1
|
Álvarez D, Winter HE, Velasquez Franco CJ, Castellanos Gutierrez AS, Baños N, Markert UR, Cadavid ÁP, Morales‐Prieto DM. Improving diagnosis in patients with obstetric antiphospholipid syndrome through the evaluation of non-criteria antibodies. Clin Transl Immunology 2024; 13:e70021. [PMID: 39678173 PMCID: PMC11638733 DOI: 10.1002/cti2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/13/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Objectives Antiphospholipid syndrome (APS) is an autoimmune disease driven by antiphospholipid antibodies (aPL). Currently, APS diagnosis requires a combination of clinical manifestations (thrombosis and/or obstetric morbidity) and the persistent presence of at least one criteria aPL: anti-cardiolipin antibodies (aCL), anti-β2-glycoprotein I antibodies (aβ2GPI) or lupus anticoagulant (LA). Patients with suggestive obstetric symptoms but lacking criteria aPL face diagnostic challenges. Non-criteria aPL screening may enhance discrimination. This study proposes a classification incorporating both criteria and non-criteria antibodies to improve obstetric APS diagnosis. Methods Blood samples from non-pregnant women (n = 68) with a history of vascular, obstetric, or vascular and obstetric manifestations were analysed. Among them, 30 had previous diagnosis of APS. Healthy women with proven gestational success were included as controls (n = 16). Criteria and non-criteria (anti-phosphatidylglycerol, anti-phosphatidylethanolamine, anti-phosphatidylinositol, anti-phosphatidylserine and anti-phosphatidic acid) IgG aPL were evaluated by ELISA and coagulation tests. Based on the resulting aPL profile, patients were reclassified. Responsiveness to treatment was obtained from medical records. Results Criteria aPL levels marginally differentiated women previously managed as obstetric APS from unexplained/other causes of obstetric morbidity. Including non-criteria aPL improved separation. The proposed classification identified an obstetric APS group that exhibits non-criteria aPL and aβ2GPI titres below the cut-off but higher than healthy women (7.88 vs. 2.47 SGU, P = 0.006). Compared to cases of other causes of obstetric morbidity, these patients retrospectively responded better to aspirin and/or heparin treatment (71.43% vs. 11.11%, P = 0.035). Conclusions Assessing non-criteria antibodies may identify isolated obstetric APS cases benefiting from established therapies.
Collapse
Affiliation(s)
- Daniel Álvarez
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de MedicinaUniversidad de Antioquia UdeAMedellínColombia
- Placenta Lab, Department of ObstetricsJena University HospitalJenaGermany
| | - Hephzibah E Winter
- Placenta Lab, Department of ObstetricsJena University HospitalJenaGermany
| | | | - Aleida Susana Castellanos Gutierrez
- Placenta Lab, Department of ObstetricsJena University HospitalJenaGermany
- BCNatalBarcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecologia, Obstetrícia i Neonatologia Fetal i+D Fetal Medicine Research CenterBarcelonaSpain
| | - Núria Baños
- BCNatalBarcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecologia, Obstetrícia i Neonatologia Fetal i+D Fetal Medicine Research CenterBarcelonaSpain
| | - Udo R Markert
- Placenta Lab, Department of ObstetricsJena University HospitalJenaGermany
| | - Ángela P Cadavid
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de MedicinaUniversidad de Antioquia UdeAMedellínColombia
- Grupo de Investigación en Trombosis, Departamento Medicina Interna, Facultad de MedicinaUniversidad de Antioquia UdeAMedellínColombia
| | | |
Collapse
|
2
|
Feng W, Qiao J, Tan Y, Liu Q, Wang Q, Yang B, Yang S, Cui L. Interaction of antiphospholipid antibodies with endothelial cells in antiphospholipid syndrome. Front Immunol 2024; 15:1361519. [PMID: 39044818 PMCID: PMC11263079 DOI: 10.3389/fimmu.2024.1361519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease with arteriovenous thrombosis and recurrent miscarriages as the main clinical manifestations. Due to the complexity of its mechanisms and the diversity of its manifestations, its diagnosis and treatment remain challenging issues. Antiphospholipid antibodies (aPL) not only serve as crucial "biomarkers" in diagnosing APS but also act as the "culprits" of the disease. Endothelial cells (ECs), as one of the core target cells of aPL, bridge the gap between the molecular level of these antibodies and the tissue and organ level of pathological changes. A more in-depth exploration of the relationship between ECs and the pathogenesis of APS holds the potential for significant advancements in the precise diagnosis, classification, and therapy of APS. Many researchers have highlighted the vital involvement of ECs in APS and the underlying mechanisms governing their functionality. Through extensive in vitro and in vivo experiments, they have identified multiple aPL receptors on the EC membrane and various intracellular pathways. This article furnishes a comprehensive overview and summary of these receptors and signaling pathways, offering prospective targets for APS therapy.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| |
Collapse
|
3
|
Andreoli L, Regola F, Caproli A, Crisafulli F, Fredi M, Lazzaroni MG, Nalli C, Piantoni S, Zatti S, Franceschini F, Tincani A. Pregnancy in antiphospholipid syndrome: what should a rheumatologist know? Rheumatology (Oxford) 2024; 63:SI86-SI95. [PMID: 38320595 DOI: 10.1093/rheumatology/kead537] [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: 07/31/2023] [Accepted: 09/03/2023] [Indexed: 02/08/2024] Open
Abstract
This review focuses on the management of reproductive issues in women who have antiphospholipid syndrome (APS) or are carriers of antiphospholipid antibodies (aPL). The importance of aPL detection during preconception counselling relies on their pathogenic potential for placental insufficiency and related obstetric complications. The risk of adverse pregnancy outcomes can be minimized by individualized risk stratification and tailored treatment aimed at preventing placental insufficiency. Combination therapy of low-dose acetylsalicylic acid and heparin is the mainstay of prophylaxis during pregnancy; immunomodulation, especially with hydroxychloroquine, should be considered in refractory cases. Supplementary ultrasound surveillance is useful to detect fetal growth restriction and correctly tailor the time of delivery. The individual aPL profile must be considered in the stratification of thrombotic risk, such as during assisted reproduction techniques requiring hormonal ovarian stimulation or during the follow-up after pregnancy in order to prevent the first vascular event.
Collapse
Affiliation(s)
- Laura Andreoli
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesca Regola
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessia Caproli
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesca Crisafulli
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Maria-Grazia Lazzaroni
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cecilia Nalli
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Sonia Zatti
- Department of Obstetrics and Gynaecology, ASST Spedali Civili, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology Unit - ERN ReCONNET, ASST Spedali Civili; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
4
|
Zhang Y, Jin S, Tian W, Shi D, Chen Y, Cui L, Zheng J. Proteomics of Serum Samples for the Exploration of the Pathological Mechanism of Obstetric Antiphospholipid Syndrome. J Proteome Res 2024; 23:289-300. [PMID: 38048430 PMCID: PMC10775856 DOI: 10.1021/acs.jproteome.3c00554] [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: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023]
Abstract
Obstetric antiphospholipid syndrome (OAPS) is a multisystem disorder characterized by thrombosis or recurrent fetal loss. In this study, we aim to explore the pathological mechanism of OAPS. Herein, we carried out data-independent acquisition (DIA) mass spectrometry quantitative proteomic analysis of serum samples from OAPS patients and healthy controls. A set of 93 differentially expressed proteins was identified, including 75 upregulated and 18 downregulated proteins compared with the levels in controls. Those proteins are enriched in KEGG pathways related to autoimmune diseases, allergic diseases, and pathogen infection. Interestingly, metabolic pathways such as fatty acid degradation and type I diabetes were enriched, indicating that OAPS is metabolic disease related. The significantly increased triglyceride also supported this idea. The differentially expressed proteins insulin-like growth factor-binding protein-1 (IGFBP-1), C-reactive protein (CRP), and ferritin light chain (FTL) were validated by ELISA. Our study presented a deep serum proteomics of OAPS and advanced our understanding of OAPS pathogenesis.
Collapse
Affiliation(s)
- Yinmei Zhang
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Shangjia Jin
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Wenmin Tian
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Dongxue Shi
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yang Chen
- Department
of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics
Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Liyan Cui
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| | - Jiajia Zheng
- Department
of Laboratory Medicine, Peking University
Third Hospital, Beijing 100191, China
| |
Collapse
|
5
|
Ji L, Zhang Z, Perl A. The mTOR pathway in the antiphospholipid syndrome. MEDICAL REVIEW (2021) 2023; 3:511-513. [PMID: 38282799 PMCID: PMC10808841 DOI: 10.1515/mr-2023-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/26/2023] [Indexed: 01/30/2024]
Abstract
This perspective discussed the available evidence on the involvement of mTOR pathway in antiphospholipid syndrome (APS), from the aspects of endothelial cells, platelets, monocytes and anti-phospholipid antibodies (PLs), which may lead to future therapeutic applications of mTOR inhibition in APS.
Collapse
Affiliation(s)
- Lanlan Ji
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Zhuoli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, Division of Rheumatology and Clinical Immunology, State University of New York, UMU, College of Medicine, Syracuse, NY, USA
| |
Collapse
|
6
|
Qin R, Wu H, Guan H, Tang C, Zheng Z, Deng C, Chen C, Zou Q, Lu L, Ma K. Anti-phospholipid autoantibodies in human diseases. Clin Immunol 2023; 256:109803. [PMID: 37821073 DOI: 10.1016/j.clim.2023.109803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
Anti-phospholipid autoantibodies are a group of antibodies that can specifically bind to anionic phospholipids and phospholipid protein complexes. Recent studies have reported elevated serum anti-phospholipid autoantibody levels in patients with antiphospholipid syndrome, systemic lupus erythematosus, rheumatoid arthritis, metabolic disorders, malaria, SARS-CoV-2 infection, obstetric diseases and cardiovascular diseases. However, the underlying mechanisms of anti-phospholipid autoantibodies in disease pathogenesis remain largely unclear. Emerging evidence indicate that anti-phospholipid autoantibodies modulate NETs formation, monocyte activation, blockade of apoptotic cell phagocytosis in macrophages, complement activation, dendritic cell activation and vascular endothelial cell activation. Herein, we provide an update on recent advances in elucidating the effector mechanisms of anti-phospholipid autoantibodies in the pathogenesis of various diseases, which may facilitate the development of potential therapeutic targets for the treatment of anti-phospholipid autoantibody-related disorders.
Collapse
Affiliation(s)
- Rencai Qin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Haiqi Wu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Hui Guan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chun Tang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhihua Zheng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chong Deng
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China
| | - Chengshun Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China.
| | - Kongyang Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
7
|
An R, Yang Y, Liu L, Li P. SAMD1 attenuates antiphospholipid syndrome-induced pregnancy complications. Immun Inflamm Dis 2023; 11:e1006. [PMID: 37904675 PMCID: PMC10614121 DOI: 10.1002/iid3.1006] [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: 03/07/2023] [Revised: 06/28/2023] [Accepted: 08/31/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE This study was intended to investigate the effect of SAMD1 on antiphospholipid syndrome (APS)-induced pregnancy complications in mice. METHODS The mRNA and protein expression of SAMD1 in APS patients and healthy controls was detected by qRT-PCR and western blot. Anti-B2 GPI and ACA levels were tested by ELISA, MMP-9, iNOS, ICAM-1 and MCP-1 mRNA and protein levels determined by qRT-PCR and western blot, cellular senescence detected by β-galactosidase staining, cell proliferation ability detected by CCK-8 assay, cell viability detected by trypan blue staining, cell mobility detected by Transwell, and cell angiogenesis ability detected by matrigel tube formation assay. An APS pregnant mouse model was constructed, and the embryo absorption rate was calculated. RESULTS SAMD1 expression was low in serum of APS patients, which was correlated with the history of thrombosis and the number of adverse pregnancies. Anti-B2 GPI and ACA levels were increased in APS. The expressions of MMP-9, iNOS, ICAM-1, and MCP-1 were also significantly upregulated in HUVECs treated with APS serum. APS promoted HUVEC senescence and inhibited cell proliferation, migration and angiogenesis. Overexpression of SAMD1 reversed the above results. Experiments on the APS pregnant mouse model confirmed that overexpression of SAMD1 reduced the rate of fetal loss. CONCLUSION SAMD1 may reduce APS-induced embryo loss by regulating cellular senescence, proliferation, migration, and angiogenesis.
Collapse
Affiliation(s)
- Ran An
- Department of Obstetrics and GynecologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangP.R. China
| | - Yanqi Yang
- Department of Obstetrics and GynecologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangP.R. China
| | - Lei Liu
- Department of Obstetrics and GynecologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangP.R. China
| | - Peiling Li
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangP.R. China
| |
Collapse
|
8
|
Mineo C, Shaul PW, Bermas BL. The pathogenesis of obstetric APS: a 2023 update. Clin Immunol 2023; 255:109745. [PMID: 37625670 PMCID: PMC11366079 DOI: 10.1016/j.clim.2023.109745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the persistent presence of antibodies directed against phospholipids and phospholipid-binding proteins that are associated with thrombosis and pregnancy-related morbidity. The latter includes fetal deaths, premature birth and maternal complications. In the early 1990s, a distinct set of autoantibodies, termed collectively antiphospholipid antibodies (aPL), were identified as the causative agents of this disorder. Subsequently histological analyses of the placenta from APS pregnancies revealed various abnormalities, including inflammation at maternal-fetal interface and poor placentation manifested by reduced trophoblast invasion and limited uterine spiral artery remodeling. Further preclinical investigations identified the molecular targets of aPL and the downstream intracellular pathways of key placental cell types. While these discoveries suggest potential therapeutics for this disorder, definitive clinical trials have not been completed. This concise review focuses on the recent developments in the field of basic and translational research pursuing novel mechanisms underlying obstetric APS.
Collapse
Affiliation(s)
- Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Bonnie L Bermas
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Granada-Gómez M, Velásquez-Berrío M, Molina CR, Martín SS, Escudero C, Alvarez AM, Cadavid AP. Modulation of the activation of endothelial nitric oxide synthase and nitrosative stress biomarkers by aspirin triggered lipoxins: A possible mechanism of action of aspirin in the antiphospholipid syndrome. Am J Reprod Immunol 2023; 90:e13753. [PMID: 37491919 DOI: 10.1111/aji.13753] [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: 04/09/2022] [Revised: 06/20/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Antiphospholipid syndrome (APS) is characterized by the clinical manifestation of vascular thrombosis (VT) or pregnancy morbidity (PM) and antiphospholipid antibodies (aPL) that can modify the nitric oxide production. Low-dose aspirin is used in the prevention and treatment of diverse alterations of pregnancy. One of the mechanisms of action of aspirin is to induce the production of aspirin-triggered-lipoxins (ATL). The aim of this study was to evaluate the modulatory effect of ATL over the activation of endothelial nitric oxide synthase (eNOS) and nitrosative stress biomarkers induced by aPL. METHODS We used polyclonal IgG and sera from women with aPL and PM/VT or VT only, and from women with PM only and positive for non-criteria aPL (SN-OAPS). In these sera, biomarkers of nitrosative stress (nitrites and nitrotyrosine) were measured. The protein expression of nitrotyrosine and the phosphorylation of eNOS (at Ser1177) were estimated in human umbilical vein endothelial cells (HUVECs) stimulated with polyclonal IgG with or without ATL. RESULTS Women with SN-OAPS showed increased circulating levels of nitrites and nitrotyrosine. Likewise, polyclonal IgG from either SN-OAPS or VT patients stimulated nitrotyrosine expression in HUVECs. ATL decreased the nitrotyrosine expression induced by polyclonal IgG from the SN-OAPS group. ATL also recovered the reduced eNOS phosphorylation at Ser1177 in HUVECs stimulated with polyclonal IgG from women with PM/VT or SN-OAPS. CONCLUSIONS Increased nitrosative stress present in serum of women with SN-OAPS is associated with IgG-mediated impaired endothelial NO synthesis in endothelial cells. ATL prevent these cellular changes.
Collapse
Affiliation(s)
- Manuel Granada-Gómez
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Manuela Velásquez-Berrío
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Carolina Rúa Molina
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Sebastián San Martín
- Biomedical Research Center School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Basic Sciences Department, Faculty of Sciences, Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM)
| | - Angela M Alvarez
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Angela P Cadavid
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM)
| |
Collapse
|
10
|
Patsouras M, Alexopoulou E, Foutadakis S, Tsiki E, Karagianni P, Agelopoulos M, Vlachoyiannopoulos PG. Antiphospholipid antibodies induce proinflammatory and procoagulant pathways in endothelial cells. J Transl Autoimmun 2023; 6:100202. [PMID: 37216142 PMCID: PMC10197110 DOI: 10.1016/j.jtauto.2023.100202] [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: 08/30/2022] [Revised: 03/01/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia characterized by recurrent thrombotic events and/or pregnancy morbidity in the presence of antiphospholipid antibodies detected either as anti-cardiolipin, anti-β2 Glycoprotein I (anti-β2GPI) or Lupus anticoagulant (LA). Endothelial deregulation characterizes the syndrome. To address gene expression changes accompanying the development of autoimmune phenotype in endothelial cells in the context of APS, we performed transcriptomics analysis in Human Umbilical Vein Endothelial Cells (HUVECs) stimulated with IgG from APS patients and β2GPI, followed by intersection of RNA-seq data with published microarray and ChIP-seq results (Chromatin Immunoprecipitation). Our strategy revealed that during HUVEC activation diverse signaling pathways such as TNF-α, TGF-β, MAPK38, and Hippo are triggered as indicated by Gene Ontology (GO) classification and pathway analysis. Finally, cell biology approaches performed side-by-side in naïve and stimulated cultured HUVECs, as well as, in placenta specimens derived from Healthy donors (HDs) and APS-patients verified the evolution of an APS-characteristic gene expression program in endothelial cells during the initial stages of the disease's development.
Collapse
Affiliation(s)
- Markos Patsouras
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eirini Alexopoulou
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Spyros Foutadakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | - Eirini Tsiki
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Panagiota Karagianni
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Marios Agelopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | | |
Collapse
|
11
|
Álvarez D, Rúa C, Velásquez Berrío M, Cataño JU, Escudero C, Cadavid J ÁP. Extracellular vesicles released upon stimulation with antiphospholipid antibodies: An actual direct procoagulant mechanism or a new factor in the lupus anticoagulant paradox? J Autoimmun 2022; 133:102905. [PMID: 36115210 DOI: 10.1016/j.jaut.2022.102905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 12/13/2022]
Abstract
Antiphospholipid antibodies (aPL) lead to a hypercoagulable state in vivo. Paradoxically, some of these autoantibodies perform as inhibitors of the coagulation cascade in vitro (a phenomenon referred to as "lupus anticoagulant"). The presence of lupus anticoagulant has been related to an increased quantity of plasma extracellular vesicles, which may constitute a direct procoagulant mechanism in antiphospholipid syndrome. This study investigates whether or not endothelial cell-derived extracellular vesicles released upon stimulation with aPL (aPL-EDEVs) are related to a higher direct coagulation activity. Using an in vitro model of endothelium, flow cytometry and a recalcified plasma-based assay, we found that the coagulation activity of aPL-EDEVs is mainly conditioned by the lupus anticoagulant-like activity of autoantibodies. Nevertheless, in the presence of β2 glycoprotein I, a cofactor of aPL during the stimulation of endothelial cells, the coagulation activity of EDEVs is restored in a mitogen-activated protein kinase kinases 1 and 2 (MEK1/2)-dependent manner. This phenomenon was especially evident when using immunoglobulins G from patients with vascular and obstetric primary antiphospholipid syndrome who manifest refractoriness to treatment. Our findings suggest that the role of aPL-EDEVs in the antiphospholipid syndrome-related hypercoagulable state may not rely on their capacity to enhance clotting directly. While β2 glycoprotein I performs as a procoagulant cofactor and restores the coagulation activity of extracellular vesicles via MEK1/2 pathway, proportionally, autoantibodies interact with aPL-EDEVs and exhaust their coagulation properties. Further analysis is required to establish whether lupus anticoagulant-like autoantibodies opsonise extracellular vesicles and whether opsonised vesicles may lead to thrombosis by indirect means.
Collapse
Affiliation(s)
- Daniel Álvarez
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Carolina Rúa
- Grupo de Investigación en Trombosis, Departamento Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Manuela Velásquez Berrío
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - John Ubeimar Cataño
- Grupo de Investigación en Trombosis, Departamento Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia; Hospital San Vicente Fundación, Medellín, Colombia
| | - Carlos Escudero
- Vascular Physiology Laboratory, Group of Research and Innovation in Vascular Health (GRIVAS Health), Basic Sciences Department, Faculty of Sciences, Universidad del Bio-Bio, Chillán, Chile
| | - Ángela P Cadavid J
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Grupo de Investigación en Trombosis, Departamento Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
12
|
An R, Yang Y, Liu L, Li P. SAMD1 attenuates antiphospholipid syndrome-induced vascular injury and pregnancy complications. Immun Inflamm Dis 2022; 10:e678. [PMID: 36039649 PMCID: PMC9382866 DOI: 10.1002/iid3.678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE This study was intended to investigate the effect of SAMD1 on antiphospholipid syndrome (APS)-induced vascular injury in human umbilical vein endothelial cells (HUVECs) and pregnancy complications in mice. METHODS The expression of SAMD1 in APS patients and healthy controls was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Anti-B2 GPI and anticardiolipin antibody (ACA) levels were tested by enzyme-linked immunosorbent assay, MMP-9, iNOS, ICAM-1, and MCP-1 mRNA and protein levels determined by qRT-PCR and Western blot, cellular senescence detected by β-galactosidase staining, cell proliferation ability detected by CCK-8 assay, cell viability detected by trypan blue staining, cell mobility detected by Transwell, and cell angiogenesis ability detected by matrigel tube formation assay. An APS pregnant mouse model was constructed, and the embryo absorption rate was calculated. RESULTS SAMD1 expression was low in serum of APS patients, which was correlated with the history of thrombosis and the number of adverse pregnancies. Anti-B2 GPI and ACA levels were increased in APS. The expressions of MMP-9, iNOS, ICAM-1, and MCP-1 were also significantly upregulated in HUVECs treated with APS serum. APS promoted HUVEC senescence and inhibited cell proliferation, migration, and angiogenesis. Overexpression of SAMD1 reversed the above results. Experiments on the APS pregnant mouse model confirmed that overexpression of SAMD1 reduced the rate of fetal loss. CONCLUSION SAMD1 may reduce APS-induced vascular injury and embryo loss by regulating cellular senescence, proliferation, migration, and angiogenesis.
Collapse
Affiliation(s)
- Ran An
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Yanqi Yang
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Lei Liu
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Peiling Li
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|