1
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
2
|
Lopez-Pedrera C, Pérez-Sánchez C, Tektonidou MG. Towards precision medicine in antiphospholipid syndrome. THE LANCET. RHEUMATOLOGY 2025:S2665-9913(25)00094-3. [PMID: 40418947 DOI: 10.1016/s2665-9913(25)00094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 05/28/2025]
Abstract
Antiphospholipid syndrome is a rare systemic autoimmune disorder with complex pathophysiology and high heterogeneity in clinical presentation and treatment responses. The core idea of precision medicine is that the varying treatment responses among patients with the same clinical diagnosis are due to differences in their underlying pathogenetic mechanisms and genetic makeup. A better understanding of the pathophysiology and multiple clinical subtypes of antiphospholipid syndrome has led to better classification and subphenotyping of the syndrome. Advances in microarray analysis, cytometry, and omic technologies have helped to identify genes, epigenetic variations, and pathway-informed biomarkers and identified new factors in disease development. By stratifying patients with antiphospholipid syndrome based on clinical or laboratory phenotypes and cellular and molecular profiles in the blood and affected tissues, treatments can be more effectively tailored, improving efficacy and reducing toxicity. This Review explores the current evidence on clinical, genetic, and biomolecular stratification in antiphospholipid syndrome and how artificial intelligence algorithms from clinical and molecular profiles can guide precision medicine in antiphospholipid syndrome.
Collapse
Affiliation(s)
- Chary Lopez-Pedrera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Carlos Pérez-Sánchez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Maria G Tektonidou
- Rheumatology Unit, First Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
3
|
Heimerl S, Höring M, Burkhardt R, Höpting M, Sigruener A, Liebisch G, Hart C. Platelet lipidomics indicates enhanced thrombocyte activation in patients with antiphospholipid syndrome in vivo. J Thromb Haemost 2025; 23:1689-1697. [PMID: 39952362 DOI: 10.1016/j.jtha.2025.01.020] [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: 09/20/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by the presence of antiphospholipid antibodies in patients with thromboembolic/thromboinflammatory events and/or obstetric complications. OBJECTIVES The aim of this study was to examine whether there are alterations in the platelet lipidome of APS patients in comparison with patients affected by thromboembolism without APS (control) and healthy volunteers. METHODS We applied quantitative mass spectrometry-based lipidomics to investigate the platelet lipidome of isolated resting and thrombin-stimulated platelets as well as platelet release in patients with APS, controls, and healthy volunteers. RESULTS Lipidomic data revealed an increase in lysophospholipids (LPLs) in platelets from APS patients, specifically in lysophosphatidylcholine and lysophosphatidylethanolamine species. As LPLs are cleavage products generated by phospholipase A (PLA) from the corresponding phospholipid precursor, LPL/phospholipid ratios may be employed as surrogates for PLA1 and PLA2 activities. The surrogate ratios for PLA2, which participates in the release of arachidonic acid during platelet activation, were significantly increased in APS in both resting platelets and upon thrombin-induced activation for phosphatidylcholine and phosphatidylethanolamine. The phosphatidylcholine-PLA2 surrogate ratio was found to correlate with serum levels of anti-β2-glycoprotein I and anticardiolipin immunoglobulin G. Finally, receiver operator characteristic analysis demonstrated excellent discrimination of patients with APS from controls and healthy volunteers. CONCLUSION These findings provide substantial evidence that platelet activation is enhanced in APS in vivo, involving the activation of PLA2.
Collapse
Affiliation(s)
- Susanne Heimerl
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Höpting
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Sigruener
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
| | - Christina Hart
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Hsieh MS, Liu HW, Guo FY, Song DP, Li MY, Chao TY, Fong IH, Chang YS, Yeh CT. S-hydroxychloroquine prevents the antiphospholipid thrombogenic complexes for antiphospholipid syndrome treatment. Biomed Pharmacother 2025; 186:117968. [PMID: 40120554 DOI: 10.1016/j.biopha.2025.117968] [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: 11/02/2024] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
Clinically used in systemic lupus erythematosus (SLE), Hydroxychloroquine (HCQ) exerts antithrombotic effects by inhibiting anti-β2-glycoprotein I (anti-β2GPI) antibody binding to phospholipid bilayers. However, HCQ is a racemic mixture, with only one enantiomer offering therapeutic benefits, while the other may contribute to toxicity. The current study evaluated the thromboprophylactic efficacy of R-enantiomer Hydroxychloroquine (R-HCQ), S-enantiomer Hydroxychloroquine (S-HCQ), and racemic HCQ (Rac-HCQ), with a focus on their impact on APS-associated markers. Both in vitro and in vivo models were employed, with human umbilical vein endothelial cells (HUVECs) and mice immunized with human β2-glycoprotein I antibodies used to evaluate the formation of antiphospholipid thrombotic complexes and their modulation by HCQ enantiomers. S-HCQ significantly reduced β2GPI complex binding and restored the AnxA5 anticoagulant shield in vitro, demonstrating superior efficacy over R-HCQ in disrupting β2GPI/anti-β2GPI interactions and preventing endothelial dysfunction in APS models. Pretreatment of HUVECs with S-HCQ significantly attenuated the expression of proinflammatory cytokines (tumor necrosis factor-alpha, interleukin-6, interleukin-1 beta, and C-C motif ligand 2) and endothelial activation markers (intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and E-selectin). S-HCQ alleviates endothelial dysfunction by reducing proinflammatory cytokines, endothelial activation markers, and NO production while downregulating iNOS expression, highlighting its potential to mitigate oxidative stress and thrombogenic activity in APS-related endothelial damage. In vivo, S-HCQ effectively reduced clot formation in the femoral veins of APS mouse models. Among the HCQ enantiomers tested, S-HCQ demonstrated superior efficacy in modulating inflammatory and angiogenic pathways, influencing the formation of antiphospholipid thrombotic complexes and mitigating thrombosis. These findings underscore the potential of S-HCQ as a therapeutic alternative for APS management.
Collapse
Affiliation(s)
- Ming-Shou Hsieh
- Department of Medical Research & Education, Taipei Medical University- Shuang Ho Hospital, New Taipei 23561, Taiwan
| | - Heng-Wei Liu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei 23561, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Fu-You Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Deng-Pan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng-Yuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tsu-Yi Chao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Iat-Hang Fong
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei 23561, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Sheng Chang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University- Shuang Ho Hospital, New Taipei 23561, Taiwan; Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan.
| |
Collapse
|
5
|
Tang W, Zhu L, Shi L, Gu B. P-Selectin, Vascular Endothelial Cadherin, and Vascular Cell Adhesion Molecule-1 as Novel Biomarkers for ABO Hemolytic Disease of the Fetus and Newborn. Anal Cell Pathol (Amst) 2025; 2025:9411137. [PMID: 40331036 PMCID: PMC12055318 DOI: 10.1155/ancp/9411137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/22/2025] [Indexed: 05/08/2025] Open
Abstract
Objective: This study aims to assess the potential of vascular endothelial injury markers, namely, P-selectin (PS), vascular endothelial cadherin (VE-Cad), and vascular cell adhesion molecule-1 (VCAM-1), as diagnostic and prognostic biomarkers for ABO hemolytic disease of the fetus and newborn (HDFN). Methods: A total of 218 pregnant women with ABO blood group incompatibility were recruited from the Third People's Hospital of Bengbu Affiliated to Bengbu Medical University. The serum levels of PS, VCAM-1, and VE-Cad were measured, and the participants were followed up until postpartum. The women were divided into an HDFN group and a control group based on the occurrence of ABO-HDFN. The correlations between the three vascular endothelial injury markers, pregnant anti-A/B antibody titers, and the occurrence and severity of HDFN were analyzed. Results: Compared to the control group, the levels of PS, VCAM-1, and VE-Cad were significantly elevated in the HDFN group. Additionally, these markers increased with higher IgG anti-A/B titers. For diagnosing HDFN, the area under the curve (AUC) for PS, VCAM-1, and VE-Cad were 0.826, 0.765, and 0.799, respectively. Moreover, the combined AUC of the three markers with IgG anti-A/B titers was 0.9. The levels of the three biomarkers were significantly negatively correlated with neonatal hemoglobin (Hb) and significantly positively correlated with reticulocyte percentage (Ret%), indirect bilirubin (IBIL), and lactate dehydrogenase (LDH). Univariate logistic regression indicated that increased levels of PS, VCAM-1, and VE-Cad were associated with a higher probability of ABO-HDFN. Multivariate logistic regression revealed that PS is an independent positive factor for HDFN. Conclusion: PS, VCAM-1, and VE-Cad provide experimental evidence for prenatal screening, diagnosis, early prevention and treatment of ABO-HDFN.
Collapse
Affiliation(s)
- Weichun Tang
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Linlin Zhu
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Liwei Shi
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Biao Gu
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| |
Collapse
|
6
|
Wang C, Li A, Zhang C, Yang Z, Yang X, Qi W, Liu T, Zhang M, Wang X. Neutrophil extracellular traps aggravate placental injury in OAPS by facilitating activation of BNIP3 mediated mitophagy. Free Radic Biol Med 2025; 235:109-123. [PMID: 40286883 DOI: 10.1016/j.freeradbiomed.2025.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Obstetric antiphospholipid syndrome (OAPS) is an autoimmune disease mediated by antiphospholipid antibodies (aPL), characterized by miscarriage, pre-mature birth caused by pre-eclampsia, intrauterine growth restriction, and other pregnancy complications. Neutrophil extracellular traps (NETs) consist of chromatin scaffolds coated with histones, proteases, granules, and cytosolic proteins. Here, we have observed increased levels of NETs in both human OAPS placental tissues and animal models, accompanied by apoptosis increases. In vitro, NETs induce a decrease in trophoblasts proliferation and an increase in apoptosis. In addition, NETs induce oxidative stress and mitochondrial reactive oxygen species (mtROS) production, upregulation of fission-associated mitochondrial proteins coupled with reduced fusion-associated protein levels. It also led to a rise in BNIP3 and autophagy-related protein expressions, along with an increase in autophagosomal numbers. Moreover, mtROS scavenging or knockdown strategies targeting BNIP3 effectively attenuated mitophagic activation alongside trophoblast apoptosis induction. Furthermore, upregulation of BNIP3 expression was evident in placentas from both APS murine models and human OAPS cases. These results suggest that aPL-induced NETs trigger BNIP3-mediated mitophagy, elevate ROS production, induce apoptosis in trophoblasts, and thereby exacerbate placental damage, with implications for our understanding of the pathogenesis of OAPS and devising novel treatment strategies.
Collapse
Affiliation(s)
- Chunying Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Anna Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Cancan Zhang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Zexin Yang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Xuan Yang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Weiyi Qi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Tengwei Liu
- Department of Obstetrics, Shandong Second Provincial General Hospital, Jinan, 250000, China
| | - Meihua Zhang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| | - Xietong Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| |
Collapse
|
7
|
Cai B, Zhou Y, Yang X, Wang Z, Huang C, Xiao Q, Jiang H, Zhao Y, Tian X, Wang Q, Li G, Li M, Zeng X, Zhao J. Remnant cholesterol predicts risk of recurrent thrombosis beyond LDL-cholesterol in patients with antiphospholipid syndrome. BMC Med 2025; 23:233. [PMID: 40264203 PMCID: PMC12016284 DOI: 10.1186/s12916-025-04063-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/10/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Antiphospholipid syndrome (APS) is notably linked to thrombotic events, particularly cardiovascular disease (CVD). The role of remnant cholesterol (RC) in predicting CVD risk is established, yet its relationship with thrombotic risk in APS patients remains to be elucidated. This study aims to assess the association between RC and recurrent thrombotic risk in patients with APS. METHODS A prospective analysis was conducted based on a cohort of APS patients who met the 2006 Sydney revised classification criteria. Thrombotic risks associated with varying levels of RC were evaluated using Kaplan-Meier survival analysis and Cox proportional hazards regression models. Mendelian randomization (MR) was applied to examine the causal link between RC and different types of thrombotic events. RESULTS A total of 325 patients with APS were enrolled in this study. Over a median follow-up of 35 months, 51 patients experienced thrombotic events, including 24 venous, 19 arterial, and 16 microvascular incidents. Patients with RC levels above 0.60 mmol/L exhibited significantly higher risks, with multivariable-adjusted hazard ratio (and 95% confidence interval) for all-cause, venous, arterial thrombosis, and microvascular disease being 5.05 (2.23-11.41), 6.34 (1.71-23.54), 3.79 (1.00-14.32), and 4.36 (1.08-17.58), respectively. Notably, elevated RC remained a significant thrombotic risk factor even in patients with normal conventional lipid profiles. MR analysis revealed a significant causal association between RC and arterial thrombosis, but not venous thrombosis. CONCLUSIONS Elevated RC is linked to a substantial increase in the risk of thrombotic events in APS patients. These findings suggest that RC could be a valuable marker for thrombotic risk in this population and a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Bin Cai
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Yangzhong Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Xinzhuang Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- Center for Bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoqing Wang
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Qingqing Xiao
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Yuan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Guanqiao Li
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
8
|
Yang L, Guo R, Liu H, Chen B, Li C, Liu R, Liao S, Xie Q, Yin G. Mechanism of antiphospholipid antibody-mediated thrombosis in antiphospholipid syndrome. Front Immunol 2025; 16:1527554. [PMID: 40181965 PMCID: PMC11966034 DOI: 10.3389/fimmu.2025.1527554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by the occurrence of thrombotic or obstetrical events in patients with persistent antiphospholipid antibodies (aPL). Thrombotic events, the primary pathological hallmarks and clinical manifestations, are among the leading causes of mortality in APS. Our understanding of the mechanism underlying APS-related thrombosis has significantly advanced in recent years. The presence of aPL, particularly anti-β2-glycoprotein I (anti-β2GPI) antibodies, is a major driver of thrombosis. The proposed pathophysiological mechanisms of aPL-mediated pro-thrombotic events can be broadly categorized into three types: disruption of anticoagulant reactions and fibrinolysis, interference with coagulation cascade cells, and complement activation. A triggering 'second hit' is typically necessary to initiate thrombosis. The development of animal models of APS has further refined our understanding of the role of aPL in thrombosis. In this review, we focused on the role of β2GPI-dependent aPL in thrombosis of thrombotic APS.
Collapse
Affiliation(s)
- Leiyi Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruibing Guo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Changpei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiting Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyi Liao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Liu L, Li M, Zhang C, Zhong Y, Liao B, Feng J, Deng L. Macrophage metabolic reprogramming: A trigger for cardiac damage in autoimmune diseases. Autoimmun Rev 2025; 24:103733. [PMID: 39716498 DOI: 10.1016/j.autrev.2024.103733] [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: 10/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Macrophage metabolic reprogramming has a central role in the progression of autoimmune and auto-inflammatory diseases. The heart is a major target organ in many autoimmune conditions and can sustain functional and structural impairments, potentially leading to irreversible cardiac damage. There is mounting clinical evidence pointing to a link between autoimmune disease and cardiac damage. However, this association remains poorly understood, and numerous patients do not receive appropriate preventive measures, which poses serious cardiovascular risks and significantly impacts their quality of life. This review discusses the relationship between macrophage metabolic reprogramming and cardiac damage in patients with autoimmune diseases and the role of adaptive immunity in macrophage reprogramming. It also provides an overview of the immunosuppressive therapies used at present. Exploiting the properties of macrophage reprogramming could lead to development of novel treatments for patients with autoimmune-related cardiac damage.
Collapse
Affiliation(s)
- Lin Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Minghao Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China.
| | - Li Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China; Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
10
|
Arachchillage DJ, Laffan M. Unresolved issues in the diagnosis and management of thrombotic antiphospholipid syndrome. Res Pract Thromb Haemost 2025; 9:102724. [PMID: 40236287 PMCID: PMC11999336 DOI: 10.1016/j.rpth.2025.102724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 04/17/2025] Open
Abstract
Antiphospholipid syndrome (APS) is a highly prothrombotic autoimmune disease characterized by the persistent presence of antiphospholipid autoantibodies (aPL) in association with thrombotic or nonthrombotic macro- and microvascular manifestations and/or pregnancy complications. This review is restricted to thrombotic APS. Since the publication of the American College of Rheumatology/European Alliance of Associations for Rheumatology classification criteria for APS, several authors have emphasized the difference between "classification" and "diagnosis" as a potential pitfall for clinicians. In addition to challenges associated with the diagnosis of APS, there are many unresolved areas in understanding pathogenesis and in the management of both thrombotic and obstetric APS. Although APS is an antibody-mediated autoimmune disease, secondary thrombosis prevention is achieved by anticoagulation, mainly with vitamin K antagonists, such as warfarin, rather than immunomodulation. Evidence is convincing for the use of vitamin K antagonists in triple-positive APS with venous thromboembolism. However, the best anticoagulant approach in the management of venous thromboembolism patients with single or dual positive aPL is not clear. Management of patients with stroke or arterial thrombosis with aPL remains a major unresolved issue, although some guidelines recommend the use of warfarin rather than antiplatelet therapy as the first-line treatment of stroke in APS. Recurrent thrombosis, despite therapeutic anticoagulation, remains a frequent problem and may be explained by the contribution of thrombo-inflammation in patients with thrombotic APS. In this narrative review, we discuss some of the unresolved issues in the diagnosis and management of thrombotic APS.
Collapse
Affiliation(s)
- Deepa J. Arachchillage
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Mike Laffan
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
11
|
Lami V, Nieri D, Pagnini M, Gattini M, Donati C, De Santis M, Cipriano A, Bazzan E, Sbrana A, Celi A, Neri T. Circulating, Extracellular Vesicle-Associated Tissue Factor in Cancer Patients with and without Venous Thromboembolism. Biomolecules 2025; 15:83. [PMID: 39858477 PMCID: PMC11762650 DOI: 10.3390/biom15010083] [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: 11/20/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer is characterized by chronic inflammation and hypercoagulability, with an excess of venous thromboembolism (VTE). Tissue factor, the initiator of blood coagulation, circulates associated with extracellular vesicles (EV-TF). Studies investigating EV-TF between cancer-associated and non-cancer-associated VTE are lacking. We therefore compared EV-TF in unprovoked VTE (U-VTE), cancer-associated VTE (C-VTE), and cancer without VTE (C-w/o VTE). We also investigated interleukin-6 (IL-6) levels between the same groups. The final population included 68 patients (U-VTE: n = 15; C-VTE: n = 24; C-w/o VTE: n = 29). All patients with VTE were enrolled within 48 h of diagnosis; non-VTE patients were recruited in the oncologic outpatient services. EV were isolated by differential centrifugation from 4 mL of peripheral blood; the final EV pellet (16,000× g for 45 min) was resuspended in 100 μL saline and tested for TF using a one-step clotting assay. There was a statistically significant difference for higher EV-TF in C-VTE and C-w/o VTE compared to U-VTE (p = 0.024; Kruskal-Wallis test). There was no significant difference between C-VTE and C-w/o VTE. Moreover, we did not find any difference in IL-6 levels. These preliminary data suggest that cancer represents, per se, a strong driver of EV-TF generation.
Collapse
Affiliation(s)
- Valentina Lami
- UO Medicina d’Urgenza e Pronto Soccorso, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (V.L.)
| | - Dario Nieri
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (D.N.); (A.C.)
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
| | - Marta Pagnini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, University of Pisa, 56126 Pisa, Italy
| | - Mario Gattini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
| | - Claudia Donati
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
| | - Mariella De Santis
- Dipartimento Cardio Toraco Vascolare, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy;
| | - Alessandro Cipriano
- UO Medicina d’Urgenza e Pronto Soccorso, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (V.L.)
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy;
| | - Andrea Sbrana
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy;
| | - Alessandro Celi
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (D.N.); (A.C.)
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, University of Pisa, 56126 Pisa, Italy
| | - Tommaso Neri
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, University of Pisa, 56126 Pisa, Italy; (M.P.); (M.G.); (C.D.)
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
12
|
Bardan CR, Ioniță I, Iordache M, Călămar-Popovici D, Todorescu V, Popescu R, Bernad BC, Bardan R, Bernad ES. Epigenetic Biomarkers in Thrombophilia-Related Pregnancy Complications: Mechanisms, Diagnostic Potential, and Therapeutic Implications: A Narrative Review. Int J Mol Sci 2024; 25:13634. [PMID: 39769397 PMCID: PMC11728153 DOI: 10.3390/ijms252413634] [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: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/14/2025] Open
Abstract
Pregnancy complications associated with thrombophilia represent significant risks for maternal and fetal health, leading to adverse outcomes such as pre-eclampsia, recurrent pregnancy loss, and intra-uterine growth restriction (IUGR). They are caused by disruptions in key physiological processes, including the coagulation cascade, trophoblast invasion, angiogenesis, and immune control. Recent advancements in epigenetics have revealed that non-coding RNAs, especially microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and extracellular vesicles (EVs) carrying these RNAs, play crucial roles in the regulation of these biological processes. This review aims to identify the epigenetic biomarkers that are the best candidates for evaluating thrombophilia-related pregnancy complications and for assessing the efficacy of anticoagulant and antiaggregant therapies. We emphasize their potential integration into personalized treatment plans, aiming to improve the risk assessment and therapy strategies for thrombophilic pregnancies. Future research should focus on validating these epigenetic biomarkers and establishing standardized protocols to enable their integration into clinical practice, paving the way for a precision medicine approach in obstetric care.
Collapse
Affiliation(s)
- Claudia Ramona Bardan
- Doctoral School, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.R.B.); (B.C.B.)
- Clinic of Hematology, Municipal Clinical Emergency Hospital, 300254 Timisoara, Romania; (I.I.); (M.I.); (D.C.-P.); (V.T.)
| | - Ioana Ioniță
- Clinic of Hematology, Municipal Clinical Emergency Hospital, 300254 Timisoara, Romania; (I.I.); (M.I.); (D.C.-P.); (V.T.)
- Department of Hematology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Maria Iordache
- Clinic of Hematology, Municipal Clinical Emergency Hospital, 300254 Timisoara, Romania; (I.I.); (M.I.); (D.C.-P.); (V.T.)
- Department of Hematology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Despina Călămar-Popovici
- Clinic of Hematology, Municipal Clinical Emergency Hospital, 300254 Timisoara, Romania; (I.I.); (M.I.); (D.C.-P.); (V.T.)
- Department of Hematology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Violeta Todorescu
- Clinic of Hematology, Municipal Clinical Emergency Hospital, 300254 Timisoara, Romania; (I.I.); (M.I.); (D.C.-P.); (V.T.)
- Department of Hematology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Roxana Popescu
- Division of Cell and Molecular Biology, Department of Microscopic Morphology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Brenda Cristiana Bernad
- Doctoral School, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.R.B.); (B.C.B.)
- Center for Neuropsychology and Behavioral Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Răzvan Bardan
- Department of Urology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Clinic of Urology, “Pius Brînzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Elena Silvia Bernad
- Department of Obstetrics and Gynecology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinic of Obstetrics and Gynecology, “Pius Brînzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
- Center for Laparoscopy, Laparoscopic Surgery and In Vitro Fertilization, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
13
|
Song X, Chen X, Wang D, Bai J. 5-oxoETE promote thrombosis in antiphospholipid syndrome by triggering NETs formation through PLC/PKC/ERK pathway. Inflamm Res 2024; 73:2165-2177. [PMID: 39377801 DOI: 10.1007/s00011-024-01956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND One mechanism by which antiphospholipid syndrome (APS) IgG contribute to thrombotic events in patients with APS is through the potentiation of neutrophil extracellular traps (NETs) release. However, the exact mechanism by which APS IgG induces NETs formation and thrombosis has not been fully elucidated. METHODS We conducted untargeted metabolomics on serum samples from thrombotic APS patients to identify metabolic changes. The effect of 5-oxoETE on NETs formation and oxidative stress was evaluated in vitro by treating neutrophils with various concentrations of 5-oxoETE. The involvement of the PLC/PKC/ERK signaling pathway in 5-oxoETE-induced NETs formation was examined using pharmacological inhibitors. In vivo, we assessed the effects of inhibiting 5-oxoETE synthesis or blocking its receptor (OXE-R) on NETs formation and thrombosis in APS mouse models. RESULTS Serum metabolomics revealed significantly elevated levels of 5-oxoETE in APS patients. In vitro experiments demonstrated that 5-oxoETE, via OXE-R activation of the PLC/PKC/ERK signaling pathway, increased NETs formation and oxidative stress in a dose-dependent manner. In vivo, inhibiting 5-oxoETE synthesis or OXE-R reduced NETs formation and attenuated venous thrombosis in APS mice models. CONCLUSION This study identifies 5-oxoETE as a critical mediator of NET formation and thrombosis in APS. Targeting 5-oxoETE or OXE-R may offer a promising therapeutic approach for thrombotic APS and other NET-associated autoimmune diseases.
Collapse
Affiliation(s)
- Xiaodong Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Xufeng Chen
- Department of Neurology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Dong Wang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Jie Bai
- Department of Infectious Disease, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
14
|
Huo R, Sun Q, Lv Q, Wang Y, Qi W, Zhang M, Li L, Wang X. Simvastatin ameliorates adverse pregnancy by inhibiting glycolysis-related NETs in obstetrical antiphospholipid syndrome. Life Sci 2024; 359:123215. [PMID: 39505298 DOI: 10.1016/j.lfs.2024.123215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024]
Abstract
AIMS Some patients with Obstetric Antiphospholipid Syndrome (OAPS) still experience miscarriage and placental dysfunction after routine treatment, which is related to an abnormal increase in neutrophil extracellular traps (NETs). The labeling of statins has been revised to remove the contraindication for use during pregnancy. Our aim is to investigate the effect of Simvastatin on pregnancy outcomes in OAPS and its correlation mechanisms with NETs. MAIN METHODS The effect of Simvastatin on pregnancy outcomes was observed. The effect of simvastatin on the function and apoptosis of neutrophils has evaluated. The effect of Simvastatin to NETs and the changes in oxidative stress levels were observed. Different groups of NETs were extracted to intervene the HTR8-Svneo.RNA-seq analysis of the mechanism of which Simvastatin reduces NETs. Seahorse experiment detected the effect of Simvastatin on neutrophil glycolysis levels. Fluorescence co-localization and flow cytometry and Co-IP were used to verify relevant mechanisms. KEY FINDINGS In the OAPS mice, Simvastatin can reduce embryo absorption rate, reshape placental blood flow perfusion. Simultaneously reducing the production of NETs both in vivo and vitro, remolding oxidative stress. Simvastatin can improve neutrophil dysfunction caused by aPL-IgG. The reduction of NETs improved HTR8-Svneo's dysfunction. The intervention of Simvastatin on neutrophils under the stimulation of aPL-IgG showed a signature in glycolytic. The key rate limiting enzyme PKM2 in glycolysis interacts with Cit-H2b and PI3K/AKT signaling pathway. SIGNIFICANCE Our study providing basic theoretical support for the treatment of OAPS with Simvastatin.
Collapse
Affiliation(s)
- Ruiheng Huo
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Qipeng Sun
- School of Clinical and Basic Medicine (Institute of Basic Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Qingfeng Lv
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Yuan Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Weiyi Qi
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, People's Republic of China
| | - Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China.
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China; School of Clinical and Basic Medicine (Institute of Basic Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, People's Republic of China.
| |
Collapse
|
15
|
Meng L, Yu Q, Zhao X, Chen L, Yang J, Wang Y, Chen H, Chen Y. Systemic risk factors of retinopathy in patients with systemic lupus erythematosus. Acta Ophthalmol 2024; 102:e774-e788. [PMID: 38396344 DOI: 10.1111/aos.16640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/07/2023] [Accepted: 01/15/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE To investigate the risk factors of lupus retinopathy (LR) in patients with systemic lupus erythematosus (SLE). METHODS This is a retrospective, cross-sectional study. LR patients admitted at Peking Union Medical College Hospital from June 2013 to April 2023 were reviewed. Age- and gender-matched SLE patients without retinopathy were selected as controls. Medical records including clinical manifestations, laboratory data and ophthalmic examination were collected. Univariate and multivariate logistic regression analyses were conducted. RESULTS One hundred and twelve LR patients (198 eyes) were included, with 12 cases (14 eyes) presenting with retinal macrovascular obstruction, and 100 cases (184 eyes) only exhibiting microvasculopathy. Multivariate analysis indicated the presence of haemolytic anaemia, decreased haemoglobin (HGB) and higher relative percentage of neutrophils were independent risk factors for LR (p < 0.05). The first two were also risk factors for retinal microvasculopathy, whereas secondary antiphospholipid syndrome (APS) was for macrovascular obstruction. In male group, LR had significant associations with decreased HGB, no matter which types of retinopathy (p < 0.05). In female group, LR was significantly associated with haemolytic anaemia, presence of antiphospholipid antibodies, decreased white blood cells and relative high percentage of neutrophils. Specifically, haemolytic anaemia (p = 0.002) was significantly associated with retinal microvasculopathy, and APS (p = 0.003) was significantly associated with macrovasculature obstruction. CONCLUSION LR was related to haemolytic anaemia, decreased HGB levels and higher percentage of neutrophils. Retinal microvasculopathy accounted for most cases and macrovasculature obstructions were rare. Male and female patients have distinct risk factors. Early ophthalmic screening is recommended especially for those with risk factors of LR.
Collapse
Affiliation(s)
- Lihui Meng
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianyi Yu
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lulu Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingyuan Yang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuelin Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youxin Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Reeves B. Blood cell JAKtivation aggravates cerebral venous thrombosis. Blood Adv 2024; 8:3327-3329. [PMID: 38916898 PMCID: PMC11258618 DOI: 10.1182/bloodadvances.2024012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024] Open
Affiliation(s)
- Brandi Reeves
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
17
|
Zhang S, Zhao X, Xue Y, Wang X, Chen XL. Advances in nanomaterial-targeted treatment of acute lung injury after burns. J Nanobiotechnology 2024; 22:342. [PMID: 38890721 PMCID: PMC11184898 DOI: 10.1186/s12951-024-02615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Acute lung injury (ALI) is a common complication in patients with severe burns and has a complex pathogenesis and high morbidity and mortality rates. A variety of drugs have been identified in the clinic for the treatment of ALI, but they have toxic side effects caused by easy degradation in the body and distribution throughout the body. In recent years, as the understanding of the mechanism underlying ALI has improved, scholars have developed a variety of new nanomaterials that can be safely and effectively targeted for the treatment of ALI. Most of these methods involve nanomaterials such as lipids, organic polymers, peptides, extracellular vesicles or cell membranes, inorganic nanoparticles and other nanomaterials, which are targeted to reach lung tissues to perform their functions through active targeting or passive targeting, a process that involves a variety of cells or organelles. In this review, first, the mechanisms and pathophysiological features of ALI occurrence after burn injury are reviewed, potential therapeutic targets for ALI are summarized, existing nanomaterials for the targeted treatment of ALI are classified, and possible problems and challenges of nanomaterials in the targeted treatment of ALI are discussed to provide a reference for the development of nanomaterials for the targeted treatment of ALI.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Yuhao Xue
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China.
| |
Collapse
|
18
|
Zen M, Tonello M, Favaro M, Del Ross T, Calligaro A, Giollo A, Vesentini F, Gennaio IA, Arru F, Ruffatti A, Doria A. Antiphospholipid antibody carriers and patients with quiescent antiphospholipid syndrome show persistent subclinical complement activation. Rheumatology (Oxford) 2024; 63:1733-1738. [PMID: 37774001 DOI: 10.1093/rheumatology/kead517] [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: 07/13/2023] [Revised: 08/20/2023] [Accepted: 09/01/2023] [Indexed: 10/01/2023] Open
Abstract
OBJECTIVES Complement activation has been advocated as one mechanism by which aPLs can induce thrombosis. In patients with catastrophic APS or re-thrombosis, enhanced complement activation has been shown, even in the quiescent phase of the disease. We aimed to assess complement activation and to investigate its association with clinical variables in aPL-positive patients with a favourable disease course. METHODS Subjects with at least two consecutive positive aPL results obtained ≥12 weeks apart were enrolled. They were subjects without a history of thrombosis or pregnancy morbidity (aPL carriers), patients with pregnancy morbidity alone, i.e. obstetric APS patients (OAPS patients), and/or patients with arterial, venous, or small-vessel thrombotic APS (TAPS patients); for enrolment, all patients were required to have been free of symptoms for ≥2 years. Patients affected with systemic autoimmune diseases were excluded. Healthy age- and sex-matched subjects were included as controls. Plasma C5a and C5b-9 levels were assessed by commercially available ELISA assays. The non-parametric Mann-Whitney test and Spearman's correlation were applied. RESULTS Thirty-seven OAPS patients, 38 TAPS patients, 42 aPL carriers and 30 healthy subjects were enrolled. The median C5a and C5b-9 levels were significantly higher in quiescent aPL-positive patients (OAPS, TAPS, aPL carriers) compared with controls: C5a ng/ml 10.61 [interquartile range (IQR) 6.87-15.46] vs 4.06 (2.66-7.35), P < 0.001; C5b-9 ng/ml 283.95 (175.8-439.40) vs 165.90 (124.23-236.8), P < 0.001. Similar C5a and C5b-9 levels were observed in OAPS and TAPS patients and aPL carriers. A positive correlation between the median C5b-9 levels and the number of aPL-positive tests was found (P = 0.002). CONCLUSION The persistence of aPL antibodies is associated with a persistent subclinical activation of the complement cascade.
Collapse
Affiliation(s)
- Margherita Zen
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Marta Tonello
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Maria Favaro
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Teresa Del Ross
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Antonia Calligaro
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Alessandro Giollo
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Filippo Vesentini
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Ilenia Anna Gennaio
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Federico Arru
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Amelia Ruffatti
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| | - Andrea Doria
- Department of Medicine, Rheumatology Unit, University of Padua, Padova, Italy
| |
Collapse
|
19
|
Rhein AK, Rabinovich A, Abuhasira R, Lubaton-Barshishat S, Erez O. Obstetric antiphospholipid syndrome carries an increased lifetime risk for obstetric and thrombotic complications-a population-based study. Res Pract Thromb Haemost 2024; 8:102430. [PMID: 38798792 PMCID: PMC11127162 DOI: 10.1016/j.rpth.2024.102430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Background Antiphospholipid syndrome (APS) can present with either a thromboembolic event (thrombotic APS, TAPS) or an obstetric complication (obstetric APS, OAPS). Data on long-term complications in the different APS phenotypes are limited. Objectives We aimed to compare obstetric history, antiphospholipid antibody profiles, obstetric and thromboembolic complications, and pregnancy outcomes between TAPS and OAPS. Methods This retrospective cohort study included women who delivered singleton pregnancies between 1998 and 2020. One hundred sixteen thousand four hundred nine women were included, resulting in 320,455 deliveries. Among the included patients, 71 were diagnosed with APS, 49 were classified as OAPS, and 22 as TAPS. The demographics, obstetric, neonatal, and thrombotic outcomes were compared among TAPS, OAPS, and the general obstetric population. Results OAPS patients had an increased risk of thrombotic events compared with the general obstetric population (odds ratio [OR] 18.0; 95% CI, 8.7-37.2). In pregnancies following the diagnosis of APS, despite standard antithrombotic treatment, OAPS patients exhibited an elevated risk of placenta-related and neonatal complications compared with the general obstetric population (late fetal loss [adjusted OR {aOR}, 15.3; 95% CI, 0.5-27.5], stillbirth [aOR, 5.9; 95% CI, 2.2-15.4], placental abruption [aOR, 4.8; 95% CI, 1.5-15.3], preeclampsia [aOR, 4.4; 95% CI, 2.5-7.7], fetal growth restriction [aOR, 4.3; 95% CI, 8.5-27.5], small for gestational age neonate [aOR, 4.0; 95% CI, 2.4-6.6], and low Apgar scores [Apgar'1: aOR, 2.6; 95% CI, 1.3-10.4; Apgar'5: aOR, 3.7; 95% CI, 1.3-10.4]). TAPS patients exhibited increased risk of preeclampsia (aOR, 3.1; 95% CI, 1.2-8). Conclusion OAPS patients exhibit a heightened risk of thrombotic events compared with the general obstetric population. Despite treatment, OAPS and TAPS still presented obstetric complications. These findings, after confirmation in prospective studies, need to be taken into consideration when planning the treatment approach for these patients.
Collapse
Affiliation(s)
- Ariel Katherine Rhein
- The Joyce & Irving Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Rabinovich
- Thrombosis and Hemostasis Unit, Hematology Institute, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ran Abuhasira
- The Joyce & Irving Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Clinical Research Center, Soroka University Medical Center, Beer-Sheva, Israel
| | - Shir Lubaton-Barshishat
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
- Department of Obstetrics and Gynecology, Hutzel Women's Hospital, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
20
|
Foret T, Dufrost V, Lagrange J, Costa P, Mourey G, Lecompte T, Magy-Bertrand N, Regnault V, Zuily S, Wahl D. Thrombin Generation Assay in Antiphospholipid Antibodies Positive Subjects as a Personalized Thrombotic Risk Assessment: State of the Art and Perspectives. Curr Rheumatol Rep 2024; 26:178-187. [PMID: 38372872 DOI: 10.1007/s11926-024-01140-1] [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] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE OF THE REVIEW Thrombotic risk assessment in antiphospholipid positive (aPL +) subjects is a major challenge, and the study of in vitro thrombin generation (thrombin generation assays (TGA)) could provide useful information. Activated protein C (APC) sensitivity is involved in thrombotic events in antiphospholipid syndrome patients. We summarized methods used to assess APC sensitivity with TGA and evaluated the prognostic role of APC resistance through literature search. RECENT FINDINGS APC resistance induced by aPL is a complex pathway. Several cross-sectional studies assessed APC sensitivity to understand thrombotic event mechanisms in aPL + subjects. Only one prospective cohort had investigated the prognostic impact of APC resistance in aPL + subjects, with a positive and significant correlation between APC sensitivity and the risk of thrombosis during the follow up (hazard ratio, 6.07 [95% CI, 1.69-21.87]). APC resistance assessed with TGA could be associated with thrombotic events in aPL + subjects.
Collapse
Affiliation(s)
- Thomas Foret
- Vascular Medicine Unit, Vascular and Endovascular Surgery Department, CHU-Besancon, 3 BD Alexandre Fleming, F-25000, Besancon, France.
- Université de Franche-Comté, SINERGIES, F-25000, Besancon, France.
| | - Virginie Dufrost
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- Vascular Medicine Division and National Referral Center for Rare Vascular and Systemic Autoimmune Diseases, CHRU-Nancy, F-54000, Nancy, France
| | - Jeremy Lagrange
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- CHRU-Nancy, F-54000, Nancy, France
| | - Patricia Costa
- Vascular Medicine Unit, Vascular and Endovascular Surgery Department, CHU-Besancon, 3 BD Alexandre Fleming, F-25000, Besancon, France
| | - Guillaume Mourey
- Université de Franche-Comté, SINERGIES, F-25000, Besancon, France
- Medical Biology Laboratory, Biological Haemostasis Department, CHU Besançon, F-25000, Besançon, France
| | - Thomas Lecompte
- Vascular Medicine Division and National Referral Center for Rare Vascular and Systemic Autoimmune Diseases, CHRU-Nancy, F-54000, Nancy, France
- Université de Lorraine, Nancy, France
| | | | - Veronique Regnault
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- CHRU-Nancy, F-54000, Nancy, France
| | - Stéphane Zuily
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- Vascular Medicine Division and National Referral Center for Rare Vascular and Systemic Autoimmune Diseases, CHRU-Nancy, F-54000, Nancy, France
| | - Denis Wahl
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- Vascular Medicine Division and National Referral Center for Rare Vascular and Systemic Autoimmune Diseases, CHRU-Nancy, F-54000, Nancy, France
| |
Collapse
|
21
|
Hoxha A, Perin N, Lovisotto M, Calligaro A, Del Ross T, Favaro M, Tonello M, Doria A, Simioni P. Risk factors for damage accrual in primary antiphospholipid syndrome: A retrospective single-center cohort study. J Autoimmun 2024; 144:103180. [PMID: 38368768 DOI: 10.1016/j.jaut.2024.103180] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Despite anticoagulant therapy, a antiphospholipid syndrome (APS) has a higher rate of recurrent events, which can lead to damage accrual and a negative impact on life quality. OBJECTIVES To evaluate the risk factors and APS subsets associated with damage accrual. PATIENTS/METHODS We conducted a retrospective single-center study. We reviewed the medical records of 282 APS patients, with a median age of 36 (IQR 30-46) years and a median of 195 (IQR 137-272) months. The primary endpoint was damage accrual during follow-up, defined as organ/tissue impairment present for at least six months or causing permanent loss. The secondary endpoints were early organ damage within six months of disease onset and death. RESULTS Eighty (28.4%) patients presented damage accrual; 52.5% developed damage within six months of APS onset, and 41.3% had more than one organ involved. Neuropsychiatric involvement, affecting 38.8% of the patients, was the most frequent, followed by peripheral vasculopathy and renal involvement, 35% either. Death happened in 7 (2.5 %) patients; damage accrual was associated with a 6-fold risk of death [OR 6.7 (95% CI 1.3-35.1), p = 0.03]. Microangiopathy and non-criteria manifestations were independent risk factors for damage accrual with 5-fold and 4-fold higher risk, respectively [(OR 4.9 (95% CI 2.1-11.7), p < 0.0001 and (OR 3.8 (95% CI 1.5-10.1), p = 0.007]. The cumulative incidence of damage accrual increased by 5.7-fold and 3.6-fold in patients with microangiopathy and non-criteria manifestations. CONCLUSIONS APS patients had a higher frequency of damage accrual. Microangiopathy and non-criteria manifestations were independent risk factors for damage accrual.
Collapse
Affiliation(s)
- Ariela Hoxha
- Internal Medicine Unit, Thrombotic and Hemorrhagic Center, Department of Medicine, University of Padua, Italy.
| | - Nicola Perin
- Internal Medicine Unit, Thrombotic and Hemorrhagic Center, Department of Medicine, University of Padua, Italy
| | - Marco Lovisotto
- Internal Medicine Unit, Thrombotic and Hemorrhagic Center, Department of Medicine, University of Padua, Italy
| | - Antonia Calligaro
- Rheumatology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Teresa Del Ross
- Rheumatology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Maria Favaro
- Rheumatology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Marta Tonello
- Rheumatology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Paolo Simioni
- Internal Medicine Unit, Thrombotic and Hemorrhagic Center, Department of Medicine, University of Padua, Italy
| |
Collapse
|
22
|
Li J, Peng L, Wu L, Ding Y, Duan X, Xu J, Wei W, Chen Z, Zhao C, Yang M, Jiang N, Zhang S, Wang Q, Tian X, Li M, Zeng X, Zhao Y, Zhao J. Antiphospholipid antibodies as potential predictors of disease severity and poor prognosis in systemic lupus erythematosus-associated thrombocytopenia: results from a real-world CSTAR cohort study. Arthritis Res Ther 2024; 26:67. [PMID: 38475924 DOI: 10.1186/s13075-024-03305-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND To investigate the role of antiphospholipid antibodies (aPLs) in the disease severity and prognosis of SLE-related thrombocytopenia (SLE-TP). METHODS This multicenter prospective study was conducted based on data from the CSTAR registry. TP was defined as a platelet count<100 × 109/L. Demographic characteristics, platelet count, clinical manifestations, disease activity, and autoantibody profiles were collected at baseline. Relapse was defined as the loss of remission. Bone marrow aspirate reports were also collected. RESULTS A total of 350 SLE-TP patients with complete follow-up data, 194 (55.4%) were aPLs positive. At baseline, SLE-TP patients with aPLs had lower baseline platelet counts (61.0 × 109/L vs. 76.5 × 109/L, P<0.001), and a higher proportion of moderate to severe cases (24.2% vs. 14.1% ; 18.0% vs. 8.3%, P<0.001). SLE-TP patients with aPLs also had lower platelet counts at their lowest point (37.0 × 109/L vs. 51.0 × 109/L, P = 0.002). In addition, thean increasing number of aPLs types was associated with a decrease in the baseline and minimum values of platelets ( P<0.001, P = 0.001). During follow-up, SLE-TP carrying aPLs had a higher relapse rate (58.2% vs. 44.2%, P = 0.009) and a lower complete response (CR) rate. As the types of aPLs increased, the relapse rate increased, and the CR rate decreased. Furthermore, there was no significant difference in the ratio of granulocytes to red blood cells (G/E), the total number of megakaryocyte and categories. CONCLUSION SLE-TP patients with positive aPLs had more severe disease a lower remission rate but a higher relapse rate.
Collapse
Affiliation(s)
- Jun Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Liying Peng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Lijun Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Yufang Ding
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Xinwang Duan
- Department of Rheumatology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jian Xu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhen Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Cheng Zhao
- Department of Rheumatology and Immunology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Min Yang
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Nan Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China.
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China.
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
23
|
López-Pedrera C, Cerdó T, Jury EC, Muñoz-Barrera L, Escudero-Contreras A, Aguirre MA, Pérez-Sánchez C. New advances in genomics and epigenetics in antiphospholipid syndrome. Rheumatology (Oxford) 2024; 63:SI14-SI23. [PMID: 38320594 PMCID: PMC10846911 DOI: 10.1093/rheumatology/kead575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/03/2023] [Accepted: 10/10/2023] [Indexed: 02/08/2024] Open
Abstract
APS patients exhibit a wide clinical heterogeneity in terms of the disease's origin and progression. This diversity can be attributed to consistent aPL profiles and other genetic and acquired risk factors. Therefore, understanding the pathophysiology of APS requires the identification of specific molecular signatures that can explain the pro-atherosclerotic, pro-thrombotic and inflammatory states observed in this autoimmune disorder. In recent years, significant progress has been made in uncovering gene profiles and understanding the intricate epigenetic mechanisms and microRNA changes that regulate their expression. These advancements have highlighted the crucial role played by these regulators in influencing various clinical aspects of APS. This review delves into the recent advancements in genomic and epigenetic approaches used to uncover the mechanisms contributing to vascular and obstetric involvement in APS. Furthermore, we discuss the implementation of novel bioinformatics tools that facilitate the investigation of these mechanisms and pave the way for personalized medicine in APS.
Collapse
Affiliation(s)
- Chary López-Pedrera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Tomás Cerdó
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Laura Muñoz-Barrera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - M A Aguirre
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Carlos Pérez-Sánchez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| |
Collapse
|
24
|
Ruiz-Irastorza G, Tektonidou MG, Khamashta M. Anticoagulant and non-anticoagulant therapy in thrombotic antiphospholipid syndrome: old drugs and new treatment targets. Rheumatology (Oxford) 2024; 63:SI96-SI106. [PMID: 38320592 DOI: 10.1093/rheumatology/kead538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 02/08/2024] Open
Abstract
In this review, we discuss the current evidence on classic and newer oral anticoagulant therapy, older drugs such as HCQ and statins, and new potential treatment targets in APS. Vitamin K antagonists (VKAs) remain the cornerstone treatment for thrombotic events in APS. In patients fulfilling criteria for definite APS presenting with a first venous thrombosis, treatment with VKAs with a target international normalized ratio (INR) 2.0-3.0 is recommended. In patients with arterial thrombosis, treatment with VKA with target INR 2.0-3.0 or 3.0-4.0 is recommended by recent guidelines, considering the individual's bleeding and thrombosis recurrence risk. A combination of VKAs and low-dose aspirin (75-100 mg/daily) may also be considered. According to available evidence direct oral anticoagulants should be avoided in patients with arterial thrombosis and/or those with triple aPL positivity. Adjunctive treatment with HCQ and/or statins can be considered, especially in anticoagulation treatment-refractory APS. Potential targeted treatments in APS include B-cell targeting, complement inhibition, mammalian target of rapamycin inhibition, IFN targeting, adenosine receptors agonists, CD38 targeting or chimeric antigen receptor T-cell therapy. The safety and efficacy of these treatment targets needs to be examined in well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Guillermo Ruiz-Irastorza
- Autoimmune Diseases Research Unit, Biocruces Bizkaia Health Research Institute, The Basque Country, Bizkaia, Spain
- University of The Basque Country, The Basque Country, Bizkaia, Spain
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Munther Khamashta
- Department of Women & Children's Health, King's College London, London, UK
| |
Collapse
|
25
|
Sachetto ATA, Mackman N. Monocyte Tissue Factor Expression: Lipopolysaccharide Induction and Roles in Pathological Activation of Coagulation. Thromb Haemost 2023; 123:1017-1033. [PMID: 37168007 PMCID: PMC10615589 DOI: 10.1055/a-2091-7006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The coagulation system is a part of the mammalian host defense system. Pathogens and pathogen components, such as bacterial lipopolysaccharide (LPS), induce tissue factor (TF) expression in circulating monocytes that then activates the coagulation protease cascade. Formation of a clot limits dissemination of pathogens, enhances the recruitment of immune cells, and facilitates killing of pathogens. However, excessive activation of coagulation can lead to thrombosis. Here, we review studies on the mechanism of LPS induction of TF expression in monocytes and its contribution to thrombosis and disseminated intravascular coagulation. Binding of LPS to Toll-like receptor 4 on monocytes induces a transient expression of TF that involves activation of intracellular signaling pathways and binding of various transcription factors, such as c-rel/p65 and c-Fos/c-Jun, to the TF promoter. Inhibition of TF in endotoxemia and sepsis models reduces activation of coagulation and improves survival. Studies with endotoxemic mice showed that hematopoietic cells and myeloid cells play major roles in the activation of coagulation. Monocyte TF expression is also increased after surgery. Activated monocytes release TF-positive extracellular vesicles (EVs) and levels of circulating TF-positive EVs are increased in endotoxemic mice and in patients with sepsis. More recently, it was shown that inflammasomes contribute to the induction of TF expression and activation of coagulation in endotoxemic mice. Taken together, these studies indicate that monocyte TF plays a major role in activation of coagulation. Selective inhibition of monocyte TF expression may reduce pathologic activation of coagulation in sepsis and other diseases without affecting hemostasis.
Collapse
Affiliation(s)
- Ana T. A. Sachetto
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
26
|
Reshetnyak T, Nurbaeva K. The Role of Neutrophil Extracellular Traps (NETs) in the Pathogenesis of Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Int J Mol Sci 2023; 24:13581. [PMID: 37686381 PMCID: PMC10487763 DOI: 10.3390/ijms241713581] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/10/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease of unknown aetiology [...].
Collapse
Affiliation(s)
- Tatiana Reshetnyak
- Department of Thromboinflammation, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russia;
| | | |
Collapse
|