies have established a strong connection of inflammatory processes with this blood coagulation disorder. The hemostatic balance in thrombosis gets altered by the inflammatory mechanisms resulting in endothelial and platelet activation that subsequently increases secretion of several prothrombotic and antifibrinolytic factors. The upregulation of these factors is the critical event in the pathogenesis of thrombosis. Among various inflammasome, nucleotide-binding domain, leucine-rich-containing family, pyrin domain containing 3 (NLRP3) is one of the best-studied sterile inflammasome strengthening a link between inflammation and coagulation in thrombosis. NLRP3 activation results in the catalytic conversion of procaspase-1 to active caspase-1, which facilitate the maturation of interleukin-1β (IL-1β) and interleukin-18. These cytokines are responsible for immune cells activation critical for immune responses. These responses further results in endothelial and platelet activation and aggregation. However, the exact molecular mechanism related to the pathogenesis of thrombosis is still elusive. There have been several reports that demonstrate Tissue factor (TF)-mediated signaling in the production of pro-inflammatory cytokines enhancing inflammation by activating protease-activated receptors on various cells, which lead to additional cytokine expression. Therefore, it would be illuminating to interpret the inflammasomes regulation in coagulation and inflammation. This review, thus, tries to comprehensively compile emerging regulatory roles of the inflammasomes in thrombosis and discusses their molecular pathways in the manifestation of thrombotic phenotypes.
Collapse
Affiliation(s)
- Shankar Chanchal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Aastha Mishra
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Manvendra Kumar Singh
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Mohammad Zahid Ashraf
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
17
|
Serrano M, Morán L, Martinez-Flores JA, Mancebo E, Pleguezuelo D, Cabrera-Marante O, Delgado J, Serrano A. Immune Complexes of Beta-2-Glycoprotein I and IgA Antiphospholipid Antibodies Identify Patients With Elevated Risk of Thrombosis and Early Mortality After Heart Transplantation. Front Immunol 2019; 10:2891. [PMID: 31921152 PMCID: PMC6935976 DOI: 10.3389/fimmu.2019.02891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/25/2019] [Indexed: 12/28/2022] Open
Abstract
Background: The presence of anti-Beta 2 glycoprotein antibodies (aB2GP1) of IgA isotype is common in patients with functional impairment of the organs in which B2GP1 is elaborated. Pretransplant IgA aB2GP1 has been associated with increased risk of thrombosis in kidney and heart transplanted patients and has also been related with early mortality after heart transplantation. Circulating immune complexes between IgA and B2GP1 (B2A-CIC) have been described in the blood of patients positive for IgA aB2GP1 with thrombotic clinical symptoms. In kidney transplanted patients, B2A-CIC is a biomarker that predicts which patients IgA aB2GP1 positive are at risk of thrombosis events following kidney transplantation and may lead to early prophylactic treatment. The prevalence of B2A-CIC and its relation with outcomes after heart transplantation is not known. Methods: Follow-up study based on 151 consecutive patients who received a heart transplant. Autoantibodies and B2A-CIC were quantified in pre-transplant serum samples. Three groups of patients were followed-up for 2 years: Group-1, positive for IgA aB2GP1 and B2A-CIC (N = 19). Group-2, only positive for IgA aB2GP1 (N = 28). Group-0 (control group): IgA aB2GP1 negative (N = 104). Results: Kaplan-Meir survival analysis showed that mortality in B2A-CIC positive was higher than group-0 at 3 months (HR:5.08; 95%CI: 1.36–19.01) and at 2 years (HR:3.82; 95%CI: 1.54–12.66). No significant differences were observed between group-2 and group-0. Multivariate analysis identified B2A-CIC as the most important independent risk factor for early mortality (OR = 6.12; 95% CI: 1.93–19.4). Post-transplant incidence of thrombosis was significantly higher in B2A-CIC positive patients than in the control group (OR: 6.42; 95%CI: 2.1–19.63). Multivariate analysis identified the presence of B2A-CIC (OR: 6.13; 95%CI: 2.1–19.63) and the pre-transplant habit of smoking actively (OR: 4.18; 95%CI: 1.35–12.94) as independent risk factor for thrombosis. The proportion of patients who had thrombotic events or died in the first trimester was significantly higher in group-1 (73.7%) than in group-0 (16.3%; p < 0.001) and in group-2 (39.3%; p = 0.02). Multivariate analysis identified B2A-CIC as the main independent risk factor for early outcomes (mortality or thrombosis) in the first 3 months after heart transplant (OR = 11.42, 95% CI: 1.69–9.68). Conclusion: B2A-CIC are a predictor of early mortality and thrombosis after heart transplant.
Collapse
Affiliation(s)
- Manuel Serrano
- Immunology Department, Healthcare Research Institute, Hospital "12 de Octubre", Madrid, Spain.,Immunology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Laura Morán
- Cardiology Department, CIBERCV, Facultad de Medicina, Healthcare Research Institute, Hospital "12 de Octubre", Universidad Complutense de Madrid, Madrid, Spain
| | | | - Esther Mancebo
- Immunology Department, Healthcare Research Institute, Hospital "12 de Octubre", Madrid, Spain
| | - Daniel Pleguezuelo
- Immunology Department, Healthcare Research Institute, Hospital "12 de Octubre", Madrid, Spain
| | - Oscar Cabrera-Marante
- Immunology Department, Healthcare Research Institute, Hospital "12 de Octubre", Madrid, Spain
| | - Juan Delgado
- Cardiology Department, CIBERCV, Facultad de Medicina, Healthcare Research Institute, Hospital "12 de Octubre", Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio Serrano
- Immunology Department, Healthcare Research Institute, Hospital "12 de Octubre", Madrid, Spain.,Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
18
|
Wirestam L, Arve S, Linge P, Bengtsson AA. Neutrophils-Important Communicators in Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Front Immunol 2019; 10:2734. [PMID: 31824510 PMCID: PMC6882868 DOI: 10.3389/fimmu.2019.02734] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) are two autoimmune diseases that can occur together or separately. Insights into the pathogenesis have revealed similarities, such as development of autoantibodies targeting subcellular antigens as well as a shared increased risk of cardiovascular morbidity, potentially due to mutual pathologic mechanisms. In this review, we will address the evidence implicating neutrophils in the pathogenesis of these conditions, highlighting their shared features. The neutrophil is the most abundant leukocyte, recognized for its role in infectious and inflammatory diseases, but dysregulation of neutrophil effector functions, including phagocytosis, oxidative burst and formation of neutrophil extracellular traps (NETs) may also contribute to an autoimmune process. The phenotype of neutrophils in SLE and APS differs from neutrophils of healthy individuals, where neutrophils in SLE and APS are activated and prone to aggregate. A specific subset of low-density neutrophils with different function compared to normal-density neutrophils can also be found within the peripheral blood mononuclear cell (PBMC) fraction after density gradient centrifugation of whole blood. Neutrophil phagocytosis is required for regular clearance of cell remnants and nuclear material. Reactive oxygen species (ROS) released by neutrophils during oxidative burst are important for immune suppression and impairment of ROS production is seen in SLE. NETs mediate pathology in both SLE and APS via several mechanisms, including exposure of autoantigens, priming of T-cells and activation of autoreactive B-cells. NETs are also involved in cardiovascular events by forming a pro-thrombotic scaffolding surface. Lastly, neutrophils communicate with other cells by producing cytokines, such as Interferon (IFN) -α, and via direct cell-cell contact. Physiological neutrophil effector functions are necessary to prevent autoimmunity, but in SLE and APS these are altered.
Collapse
Affiliation(s)
- Lina Wirestam
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sabine Arve
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Petrus Linge
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anders A Bengtsson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Tissue factor pathway inhibitor primes monocytes for antiphospholipid antibody-induced thrombosis. Blood 2019; 134:1119-1131. [PMID: 31434703 DOI: 10.1182/blood.2019001530] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022] Open
Abstract
Antiphospholipid antibodies (aPLs) with complex lipid and/or protein reactivities cause complement-dependent thrombosis and pregnancy complications. Although cross-reactivities with coagulation regulatory proteins contribute to the risk for developing thrombosis in patients with antiphospholipid syndrome, the majority of pathogenic aPLs retain reactivity with membrane lipid components and rapidly induce reactive oxygen species-dependent proinflammatory signaling and tissue factor (TF) procoagulant activation. Here, we show that lipid-reactive aPLs activate a common species-conserved TF signaling pathway. aPLs dissociate an inhibited TF coagulation initiation complex on the cell surface of monocytes, thereby liberating factor Xa for thrombin generation and protease activated receptor 1/2 heterodimer signaling. In addition to proteolytic signaling, aPLs promote complement- and protein disulfide isomerase-dependent TF-integrin β1 trafficking that translocates aPLs and NADPH oxidase to the endosome. Cell surface TF pathway inhibitor (TFPI) synthesized by monocytes is required for TF inhibition, and disabling TFPI prevents aPL signaling, indicating a paradoxical prothrombotic role for TFPI. Myeloid cell-specific TFPI inactivation has no effect on models of arterial or venous thrombus development, but remarkably prevents experimental aPL-induced thrombosis in mice. Thus, the physiological control of TF primes monocytes for rapid aPL pathogenic signaling and thrombosis amplification in an unexpected crosstalk between complement activation and coagulation signaling.
Collapse
|
20
|
Pastori D, Bucci T, Triggiani M, Ames PRJ, Parrotto S, Violi F, Pignatelli P, Farcomeni A. Immunoglobulin G (IgG) anticardiolipin antibodies and recurrent cardiovascular events. A systematic review and Bayesian meta-regression analysis. Autoimmun Rev 2019; 18:519-525. [PMID: 30844560 DOI: 10.1016/j.autrev.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 12/13/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Anticardiolipin antibodies of the immunoglobulin G isotype (IgG aCL) have been suggested as risk factor for arterial and venous thrombosis. No conclusive data in patients with coronary artery disease (CAD) do exist. We investigate the risk of recurrent CAD according to the presence of IgG aCL. METHODS We performed a systematic review and meta-analysis to evaluate the risk of recurrent major adverse cardiac events (MACE) associated with the presence of IgG aCL in patients with CAD. MEDLINE and Cochrane databases were searched. We conducted a meta-analysis of the relative risk (RR) both at 12 and 24 months. RESULTS We included 11 eligible studies with a total of 2425 patients, 283 IgG aCL+ and 2142 IgG aCL-. The prevalence of IgG aCL+ ranged from 6.1% to 43.3%. A total of 341 cardiac events were reported: 71 (25.1%) in IgG aCL+ and 270 (12.6%) in IgG aCL- patients. We found an increased risk of recurrent MACE in patients with high IgG aCL both at 12 (RR 2.17, 2.5-97.5%CI, 1.54-3.00) and 24 months (RR 2.11, 2.5-97.5%CI, 1.62-2.66). This association was even stronger in patients with juvenile CAD (i.e. <50 years) at both 12 (RR 3.21, 2.5-97.5%CI, 1.74-5.41) and 24 months (RR 3.24, 2.5-97.5%CI, 1.84-5.21). CONCLUSION Patients with CAD and elevated IgG aCL have a doubled risk of recurrent MACE at 12 and 24 months. The presence of aCL should be suspected in patients with recurrent CAD events or in patients with juvenile CAD.
Collapse
Affiliation(s)
- Daniele Pastori
- I Clinica Medica, Atherothrombosis Centre, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy.
| | - Tommaso Bucci
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Salerno, Salerno, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Salerno, Salerno, Italy
| | - Paul R J Ames
- Immune Response and Vascular Disease Unit, Nova University, Lisbon, Portugal; Dumfries and Galloway Royal Infirmary, Dumfries, UK
| | - Sandro Parrotto
- I Clinica Medica, Atherothrombosis Centre, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Francesco Violi
- I Clinica Medica, Atherothrombosis Centre, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Pasquale Pignatelli
- I Clinica Medica, Atherothrombosis Centre, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Alessio Farcomeni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
21
|
Lackner KJ, Müller-Calleja N. Pathogenesis of antiphospholipid syndrome: recent insights and emerging concepts. Expert Rev Clin Immunol 2018; 15:199-209. [PMID: 30412684 DOI: 10.1080/1744666x.2019.1546578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Even though our understanding of the antiphospholipid syndrome (APS) has improved tremendously over the last decades, we are still not in a position to replace symptomatic anticoagulation by pathogenesis based causal treatments. Areas covered: Recent years have provided further insights into pathogenetically relevant mechanisms. These include a differentiation of pathogenic subtypes of antiphospholipid antibodies (aPL), novel mechanisms modulating disease activity, for example, extracellular vesicles and microRNA, and novel players in pathogenesis, for example, neutrophils and neutrophil extracellular traps (NETs). Expert commentary: It is evident that aPL induce a proinflammatory and procoagulant state and recent data suggest that different aPL species activate different signaling pathways which sometimes converge into a common cellular response. This implies that presence of more than one aPL species may disproportionally increase the risk for the major manifestations of APS, that is, thrombosis and fetal loss. Further delineation of the pathogenic mechanisms will hopefully provide clues to causal rather than symptomatic treatments of APS.
Collapse
Affiliation(s)
- Karl J Lackner
- a Institute of Clinical Chemistry and Laboratory Medicine , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,b Center for Translational Vascular Biology , University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| | - Nadine Müller-Calleja
- a Institute of Clinical Chemistry and Laboratory Medicine , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,b Center for Translational Vascular Biology , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,c Center for Thrombosis and Hemostasis , University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| |
Collapse
|
22
|
García-Carrasco M, Jiménez-Herrera EA, Gálvez-Romero JL, Mendoza-Pinto C, Méndez-Martínez S, Etchegaray-Morales I, Munguía-Realpozo P, Vázquez de Lara-Cisneros L, Santa Cruz FJ, Cervera R. The anti-thrombotic effects of vitamin D and their possible relationship with antiphospholipid syndrome. Lupus 2018; 27:2181-2189. [DOI: 10.1177/0961203318801520] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The importance of the immunomodulatory effects of vitamin D has recently been associated with autoimmune and chronic inflammatory diseases. Vitamin D deficiency has been linked to the development of autoimmune conditions. Antiphospholipid syndrome is an autoimmune disease characterized by thrombotic events and obstetric complications in patients with antiphospholipid antibodies. Current data show that patients with antiphospholipid syndrome have a high prevalence of vitamin D deficiency even without classic risk factors. Several studies have suggested vitamin D may have anti-thrombotic functions. In antiphospholipid syndrome, low vitamin D serum levels have been associated with thrombotic manifestations, suggesting a possible protective role of vitamin D in antiphospholipid syndrome. This literature review presents current evidence on the haemostatic functions of vitamin D and their possible relationship with the clinical manifestations of antiphospholipid syndrome.
Collapse
Affiliation(s)
- M García-Carrasco
- Systemic Autoimmune Diseases Research Unit, General Regional Hospital No. 36, Puebla, Mexico
- Department of Rheumatology, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Department of Immunology, Benemérita Universidad Autónomade Puebla, Puebla, Mexico
| | | | - J L Gálvez-Romero
- Department of Immunology, Benemérita Universidad Autónomade Puebla, Puebla, Mexico
- Department of Immunology and Allergology, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Puebla, Mexico
| | - C Mendoza-Pinto
- Systemic Autoimmune Diseases Research Unit, General Regional Hospital No. 36, Puebla, Mexico
- Department of Rheumatology, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Department of Immunology, Benemérita Universidad Autónomade Puebla, Puebla, Mexico
| | - S Méndez-Martínez
- Puebla Research Coordination, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - I Etchegaray-Morales
- Systemic Autoimmune Diseases Research Unit, General Regional Hospital No. 36, Puebla, Mexico
| | - P Munguía-Realpozo
- Department of Rheumatology, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | | | - F J Santa Cruz
- Dermatology Centre ‘Dr Ladislao de la Pascua’, México, Mexico
| | - R Cervera
- Department of Autoimmune Diseases, Institut Clínic de Medicina I Dermatologia, Barcelona, Spain
| |
Collapse
|
23
|
Lackner KJ, Müller-Calleja N. Laboratory criteria for antiphospholipid syndrome: comment. J Thromb Haemost 2018; 16:2115-2116. [PMID: 30007114 DOI: 10.1111/jth.14237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Indexed: 11/29/2022]
Affiliation(s)
- K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Mainz, Germany
| |
Collapse
|
24
|
Devreese KMJ, Ortel TL, Pengo V, de Laat B. Laboratory criteria for antiphospholipid syndrome: reply. J Thromb Haemost 2018; 16:2117-2119. [PMID: 30007111 DOI: 10.1111/jth.14238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- K M J Devreese
- Coagulation Laboratory, Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - T L Ortel
- Departments of Medicine and Pathology, Duke University Medical Center, Durham, NC, USA
| | - V Pengo
- Cardiology Department, University of Padova, Padova, Italy
| | - B de Laat
- Department of Biochemistry, Maastricht University, Synapse BV, Maastricht, the Netherlands
| |
Collapse
|
25
|
Manukyan D, Müller-Calleja N, Lackner K. Pathophysiological insights into the antiphospholipid syndrome. Hamostaseologie 2017; 37:202-207. [DOI: 10.5482/hamo-16-07-0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/06/2016] [Indexed: 11/05/2022] Open
Abstract
SummaryThe antiphospholipid syndrome (APS) is characterized by venous and/or arterial thrombosis and severe pregnancy morbidity in presence of antiphospholipid antibodies (aPL). While there is compelling evidence that aPL cause the clinical manifestations of APS, the underlying mechanisms are still a matter of scientific debate. This is mainly related to the broad heterogeneity of aPL. There are three major types of aPL: The first one binds to (anionic) phospholipids, e.g. cardiolipin, in absence of other factors (cofactor independent aPL). The second type binds to phospholipids only in presence of protein cofactors, e.g. ß2-glycoprotein I (ß2GPI) (cofactor dependent aPL). The third type binds to cofactor proteins directly without need for phospholipids. It is widely believed that cofactor independent aPL (type 1) are associated with infections and, more importantly, non-pathogenic, while pathogenic aPL belong to the second and in particular to the third type. This view, in particular with regard to type 1 aPL, has not been undisputed and novel research data have shown that it is in fact untenable. We summarize the available data on the pathogenetic role of aPL and the implications for diagnosis of APS and future research.
Collapse
|
26
|
Lv J, He X, Wang H, Wang Z, Kelly GT, Wang X, Chen Y, Wang T, Qian Z. TLR4-NOX2 axis regulates the phagocytosis and killing of Mycobacterium tuberculosis by macrophages. BMC Pulm Med 2017; 17:194. [PMID: 29233104 PMCID: PMC5727946 DOI: 10.1186/s12890-017-0517-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 11/21/2017] [Indexed: 11/23/2022] Open
Abstract
Background Macrophages stand at the forefront of both innate and adapted immunity through their capacities to recognize, engulf, and eliminate foreign particles, and to stimulate adapted immune cells. They are also involved in controlling pro- and anti-inflammatory pathways. Macrophage activity against Mycobacterium tuberculosis (M. tuberculosis) has been shown to involve Toll-like receptor (TLR) activation and ROS production. Previous studies have shown that lipopolysaccharide (LPS), through TLR4, could activate macrophages, improve their bactericidal ROS production, and facilitate anti-infective immune responses. We sought to better understand the role of the TLR4-NOX2 axis in macrophage activation during M. tuberculosis infection. Methods THP-1 macrophages and PMA primed THP-1 macrophages [THP-1(A)] were treated with LPS and infected by M. tuberculosis. Cells were analyzed by flow cytometry for TLR4 expression, ROS production, phagocytosis, and killing of M. tuberculosis. Western blotting was used to analyze NOX2 expression. Inhibitors of the TLR4-NOX2 pathway were used to assess this pathway’s role in these processes, and their role in LPS activation of macrophages. Results We found that THP1-derived macrophages or PMA primed THP-1 macrophages exhibit higher surface TLR4 levels and increased NOX2 expression levels following LPS treatment. M. tuberculosis infection reduced these levels, but LPS was able to limit the negative effects of M.tb. Additionally, LPS increases THP-1(A) cells’ bactericidal activities including phagocytosis, ROS production, and destruction of M. tuberculosis. Significantly, all of these activities are impaired when TLR4 or NOX2 are inhibited. Conclusion These studies demonstrate the importance of the TLR4-NOX2 axis in M. tuberculosis elimination by macrophages and may lead to novel therapies for tuberculosis and other bacterial infections. Electronic supplementary material The online version of this article (10.1186/s12890-017-0517-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingzhu Lv
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, 233003, China
| | - Xiaoyan He
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, 233003, China
| | - Hongtao Wang
- Key Laboratory of Anhui Province for Infection and Immunology, Bengbu Medical College, 2600 Donghai Ave, Bengbu, Anhui, 233003, China
| | - Zhaohua Wang
- Department of Pulmonary Medicine, Bengbu Infectious Disease Hospital, Bengbu, Anhui, 233003, China
| | - Gabriel T Kelly
- Department of Medicine, The University of Arizona, 1656 E. Mabel St, P.O. Box 245218, Tucson, AZ, 85724, USA
| | - Xiaojing Wang
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital; Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Yin Chen
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ, 85724, USA
| | - Ting Wang
- Department of Medicine, The University of Arizona, 1656 E. Mabel St, P.O. Box 245218, Tucson, AZ, 85724, USA.
| | - Zhongqing Qian
- Key Laboratory of Anhui Province for Infection and Immunology, Bengbu Medical College, 2600 Donghai Ave, Bengbu, Anhui, 233003, China.
| |
Collapse
|
27
|
Müller-Calleja N, Hollerbach A, Häuser F, Canisius A, Orning C, Lackner KJ. Antiphospholipid antibody-induced cellular responses depend on epitope specificity : implications for treatment of antiphospholipid syndrome. J Thromb Haemost 2017; 15:2367-2376. [PMID: 29024318 DOI: 10.1111/jth.13865] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Indexed: 01/18/2023]
Abstract
Essentials Antiphospholipid antibodies (aPL) are heterogeneous and induce different cellular responses. We analyzed signaling events induced by different monoclonal and patient aPL in monocytes. Two major signaling pathways involving either NADPH-oxidase or LRP8 were identified. Our data suggest that these two pathways mediate the majority of aPL effects on monocytes. SUMMARY Background Antiphospholipid antibodies (aPLs) contribute to the pathogenesis of the antiphospholipid syndrome (APS) by induction of an inflammatory and procoagulant state in different cell types, and several signaling pathways have been described. Objectives To investigate whether signaling depends on the epitope specificity of aPLs. Methods Cellular effects of three human monoclonal aPLs with distinctly different epitope specificities were analyzed in vitro. Expression of tumor necrosis factor-α mRNA by mouse and human monocytes was the major readout. Analysis included cells from genetically modified mice, and the use of specific inhibitors in human monocytes. Data were validated with IgG isolated from 20 APS patients. Results Cofactor-independent anticardiolipin aPLs activated monocytes by induction of endosomal NADPH oxidase. Activation could be blocked by hydroxychloroquine (HCQ). Anti-β2 -glycoprotein I aPL activated monocytes by interacting with LDL receptor-related protein 8 (LRP8). This could be blocked by rapamycin. Analysis of 20 APS patients' IgG showed that all IgG fractions activated the same two pathways as the monoclonal aPL, depending on their epitope patterns as determined by ELISA. Monocyte activation by APS IgG could be blocked completely by HCQ and/or rapamycin, suggesting that in most, if not all, APS patients there is no other relevant signaling pathway. Conclusions aPLs activate two major proinflammatory signal transduction pathways, depending on their epitope specificity. HCQ and rapamycin, either alone or in combination, completely suppress signaling by APS IgG. These observations may provide a rationale for specific treatment of APS patients according to their aPL profile.
Collapse
Affiliation(s)
- N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - A Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - F Häuser
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - A Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - C Orning
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW It is well established that the antiphospholipid syndrome (APS) is caused by antiphospholipid antibodies (aPL). While several underlying mechanisms have been described in the past, many open questions remain. Here, we will review data on endosomal signaling and, in particular, redox signaling in APS. RECENT FINDINGS Endosomal redox signaling has been implicated in several cellular processes including signaling of proinflammatory cytokines. We have shown that certain aPL can activate endosomal NADPH-oxidase (NOX) in several cell types followed by induction of proinflammatory and procoagulant cellular responses in vitro. Involvement of endosomes in aPL signaling has also been reported by others. In wild-type mice but not in NOX-deficient mice, aPL accelerate venous thrombus formation underscoring the relevance of endosomal NOX. Furthermore, hydroxychloroquine (HCQ) inhibits activation of endosomal NOX and prevents thrombus formation in aPL-treated mice. Endosomal redox signaling is an important novel mechanism involved in APS pathogenesis. This makes endosomes a potential target for future treatment approaches of APS.
Collapse
|
29
|
DiNicolantonio JJ, O'Keefe JH, McCarty MF. Supplemental N-acetylcysteine and other measures that boost intracellular glutathione can downregulate interleukin-1β signalling: a potential strategy for preventing cardiovascular events? Open Heart 2017; 4:e000599. [PMID: 28878946 PMCID: PMC5574421 DOI: 10.1136/openhrt-2017-000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/13/2017] [Indexed: 11/03/2022] Open
|
30
|
McCarty MF, O'Keefe JH, DiNicolantonio JJ. Interleukin-1beta may act on hepatocytes to boost plasma homocysteine - The increased cardiovascular risk associated with elevated homocysteine may be mediated by this cytokine. Med Hypotheses 2017; 102:78-81. [PMID: 28478836 DOI: 10.1016/j.mehy.2017.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/11/2017] [Indexed: 11/17/2022]
Abstract
The results of multi-center trials of B vitamin supplementation reveal that, whereas moderately elevated homocysteine predicts increased risk for coronary disease, it does not play a mediating role in this regard. This essay proposes that interleukin-1beta can act on hepatocytes to suppress expression of the hepatocyte-specific forms of methionine adenosyltransferase; this in turn can be expected to decrease hepatic activity of cystathionine-β-synthase, leading to an increase in plasma homocysteine. It is further proposed that interleukin-1beta (IL-1β) is a true mediating risk factor for cardiovascular disease, and that elevated homocysteine predicts coronary disease because it can serve as a marker for increased IL-1β activity. Potent statin therapy may decrease IL-1β production by suppressing inflammasome activation - thereby accounting for the marked protection from cardiovascular events observed in the classic JUPITER study, in which the enrolled subjects had low-normal Low Density Lipoprotein cholesterol but elevated C-reactive protein.
Collapse
|
31
|
Müller-Calleja N, Manukyan D, Canisius A, Strand D, Lackner KJ. Hydroxychloroquine inhibits proinflammatory signalling pathways by targeting endosomal NADPH oxidase. Ann Rheum Dis 2017; 76:891-897. [PMID: 27903507 DOI: 10.1136/annrheumdis-2016-210012] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/28/2016] [Accepted: 11/05/2016] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Hydroxychloroquine (HCQ) has been used for decades to treat patients with rheumatic diseases, for example, systemic lupus erythematosus (SLE), rheumatoid arthritis or the antiphospholipid syndrome (APS). We hypothesise that HCQ might target endosomal NADPH oxidase (NOX), which is involved in the signal transduction of cytokines as well as antiphospholipid antibodies (aPL). METHODS For in vitro experiments, monocytic cells were stimulated with tumour necrosis factor α (TNFα), interleukin-1β (IL-1β) or a human monoclonal aPL and the activity of NOX was determined by flow cytometry. The expression of genes known to be induced by these stimuli was quantified by quantitative reverse transcription PCR. Live cell imaging was performed by confocal laser scanning microscopy. Finally, the effects of HCQ on NOX-induced signal transduction were analysed in an in vivo model of venous thrombosis. RESULTS HCQ strongly reduces or completely prevents the induction of endosomal NOX by TNFα, IL-1β and aPL in human monocytes and MonoMac1 cells. As a consequence, induction of downstream genes by these stimuli is reduced or abrogated. This effect of HCQ is not mediated by direct interference with the agonists but by inhibiting the translocation of the catalytic subunit of NOX2 (gp91phox) into the endosome. In vivo, HCQ protects mice from aPL-induced and NOX2-mediated thrombus formation. CONCLUSIONS We describe here a novel mechanism of action of HCQ, that is, interference with the assembly of endosomal NOX2. Since endosomal NOX2 is involved in many inflammatory and prothrombotic signalling pathways, this activity of HCQ might explain many of its beneficial effects in rheumatic diseases including the APS.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Davit Manukyan
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Dennis Strand
- Department of Medicine 1, University Medical Center Mainz, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
32
|
Lackner KJ, Müller-Calleja N. Pathogenesis of the antiphospholipid syndrome revisited: time to challenge the dogma: reply. J Thromb Haemost 2016; 14:2563-2564. [PMID: 27696707 DOI: 10.1111/jth.13523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
33
|
Lu LL, Chung AW, Rosebrock T, Ghebremichael M, Yu WH, Grace PS, Schoen MK, Tafesse F, Martin C, Leung V, Mahan AE, Sips M, Kumar M, Tedesco J, Robinson H, Tkachenko E, Draghi M, Freedberg KJ, Streeck H, Suscovich TJ, Lauffenburger D, Restrepo BI, Day C, Fortune SM, Alter G. A Functional Role for Antibodies in Tuberculosis. Cell 2016; 167:433-443.e14. [PMID: 27667685 PMCID: PMC5526202 DOI: 10.1016/j.cell.2016.08.072] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/04/2016] [Accepted: 08/26/2016] [Indexed: 11/18/2022]
Abstract
While a third of the world carries the burden of tuberculosis, disease control has been hindered by a lack of tools, including a rapid, point-of-care diagnostic and a protective vaccine. In many infectious diseases, antibodies (Abs) are powerful biomarkers and important immune mediators. However, in Mycobacterium tuberculosis (Mtb) infection, a discriminatory or protective role for humoral immunity remains unclear. Using an unbiased antibody profiling approach, we show that individuals with latent tuberculosis infection (Ltb) and active tuberculosis disease (Atb) have distinct Mtb-specific humoral responses, such that Ltb infection is associated with unique Ab Fc functional profiles, selective binding to FcγRIII, and distinct Ab glycosylation patterns. Moreover, compared to Abs from Atb, Abs from Ltb drove enhanced phagolysosomal maturation, inflammasome activation, and, most importantly, macrophage killing of intracellular Mtb. Combined, these data point to a potential role for Fc-mediated Ab effector functions, tuned via differential glycosylation, in Mtb control.
Collapse
Affiliation(s)
- Lenette L. Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Amy W. Chung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Microbiology and Immunology, University of Melbourne, Doherty Institute for Infection and Immunity, Melbourne, 3000, Australia
| | - Tracy Rosebrock
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | | | - Wen Han Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | | | - Fikadu Tafesse
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Constance Martin
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Vivian Leung
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Alison E. Mahan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Magdalena Sips
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biomedical Molecular Biology, Ghent University, Ghent, 9000, Belgium
| | - Manu Kumar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | - Hannah Robinson
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Monia Draghi
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | | | | | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Blanca I. Restrepo
- School of Public Health, University of Texas Health Houston, Brownsville, TX, 78520, USA
| | - Cheryl Day
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30332, USA
- South African Tuberculosis Vaccine Initiative (SATVI) and School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory 7925 South Africa
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| |
Collapse
|
34
|
Antiphospholipid Antibodies: Their Origin and Development. Antibodies (Basel) 2016; 5:antib5020015. [PMID: 31557996 PMCID: PMC6698834 DOI: 10.3390/antib5020015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 11/25/2022] Open
Abstract
Antiphospholipid antibodies (aPL) are a hallmark of the antiphospholipid syndrome (APS), which is the most commonly acquired thrombophilia. To date there is consensus that aPL cause the clinical manifestations of this potentially devastating disorder. However, there is good evidence that not all aPL are pathogenic. For instance, aPL associated with syphilis show no association with the manifestations of APS. While there has been intensive research on the pathogenetic role of aPL, comparably little is known about the origin and development of aPL. This review will summarize the current knowledge and understanding of the origin and development of aPL derived from animal and human studies.
Collapse
|
35
|
Lackner KJ, Müller-Calleja N. Pathogenesis of the antiphospholipid syndrome revisited: time to challenge the dogma. J Thromb Haemost 2016; 14:1117-20. [PMID: 26998919 DOI: 10.1111/jth.13320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/10/2016] [Indexed: 01/27/2023]
Abstract
For more than a decade the antiphospholipid syndrome (APS) has been reported to be caused mainly by antiphospholipid antibodies (aPL), which are not directed against phospholipids but against a complex of phospholipids and phospholipid binding proteins, so called cofactors (e.g. β2-glycoprotein I [β2GPI]). In fact, many researchers propose that the only relevant antigens in the APS are the cofactors themselves, with β2GPI being the most important. Antibodies that bind to phospholipids in a cofactor-independent manner are considered insignificant for the pathogenesis of the APS. We review the evidence for this current pathophysiologic concept and argue that it has never been proven and is now clearly no longer tenable. First, there is undisputable evidence that cofactor-independent aPL are pathogenic and present in the blood of APS patients. Second, available epidemiologic and clinical studies do not support a dominant pathogenic role for anti-β2GPI.
Collapse
Affiliation(s)
- K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Mainz, Germany
| |
Collapse
|
36
|
Chen X, Pang S, Lin J, Xia J, Wang Y. Allicin prevents oxidized low-density lipoprotein-induced endothelial cell injury by inhibiting apoptosis and oxidative stress pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:133. [PMID: 27207282 PMCID: PMC4875621 DOI: 10.1186/s12906-016-1126-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/13/2016] [Indexed: 12/26/2022]
Abstract
Background Vascular endothelial apoptosis is significantly associated with atherosclerosis and cardiovascular diseases, for which oxidized low-density lipoprotein (ox-LDL) is a major risk factor. Allicin, the primary active ingredient of garlic, has been found to have cardiovascular protective effect by changing the fatty-acid composition, but its effect on ox-LDL-induced vascular endothelial injury remains unclear. We investigated the protective effect of allicin on cell viability, LDH release, apoptosis and apoptotic signaling in human umbilical vein endothelial cells (HUVECs). Methods In cultured HUVEC cell line, ox-LDL induced injury was investigated. The cell viability and injury were evaluated by using cell proliferation Assay kit and LDH release assay. The apoptosis was evaluated by the Annexin V-FITC kit. The activity of caspase-3 was assessed using a colorimetric caspase-3 assay kit. The ROS production was evaluated by fluorometric assay and NADPH oxidase activity was assessed with a GENMED kit. Results Exposure of HUVECs to ox-LDL (150 μg/ml) reduced cell viability, induced apoptosis and increased activity of caspase-3, NADPH oxidase, and reactive oxygen species (ROS) production. The pretreatment with allicin (30 and 100 μM) significantly rescued the cell viability, inhibited ox-LDL-induced apoptosis and activity of caspase-3, NADPH oxidase and ROS production in HUVECs, and the protective effect is concentration-dependent. The allicin (100 μM) alone did not show significant difference from control. Our study demonstrated that allicin protected HUVECs from ox-LDL-induced endothelial injury by reducing the apoptosis, mediated by inhibition of caspase-3 and NADPH oxidase related apoptotic signaling. Conclusions Allicin prevents ox-LDL-induced endothelial cell injury by inhibiting apoptosis and oxidative stress pathway.
Collapse
|
37
|
Manukyan D, Müller-Calleja N, Jäckel S, Luchmann K, Mönnikes R, Kiouptsi K, Reinhardt C, Jurk K, Walter U, Lackner KJ. Cofactor-independent human antiphospholipid antibodies induce venous thrombosis in mice. J Thromb Haemost 2016; 14:1011-20. [PMID: 26786324 DOI: 10.1111/jth.13263] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Essentials Cofactor-independent antiphospholipid antibodies (CI-aPL) are generally considered non-pathogenic. We analyzed the effects of human monoclonal CI-aPL in a mouse model of venous thrombosis. As shown in vitro, CI-aPL induce a procoagulant state in vivo by activation of endosomal NADPH-oxidase. Contrary to common belief, CI-aPL induce venous thrombosis in vivo. SUMMARY Background There is general consensus that the antiphospholipid syndrome (APS) is caused by antiphospholipid antibodies (aPL) with antibodies against β2-glycoprotein-I being the most relevant. aPL that bind phospholipids in the absence of protein cofactors are generally considered pathogenetically irrelevant. We showed that cofactor-independent human monoclonal aPL isolated from APS patients induce proinflammatory and procoagulant cellular responses by activating endosomal NADPH-oxidase 2 (NOX2). Similar aPL were detected in all IgG fractions from APS patients analyzed. Objectives We aimed to clarify if cofactor-independent aPL can be thrombogenic in vivo and, if so, whether these effects are mediated via activation of NOX2. Methods Two cofactor-independent human monoclonal aPL, HL5B and RR7F, were tested in a mouse model of venous thrombosis. Genetically modified mice and in vitro assays were used to delineate the mechanisms underlying thrombus induction. Results HL5B and RR7F dramatically accelerate thrombus formation in this mouse model. Thrombus formation depends on tissue factor activation. It cannot be induced in NOX2-deficient mice. Bone marrow chimeras of C57BL/6J mice reconstituted with NOX2-deficient bone marrow showed that leukocyte activation plays a major role in thrombus formation. Neither TLR4 signaling nor platelet activation by our aPL is required for venous thrombus formation. Conclusions Cofactor-independent aPL can induce thrombosis in vivo. This effect is mainly mediated by leukocyte activation, which depends on the previously described signal transduction via endosomal NOX2. Because most APS patients have been shown to harbor aPL with similar activity, our data are of general relevance for the APS.
Collapse
Affiliation(s)
- D Manukyan
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| | - S Jäckel
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K Luchmann
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - R Mönnikes
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| | - K Kiouptsi
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - C Reinhardt
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K Jurk
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - U Walter
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| |
Collapse
|
38
|
Müller-Calleja N, Rossmann H, Müller C, Wild P, Blankenberg S, Pfeiffer N, Binder H, Beutel ME, Manukyan D, Zeller T, Lackner KJ. Antiphospholipid antibodies in a large population-based cohort: genome-wide associations and effects on monocyte gene expression. Thromb Haemost 2016; 116:115-23. [PMID: 27098658 DOI: 10.1160/th15-12-0947] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/15/2016] [Indexed: 11/05/2022]
Abstract
The antiphospholipid syndrome (APS) is characterised by venous and/or arterial thrombosis and pregnancy morbidity in women combined with the persistent presence of antiphospholipid antibodies (aPL). We aimed to identify genetic factors associated with the presence of aPL in a population based cohort. Furthermore, we wanted to clarify if the presence of aPL affects gene expression in circulating monocytes. Titres of IgG and IgM against cardiolipin, β2glycoprotein 1 (anti-β2GPI), and IgG against domain 1 of β2GPI (anti-domain 1) were determined in approx. 5,000 individuals from the Gutenberg Health Study (GHS) a population based cohort of German descent. Genotyping was conducted on Affymetrix Genome-Wide Human SNP 6.0 arrays. Monocyte gene expression was determined in a subgroup of 1,279 individuals by using the Illumina HT-12 v3 BeadChip. Gene expression data were confirmed in vitro and ex vivo by qRT-PCR. Genome wide analysis revealed significant associations of anti-β2GPI IgG and APOH on chromosome 17, which had been previously identified by candidate gene approaches, and of anti-domain1 and MACROD2 on chromosome 20 which has been listed in a previous GWAS as a suggestive locus associated with the occurrence of anti-β2GPI antibodies. Expression analysis confirmed increased expression of TNFα in monocytes and identified and confirmed neuron navigator 3 (NAV3) as a novel gene induced by aPL. In conclusion, MACROD2 represents a novel genetic locus associated with aPL. Furthermore, we show that aPL induce the expression of NAV3 in monocytes and endothelial cells. This will stimulate further research into the role of these genes in the APS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karl J Lackner
- Dr. Karl J. Lackner, Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany, Tel.: +49 6131 177190, E-mail:
| |
Collapse
|
39
|
Khamashta M, Taraborelli M, Sciascia S, Tincani A. Antiphospholipid syndrome. Best Pract Res Clin Rheumatol 2016; 30:133-48. [DOI: 10.1016/j.berh.2016.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Lackner KJ. More about cellular signaling by antiphospholipid antibodies. J Thromb Haemost 2015; 13:1925-7. [PMID: 26302892 DOI: 10.1111/jth.13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 11/29/2022]
Affiliation(s)
- K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
41
|
van den Hoogen LL, van Roon JAG, Radstake TRDJ, Fritsch-Stork RDE, Derksen RHWM. Delineating the deranged immune system in the antiphospholipid syndrome. Autoimmun Rev 2015; 15:50-60. [PMID: 26318678 DOI: 10.1016/j.autrev.2015.08.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022]
Abstract
The antiphospholipid syndrome (APS) is a systemic autoimmune disease that is characterized serologically by the presence of antiphospholipid antibodies (aPL) and clinically by vascular thrombosis and obstetric complications. The protein β2 glycoprotein I (β2GPI) is identified as the most important autoantigen in this syndrome. Activation of endothelial cells, thrombocytes and placental tissue by anti-β2GPI antibodies relates to the clinical manifestations of APS. This review describes genetic and environmental factors in relation to APS and summarizes the current knowledge on abnormalities in components of both the innate and adaptive immune system in APS. The role of dendritic cells, T-cells, B-cells, monocytes, neutrophils and NK-cells as well as the complement system in APS are discussed. Several gaps in our knowledge on the pathophysiology of APS are identified and a plea is made for future extensive immune cell profiling by a systems medicine approach in order to better unravel the pathogenesis of APS, to gain more insight in the role of the immune system in APS as well as having the potential to reveal biomarkers or novel therapeutic targets.
Collapse
Affiliation(s)
- Lucas L van den Hoogen
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | - Joël A G van Roon
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Ruth D E Fritsch-Stork
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Ronald H W M Derksen
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
42
|
Lamb FS. Localising oxidant "stress" in the antiphospholipid syndrome. Thromb Haemost 2015; 113:915. [PMID: 25832773 DOI: 10.1160/th15-03-0218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Fred S Lamb
- Fred S. Lamb, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA, Tel.: +1 615 936 1302, E-mail:
| |
Collapse
|