1
|
Murvai VR, Galiș R, Panaitescu A, Radu CM, Ghitea TC, Trif P, Onița-Avram M, Vesa AA, Huniadi A. Antiphospholipid syndrome in pregnancy: a comprehensive literature review. BMC Pregnancy Childbirth 2025; 25:337. [PMID: 40128683 PMCID: PMC11934569 DOI: 10.1186/s12884-025-07471-w] [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: 12/08/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Antiphospholipid syndrome (APS) is an autoimmune disorder associated with thrombotic events and adverse obstetric outcomes, particularly in its obstetric form (OAPS). Affecting approximately 0.5% of the population, APS is a leading contributor to recurrent pregnancy loss (RPL), preeclampsia (PE), and fetal growth restriction ((FGR). Despite advancements in understanding its pathophysiology and management, optimal treatment strategies for APS in pregnancy remain challenging and require systematic evaluation. This review synthesizes current evidence on APS mechanisms, diagnostic criteria, and therapeutic interventions, with a focus on maternal and fetal outcomes in OAPS. METHODS A comprehensive search of PubMed, was conducted to identify studies exploring APS pathogenesis, diagnostic standards, and treatment efficacy in obstetric settings. Inclusion criteria prioritized randomized controlled trials, cohort studies, and systematic reviews with a clear focus on APS and pregnancy. RESULTS The review confirmed that APS current accepted pathogenesis is governed by a "two-hit" model, where antiphospholipid antibodies (aPLs) initiate endothelial damage, culminating in thrombosis and placental insufficiency. Epidemiological analysis underscores the prevalence and severity of APS in obstetric contexts, with lupus anticoagulant (LA) emerging as a significant predictor of adverse outcomes. Evidence supports the use of low-dose aspirin (LDA) and heparin to reduce miscarriage rates, while adjunctive treatments, such as hydroxychloroquine (HCQ), have shown promise in improving live birth rates and reducing preterm delivery in high-risk cases. Emerging therapies, including tumoral necrosis factor (TNF-alpha) inhibitors and nitric oxide modulators, may offer additional benefits in refractory cases. CONCLUSION APS remains a critical determinant of adverse pregnancy outcomes, necessitating precise diagnostic criteria and tailored management approaches. This systematic review emphasizes the importance of individualized therapeutic regimens to optimize maternal and fetal health in OAPS and highlights areas for future research, particularly regarding novel pharmacological approaches. Further studies are essential to refine treatment protocols and improve clinical guidelines for managing APS in pregnancy.
Collapse
Affiliation(s)
- Viorela Romina Murvai
- Doctoral School of Biological and Biomedical Sciences, Bihor County Emergency Clinical Hospital, University of Oradea, Oradea, 410087, Romania
- Clinics Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| | - Radu Galiș
- Poznan University of Medical Sciences, Spitalul Clinic, University of Oradea, Bihor County Emergency Clinical Hospital, Poznan, Poland
| | - Anca Panaitescu
- Carol Davila University of Medicine and Pharmacy, Kings College London, University College Hospital, London, UK
| | - Casandra Maria Radu
- Clinics Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| | - Timea Claudia Ghitea
- Faculty of Medicine and Pharmacy, Pharmacy Department, University of Oradea, Oradea, Romania.
| | - Paula Trif
- Clinics Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| | - Miruna Onița-Avram
- Preclinical Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| | - Alexandra Alina Vesa
- Clinics Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| | - Anca Huniadi
- Clinics Department, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, 410073, Romania
| |
Collapse
|
2
|
Zouein J, Naim N, Spencer DM, Ortel TL. Genetic and genomic associations in antiphospholipid syndrome: A systematic review. Autoimmun Rev 2025; 24:103712. [PMID: 39617250 DOI: 10.1016/j.autrev.2024.103712] [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: 06/27/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Numerous genes have been associated with APS in the literature. In recent years, microRNA (miRNA) and long non-coding RNA (lncRNA) have also been shown to modulate the expression of APS-related genes. OBJECTIVE We performed a systematic review to identify all studies reporting on genetic mechanisms that have been shown to be associated with APS. METHODS An extensive literature search was performed in the PubMed, Cochrane and Web of Science databases gathering all available articles through February 2024. We only selected case-control studies that met inclusion criteria and that focused on genetic contributors and modifiers related to primary APS. RESULTS Sixty studies were selected for data extraction. Selected studies were grouped into 8 broad categories for review and analysis: (1) gene expression studies; (2) thrombophilia genotypes; (3) single nucleotide polymorphisms (SNPs); (4) interferon-inducible genes; (5) microRNA studies; (6) long non-coding RNA (lncRNA) studies; (7) DNA methylation studies; and (8) differential gene expression studies. Several genes have been identified as associated with APS by more than one approach, including TF, complement associated genes, and interferon-inducible genes. It has been demonstrated that miRNA and lncRNA may alter the expression of important genes in patients with APS. CONCLUSION This systematic review has helped highlight important genes implicated in APS. Most importantly, pathways such as thrombosis/hemostasis, complement and interferon appear to be involved. Further studies are needed to help uncover important genes that could serve as biomarkers.
Collapse
Affiliation(s)
- Joseph Zouein
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Nabih Naim
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University of Beirut, Lebanon
| | - Diane M Spencer
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Thomas L Ortel
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
3
|
Lu C, Gao R, Qing P, Zeng X, Liao X, Cheng M, Qin L, Liu Y. Single-cell transcriptome analyses reveal disturbed decidual homoeostasis in obstetric antiphospholipid syndrome. Ann Rheum Dis 2024; 83:624-637. [PMID: 38331588 DOI: 10.1136/ard-2023-224930] [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: 08/29/2023] [Accepted: 11/21/2023] [Indexed: 02/10/2024]
Abstract
OBJECTIVES Obstetric antiphospholipid syndrome (OAPS) is an autoimmune disease characterised by the presence of antiphospholipid antibodies in circulation and pathological pregnancy. However, the pathogenesis of OAPS remains unknown. We aimed to reveal cellular compositions and molecular features of decidual cells involved in the development of OAPS using single-cell RNA sequencing (scRNA-seq). METHODS We performed unbiased scRNA-seq analysis on the first-trimester decidua from five OAPS patients and five healthy controls (HCs), followed by validations with flow cytometry, immunohistochemical staining and immunofluorescence in a larger cohort. Serum chemokines and cytokines were measured by using ELISA. RESULTS A higher ratio of macrophages but a lower ratio of decidual natural killer (dNK) cells was found in decidua from OAPS compared with HCs. Vascular endothelial cells shrinked in OAPS decidua while having upregulated chemokine expression and conspicuous responses to IFN-γ and TNF-α. Macrophages in OAPS had stronger phagocytosis function, complement activation signals and relied more on glycolysis. dNK cells were more activated in OAPS and had enhanced cytotoxicity and IFN-γ production. Downregulation of granules in OAPS dNK cells could be associated with suppressed glycolysis. Moreover, stromal cells had a prosenescent state with weakened immune surveillance for senescent cells in OAPS. In addition, the cellular interactions between decidual immune cells and those of immune cells with non-immune cells under disease state were altered, especially through chemokines, IFN-γ and TNF-α. CONCLUSION This study provided a comprehensive decidual cell landscape and identified aberrant decidual microenvironment in OAPS, providing some potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Rui Gao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xun Zeng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Liao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of the Central Operating Unit, West China Second University Hospital, Sichuan University/West China School of Nursing, Chengdu, Sichuan, China
| | - Meng Cheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lang Qin
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Raschi E, Borghi MO, Tedesco F, Meroni PL. Antiphospholipid syndrome pathogenesis in 2023: an update of new mechanisms or just a reconsideration of the old ones? Rheumatology (Oxford) 2024; 63:SI4-SI13. [PMID: 38320591 DOI: 10.1093/rheumatology/kead603] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/27/2023] [Indexed: 02/08/2024] Open
Abstract
Antibodies against phospholipid (aPL)-binding proteins, in particular, beta 2 glycoprotein I (β2GPI), are diagnostic/classification and pathogenic antibodies in antiphospholipid syndrome (APS). β2GPI-aPL recognize their target on endothelium and trigger a pro-thrombotic phenotype which is amplified by circulating monocytes, platelets and neutrophils. Complement activation is required as supported by the lack of aPL-mediated effects in animal models when the complement cascade is blocked. The final result is a localized clot. A strong generalized inflammatory response is associated with catastrophic APS, the clinical variant characterized by systemic thrombotic microangiopathy. A two-hit hypothesis was suggested to explain why persistent aPL are associated with acute events only when a second hit allows antibody/complement binding by modulating β2GPI tissue presentation. β2GPI/β2GPI-aPL are also responsible for obstetric APS, being the molecule physiologically present in placental/decidual tissues. Additional mechanisms mediated by aPL with different characteristics have been reported, but their diagnostic/prognostic value is still a matter of research.
Collapse
Affiliation(s)
- Elena Raschi
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Maria Orietta Borghi
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Francesco Tedesco
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Pier Luigi Meroni
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Vrzić Petronijević S, Vilotić A, Bojić-Trbojević Ž, Kostić S, Petronijević M, Vićovac L, Jovanović Krivokuća M. Trophoblast Cell Function in the Antiphospholipid Syndrome. Biomedicines 2023; 11:2681. [PMID: 37893055 PMCID: PMC10604227 DOI: 10.3390/biomedicines11102681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Antiphospholipid syndrome (APS) is a complex thrombo-inflammatory autoimmune disease characterized by the presence of antiphospholipid antibodies (aPL). Women with APS are at high risk of recurrent early pregnancy loss as well as late obstetrical complications-premature birth due to placental insufficiency or severe preeclampsia. Accumulating evidence implies that vascular thrombosis is not the only pathogenic mechanism in obstetric APS, and that the direct negative effect of aPL on the placental cells, trophoblast, plays a major role. In this review, we summarize the current findings regarding the potential mechanisms involved in aPL-induced trophoblast dysfunction. Introduction on the APS and aPL is followed by an overview of the effects of aPL on trophoblast-survival, cell function and aPL internalization. Finally, the implication of several non-coding RNAs in pathogenesis of obstetric APS is discussed, with special emphasis of their possible role in trophoblast dysfunction and the associated mechanisms.
Collapse
Affiliation(s)
- Svetlana Vrzić Petronijević
- University of Belgrade, Faculty of Medicine, University Clinical Center of Serbia Clinic for Obstetrics and Gynecology, Koste Todorovića 26, 11000 Belgrade, Serbia
| | - Aleksandra Vilotić
- University of Belgrade, Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, Banatska 31b, 11080 Belgrade, Serbia
| | - Žanka Bojić-Trbojević
- University of Belgrade, Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, Banatska 31b, 11080 Belgrade, Serbia
| | - Sanja Kostić
- University of Belgrade, Faculty of Medicine, University Clinical Center of Serbia Clinic for Obstetrics and Gynecology, Koste Todorovića 26, 11000 Belgrade, Serbia
| | - Miloš Petronijević
- University of Belgrade, Faculty of Medicine, University Clinical Center of Serbia Clinic for Obstetrics and Gynecology, Koste Todorovića 26, 11000 Belgrade, Serbia
| | - Ljiljana Vićovac
- University of Belgrade, Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, Banatska 31b, 11080 Belgrade, Serbia
| | - Milica Jovanović Krivokuća
- University of Belgrade, Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, Banatska 31b, 11080 Belgrade, Serbia
| |
Collapse
|
8
|
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
|
9
|
Verrou KM, Sfikakis PP, Tektonidou MG. Whole blood transcriptome identifies interferon-regulated genes as key drivers in thrombotic primary antiphospholipid syndrome. J Autoimmun 2023; 134:102978. [PMID: 36587511 DOI: 10.1016/j.jaut.2022.102978] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Pathogenesis of antiphospholipid syndrome (APS) isn't fully elucidated. We aimed to identify gene signatures characterizing thrombotic primary APS (thrPAPS) and subgroups at high risk for worse outcomes. METHODS We performed whole blood next-generation RNA-sequencing in 62 patients with thrPAPS and 29 age-/sex-matched healthy controls (HCs), followed by differential gene expression analysis (DGEA) and enrichment analysis. We trained models on transcriptomics data using machine learning. RESULTS DGEA of 12.306 genes revealed 34 deregulated genes in thrPAPS versus HCs; 33 were upregulated by at least 2-fold, and 14/33 were type I and II interferon-regulated genes (IRGs) as determined by interferome database. Machine learning applied to deregulated genes returned 79% accuracy to discriminate thrPAPS from HCs, which increased to 82% when only the most informative IRGs were analyzed. Comparison of thrPAPS subgroups versus HCs showed an increased presence of IRGs among upregulated genes in venous thrombosis (21/23, 91%), triple-antiphospholipid antibody (aPL) positive (30/50, 60%), and recurrent thrombosis (19/42, 45%) subgroups. Enrichment analysis of upregulated genes in triple-aPL positive patients revealed terms related to 'type I interferon signaling pathway' and 'innate immune response'. DGEA among thrPAPS subgroups revealed upregulated genes, including IRGs, in patients with venous versus arterial thrombosis (n = 11, 9 IRGs), triple-aPL versus non-triple aPL (n = 10, 9 IRGs), and recurrent versus non-recurrent thrombosis (n = 10, 3 IRGs). CONCLUSION Upregulated IRGs may better discriminate thrPAPS from HCs than all deregulated genes in peripheral blood. Taken together with DGEA data, IRGs are highly expressed in thrPAPS and high-risk subgroups of triple-aPL and recurrent thrombosis, with potential treatment implications.
Collapse
Affiliation(s)
- Kleio-Maria Verrou
- Center of New Biotechnologies & Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros P Sfikakis
- Center of New Biotechnologies & Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria G Tektonidou
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
10
|
Jacintho BC, Mazetto Fonseca BDM, Hounkpe BW, Oliveira JD, dos Santos APR, Vaz CDO, de Paula EV, Orsi FA. Evaluation of a gene signature related to thrombotic manifestations in antiphospholipid syndrome. Front Med (Lausanne) 2023; 10:1139906. [PMID: 37035297 PMCID: PMC10076702 DOI: 10.3389/fmed.2023.1139906] [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: 01/07/2023] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Thrombotic primary antiphospholipid syndrome (t-PAPS) is an acquired condition characterized by heterogeneous thrombotic manifestations, which is intriguing since venous and arterial thrombosis appear to have distinct pathogenesis. Gene expression analysis may constitute a new approach to evaluate potential similarities or differences between the clinical manifestations of t-PAPS. Recently, dysregulation of the ANXA3, TNFAIP6, TXK, BACH2, and SERPINB2 genes has been associated with both arterial and venous thrombosis in the general population. Therefore, the aim of this study was to examine whether ANXA3, TNFAIP6, TXK, BACH2, and SERPINB2 expression was associated with t-PAPS. Gene expression was quantified by qPCR of total leukocyte mRNA. In this case-control study, 102 t-PAPS patients, 17 asymptomatic antiphospholipid (aPL) carriers and 100 controls were evaluated. Increased expression of ANXA3 (P = 0.008) and TNFAIP6 (P = 0.001) and decreased expression of the TXK gene (P = 0.0001) were associated with an increased risk of t-PAPS compared to the control. ANXA3 upregulation was more evident in cases of arterial thrombosis and multiple thrombotic events. There was no difference in the expression of these genes between triple and non-triple aPL positivity. ANXA3, TNFAIP6, TXK, BACH2, and SERPINB2 expression levels were also similar between aPL carriers and controls (P = 0.77; P = 0.48; P = 0.08; P = 0.73, and P = 0.13, respectively). In conclusion, our results showed that genes related to hemostasis (ANXA3) and immunity (TNFAIP6, TXK) are dysregulated in t-PAPS compared to controls. Gene dysregulation was not detected in aPL carriers and was not related to the aPL profile, suggesting that this gene signature is related to thrombotic manifestations rather than to aPL burden. Our results suggest that innate immunity and hemostasis pathways are associated with t-PAPS at a molecular level and may play a role in disease severity.
Collapse
Affiliation(s)
| | - Bruna de Moraes Mazetto Fonseca
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Hematology and Hemotherapy Center, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Jose Diogo Oliveira
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | | | - Erich Vinicius de Paula
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Hematology and Hemotherapy Center, University of Campinas (UNICAMP), Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- *Correspondence: Fernanda Andrade Orsi,
| |
Collapse
|
11
|
Huang Z, Tang Z, Guan H, Leung W, Wang L, Xia H, Zhang W. Inactivation of Yes-Associated Protein Mediates Trophoblast Dysfunction: A New Mechanism of Pregnancy Loss Associated with Anti-Phospholipid Antibodies? Biomedicines 2022; 10:biomedicines10123296. [PMID: 36552052 PMCID: PMC9776042 DOI: 10.3390/biomedicines10123296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Pregnancy morbidity induced by anti-phospholipid antibodies (aPL+/PM+) is mainly thought to arise from placental abnormalities. We attempted to investigate the effect of aPL on the activity of Yes-associated protein (YAP) in the trophoblast and how YAP regulated human trophoblasts function. Thus, HTR-8 cells were treated with IgG purified from aPL+/PM+ women or normal controls. We found that aPL+/PM+ IgG impacted YAP activity via abrogating YAP expression. Further investigation of the anti-β2GPI-IgG/β2GPI complex showed an inhibition of nuclear YAP level and translocation in a dose-dependent manner, which might be rescued by progesterone in HTR-8 cells. YAP overexpression or knockdown HTR-8 cells were established for the evaluation of cell function and related gene expression in vitro. Loss of YAP arrested cell cycles in the G2/M phase, accelerated cell apoptosis by increasing the ratio of Bax/Bcl2, and disrupted MMP2/9-mediated cell migration and angiogenesis tube formation by VEGF. These findings support a new mechanism of PM associated with aPL through which YAP inactivation induced by aPL perturbs the trophoblast cell cycle, apoptosis, migration, and angiogenesis, finally developing into pregnancy failure.
Collapse
Affiliation(s)
- Zengshu Huang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Zhijing Tang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Haiyun Guan
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Wingting Leung
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Lu Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Hexia Xia
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Wei Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Correspondence:
| |
Collapse
|
12
|
Alijotas-Reig J, Esteve-Valverde E, Anunciación-Llunell A, Marques-Soares J, Pardos-Gea J, Miró-Mur F. Pathogenesis, Diagnosis and Management of Obstetric Antiphospholipid Syndrome: A Comprehensive Review. J Clin Med 2022; 11:675. [PMID: 35160128 PMCID: PMC8836886 DOI: 10.3390/jcm11030675] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Antiphospholipid syndrome is an autoimmune disorder characterized by vascular thrombosis and/or pregnancy morbidity associated with persistent antiphospholipid antibody positivity. Cases fulfilling the Sydney criteria for obstetric morbidity with no previous thrombosis are known as obstetric antiphospholipid syndrome (OAPS). OAPS is the most identified cause of recurrent pregnancy loss and late-pregnancy morbidity related to placental injury. Cases with incomplete clinical or laboratory data are classified as obstetric morbidity APS (OMAPS) and non-criteria OAPS (NC-OAPS), respectively. Inflammatory and thrombotic mechanisms are involved in the pathophysiology of OAPS. Trophoblasts, endothelium, platelets and innate immune cells are key cellular players. Complement activation plays a crucial pathogenic role. Secondary placental thrombosis appears by clot formation in response to tissue factor activation. New risk assessment tools could improve the prediction of obstetric complication recurrences or thromboses. The standard-of-care treatment consists of low-dose aspirin and prophylactic low molecular weight heparin. In refractory cases, the addition of hydroxychloroquine, low-dose prednisone or IVIG improve pregnancy outcomes. Statins and eculizumab are currently being tested for treating selected OAPS women. Finally, we revisited recent insights and concerns about the pathophysiology, diagnosis and management of OAPS.
Collapse
Affiliation(s)
- Jaume Alijotas-Reig
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Enrique Esteve-Valverde
- Department of Internal Medicine, Althaia Xarxa Assistencial, Carrer Dr Joan Soler 1-3, 08243 Manresa, Spain;
| | - Ariadna Anunciación-Llunell
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
| | - Joana Marques-Soares
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Josep Pardos-Gea
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Francesc Miró-Mur
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
| |
Collapse
|
13
|
Rodríguez CM, Velásquez-Berrío M, Rúa C, Viana M, Abrahams VM, Cadavid AP, Alvarez AM. Antiphospholipid Antibodies From Women With Pregnancy Morbidity and Vascular Thrombosis Induce Endothelial Mitochondrial Dysfunction, mTOR Activation, and Autophagy. Front Physiol 2021; 12:706743. [PMID: 34912234 PMCID: PMC8667788 DOI: 10.3389/fphys.2021.706743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by thrombosis and pregnancy morbidity (PM) obstetric events together with persistent high titers of circulating antiphospholipid antibodies (aPL). Several mechanisms that explain the development of thrombosis and PM in APS include the association of aPL with alterations in the coagulation cascade and inflammatory events. Other mechanisms disturbing cellular homeostases, such as mitochondrial dysfunction, autophagy, and cell proliferation, have been described in other autoimmune diseases. Therefore, the objective of this study was to investigate the impact of aPL from different patient populations on endothelial cell mitochondrial function, activation of the mammalian target of rapamycin (mTOR) and autophagy pathways, and cellular growth. Using an in vitro model, human umbilical vein endothelial cells (HUVECs) were treated with polyclonal immunoglobulin G (IgG) purified from the serum of women with both PM and vascular thrombosis (PM/VT), with VT only (VT), or with PM and non-criteria aPL (seronegative-obstetric APS, SN-OAPS). We included IgG from women with PM without aPL (PM/aPL-) and healthy women with previous uncomplicated pregnancies (normal human serum, NHS) as control groups. Mitochondrial function, mTOR activation, autophagy, and cell proliferation were evaluated by Western blotting, flow cytometry, and functional assays. IgG from women with PM/VT increased HUVEC mitochondrial hyperpolarization and activation of the mTOR and autophagic pathways, while IgG from patients with VT induced endothelial autophagy and cell proliferation in the absence of elevated mTOR activity or mitochondrial dysfunction. IgG from the SN-OAPS patient group had no effect on any of these HUVEC responses. In conclusion, aPL from women with PM and vascular events induce cellular stress evidenced by mitochondrial hyperpolarization and increased activation of the mTOR and autophagic pathways which may play a role in the pathogenesis of obstetric APS.
Collapse
Affiliation(s)
- Carlos M. Rodríguez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Manuela Velásquez-Berrío
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Carolina Rúa
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Marta Viana
- Grupo de Metabolismo y Función Vascular, Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Angela P. Cadavid
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Angela M. Alvarez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| |
Collapse
|
14
|
Proteomics and enriched biological processes in Antiphospholipid syndrome: A systematic review. Autoimmun Rev 2021; 20:102982. [PMID: 34718168 DOI: 10.1016/j.autrev.2021.102982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022]
Abstract
Identification of differentially expressed proteins in antiphospholipid syndrome (APS) is a developing area of research for unique profiles of this pathology. Advances in technologies of mass spectrometry brings improvements in proteomics and results in assessment of soluble or cellular proteins which could be candidates for clinical biomarkers of primary APS. The use of blood as a source of proteins ease the acquisition of samples for proteomics analyses and later for disease diagnosis. We performed a systematic review to explore the proteomics studies carried out in circulating released proteins (serum, plasma) or cellular proteins (monocytes and platelets) of APS patients. The study groups differentiate among clinical APS cases with the aim to translate molecular findings to disease stratification and to improve APS diagnosis and prognosis. These studies also include the unravelling of new autoantibodies in non-criteria APS or how post-translational protein modifications provides clues about the pathological mechanisms of antigen-autoantibody recognition. Herein, we identified 82 proteins that were dysregulated in APS across eleven studies. Enrichment analysis revealed its connection to cellular activation and degranulation that eventually leads to thrombosis as the main biological process highlighted by these studies. Validation of APS-relevant proteins by functional and mechanistic studies will be essential for patient stratification and the development of targeted therapies for every clinical subtype of APS.
Collapse
|
15
|
Zhou Z, You Y, Wang F, Sun Y, Teng J, Liu H, Cheng X, Su Y, Shi H, Hu Q, Chi H, Jia J, Wan L, Liu T, Wang M, Shi C, Yang C, Ye J. Urine Proteomics Differentiate Primary Thrombotic Antiphospholipid Syndrome From Obstetric Antiphospholipid Syndrome. Front Immunol 2021; 12:702425. [PMID: 34489952 PMCID: PMC8416615 DOI: 10.3389/fimmu.2021.702425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Antiphospholipid syndrome (APS) is a multisystem disorder characterized by thrombosis and/or recurrent fetal loss. This clinical phenotype heterogeneity may result in differences in response to treatment and prognosis. In this study, we aimed to identify primary thrombotic APS (TAPS) from primary obstetric APS (OAPS) using urine proteomics as a non-invasive method. Only patients with primary APS were enrolled in this study from 2016 to 2018 at a single clinical center in Shanghai. Urine samples from 15 patients with TAPS, 9 patients with OAPS, and 15 healthy controls (HCs) were collected and analyzed using isobaric tags for relative and absolute quantification (iTRAQ) labeling combined with liquid chromatography-tandem mass spectrometry analysis to identify differentially expressed proteins. Cluster analysis of urine proteomics identified differentiated proteins among the TAPS, OAPS, and HC groups. Urinary proteins were enriched in cytokine and cytokine receptor pathways. Representative secreted cytokines screened out (fold change >1.20, or <0.83, p<0.05) in these differentiated proteins were measured by enzyme-linked immunosorbent assay in a validation cohort. The results showed that the levels of C-X-C motif chemokine ligand 12 (CXCL12) were higher in the urine of patients with TAPS than in those with OAPS (p=0.035), while the levels of platelet-derived growth factor subunit B (PDGFB) were lower in patients with TAPS than in those with OAPS (p=0.041). In addition, correlation analysis showed that CXCL12 levels were positively correlated with immunoglobulin G anti-β2-glycoprotein I antibody (r=0.617, p=0.016). Our results demonstrated that urinary CXCL12 and PDGFB might serve as potential non-invasive markers to differentiate primary TAPS from primary OAPS.
Collapse
Affiliation(s)
- Zhuochao Zhou
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijun You
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Sun
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Teng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglei Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Su
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiongyi Hu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihui Chi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinchao Jia
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liyan Wan
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ce Shi
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Goulielmos GN, Zervou MI. Comment on: Obstetric antiphospholipid syndrome is not associated with an increased risk of subclinical atherosclerosis. Rheumatology (Oxford) 2021; 60:e220-e221. [PMID: 33555317 DOI: 10.1093/rheumatology/keab110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- George N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece.,Department of Internal Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Maria I Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
17
|
Pérez-Sánchez L, Patiño-Trives AM, Aguirre-Zamorano MÁ, Luque-Tévar M, Ábalos-Aguilera MC, Arias-de la Rosa I, Seguí P, Velasco-Gimena F, Barbarroja N, Escudero-Contreras A, Collantes-Estévez E, Pérez-Sánchez C, López-Pedrera C. Characterization of Antiphospholipid Syndrome Atherothrombotic Risk by Unsupervised Integrated Transcriptomic Analyses. Arterioscler Thromb Vasc Biol 2020; 41:865-877. [PMID: 33356391 DOI: 10.1161/atvbaha.120.315346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Our aim was to characterize distinctive clinical antiphospholipid syndrome phenotypes and identify novel microRNA (miRNA)-mRNA-intracellular signaling regulatory networks in monocytes linked to cardiovascular disease. Approach and Results: Microarray analysis in antiphospholipid syndrome monocytes revealed 547 differentially expressed genes, mainly involved in inflammatory, cardiovascular, and reproductive disorders. Besides, this approach identified several genes related to inflammatory, renal, and dermatologic diseases. Functional analyses further demonstrated phosphorylation of intracellular kinases related to thrombosis and immune-mediated chronic inflammation. miRNA profiling showed altered expression of 22 miRNAs, enriched in pathways related to immune functions, cardiovascular disease, and autoimmune-associated pathologies. Unbiased integrated mRNA-miRNA analysis identified a signature of 9 miRNAs as potential modulators of 17 interconnected genes related to cardiovascular disease. The altered expression of that miRNA-mRNA signature was proven to be stable along time and distinctive of nonautoimmune thrombotic patients. Transfection studies and luciferase assays established the relationship between specific miRNAs and their identified target genes and proteins, along with their involvement in the regulation of monocytes procoagulant activity and cell adhesion. Correlation analyses showed relationship among altered miRNAs and their interconnected genes with aPL (antiphospholipid antibodies)-titers, along with microvascular endothelial dysfunction. In vitro studies demonstrated modulation in healthy monocytes by IgG-aPLs of several genes/miRNAs, which further intermediated downstream effects on endothelial function. The identified transcriptomic signature allowed the unsupervised division of three clusters of patients with antiphospholipid syndrome showing distinctive clinical profiles, mainly associated with their prothrombotic risk (thrombosis, autoantibody profile, cardiovascular risk factors, and atherosclerosis). CONCLUSIONS Extensive molecular profiling of monocytes in patients with primary antiphospholipid syndrome might help to identify distinctive clinical phenotypes, thus enabling new patients' tailored treatments.
Collapse
Affiliation(s)
- Laura Pérez-Sánchez
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Alejandra M Patiño-Trives
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - M Ángeles Aguirre-Zamorano
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - María Luque-Tévar
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - M Carmen Ábalos-Aguilera
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Iván Arias-de la Rosa
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Pedro Seguí
- Radiology Service (P.S.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Francisco Velasco-Gimena
- Haematology Service (F.V.-G.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Nuria Barbarroja
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain (N.B.)
| | - Alejandro Escudero-Contreras
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Eduardo Collantes-Estévez
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| | - Carlos Pérez-Sánchez
- Deparment of Medicine, University of Cambridge, School of Clinical Medicine, Addenbroke's Hospital, Cambridge Institute for Medical Research, United Kingdom (C.P.-S.)
| | - Chary López-Pedrera
- Rheumatology Service (L.P.-S., A.M.P.-T., M.A.A.-Z., M.L.-T., M.C.A.-A., I.A.-d.l.R., N.B., A.E.-C., E.C.-E., C.L.-P.), Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC)/University of Cordoba, Spain
| |
Collapse
|
18
|
Abstract
Antiphospholipid syndrome is one of the more common acquired causes of hypercoagulability. Its major presentations are thrombotic (arterial, venous, or microvascular) and pregnancy morbidity (miscarriages, late intrauterine fetal demise, and severe pre-eclampsia). Classification criteria include 3 different antiphospholipid antibodies: lupus anticoagulant, anticardiolipin, and anti-beta 2 glycoprotein I. Management includes both preventive strategies (low-dose aspirin, hydroxychloroquine) and long-term anticoagulation after thrombosis.
Collapse
Affiliation(s)
- Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Antiphospholipid syndrome (APS) is a thrombo-inflammatory disease that is primarily treated with anticoagulation. Better understanding the inflammatory aspects of APS could lead to safer, more effective, and more personalized therapeutic options. To this end, we sought to understand recent literature related to the role of neutrophils and, in particular, neutrophil extracellular traps (NETs) in APS. RECENT FINDINGS Expression of genes associated with type I interferons, endothelial adhesion, and pregnancy regulation are increased in APS neutrophils. APS neutrophils have a reduced threshold for NET release, which likely potentiates thrombotic events and perhaps especially large-vein thrombosis. Neutrophil-derived reactive oxygen species also appear to play a role in APS pathogenesis. There are new approaches for preventing and disrupting NETs that could potentially be leveraged to reduce the risk of APS-associated thrombosis. Neutrophils and NETs contribute to APS pathophysiology. More precisely understanding their roles at a mechanistic level should help identify new therapeutic targets for inhibiting NET formation, enhancing NET dissolution, and altering neutrophil adhesion. Such approaches may ultimately lead to better clinical management of APS patients and thereby reduce the chronic burden of this disease.
Collapse
|
20
|
Zuily S, Clerc-Urmès I, Bauman C, Andrade D, Sciascia S, Pengo V, Tektonidou MG, Ugarte A, Gerosa M, Michael Belmont H, Zamorano MAA, Fortin P, Ji L, Efthymiou M, Cohen H, Branch DW, Jesus GRD, Nalli C, Petri M, Rodriguez E, Cervera R, Knight JS, Atsumi T, Willis R, Bertolaccini ML, Vega J, Wahl D, Erkan D. Cluster analysis for the identification of clinical phenotypes among antiphospholipid antibody-positive patients from the APS ACTION Registry. Lupus 2020:961203320940776. [PMID: 32703117 DOI: 10.1177/0961203320940776] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study aimed to use cluster analysis (CA) to identify different clinical phenotypes among antiphospholipid antibodies (aPL)-positive patients. METHODS The Alliance for Clinical Trials and International Networking (APS ACTION) Registry includes persistently positive aPL of any isotype based on the Sydney antiphospholipid syndrome (APS) classification criteria. We performed CA on the baseline characteristics collected retrospectively at the time of the registry entry of the first 500 patients included in the registry. A total of 30 clinical data points were included in the primary CA to cover the broad spectrum of aPL-positive patients. RESULTS A total of 497 patients from international centres were analysed, resulting in three main exclusive clusters: (a) female patients with no other autoimmune diseases but with venous thromboembolism (VTE) and triple-aPL positivity; (b) female patients with systemic lupus erythematosus, VTE, aPL nephropathy, thrombocytopaenia, haemolytic anaemia and a positive lupus anticoagulant test; and (c) older men with arterial thrombosis, heart valve disease, livedo, skin ulcers, neurological manifestations and cardiovascular disease (CVD) risk factors. CONCLUSIONS Based on our hierarchical cluster analysis, we identified different clinical phenotypes of aPL-positive patients discriminated by aPL profile, lupus or CVD risk factors. Our results, while supporting the heterogeneity of aPL-positive patients, also provide a foundation to understand disease mechanisms, create new approaches for APS classification and ultimately develop new management approaches.
Collapse
Affiliation(s)
- Stéphane Zuily
- Vascular Medicine Division and Regional Competence Centre for Systemic And Autoimmune Diseases, Nancy Academic Hospital, Nancy, France.,Inserm UMR_S 1116, Lorraine University, Nancy, France
| | - Isabelle Clerc-Urmès
- ESPRI-BioBase, Platform of Clinical Research Support PARC (MDS unity), Nancy Academic Hospital, Nancy, France
| | - Cédric Bauman
- ESPRI-BioBase, Platform of Clinical Research Support PARC (MDS unity), Nancy Academic Hospital, Nancy, France
| | - Danieli Andrade
- Department of Rheumatology, Faculty of Medicine, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Savino Sciascia
- Centre of Research of Immunopathology and Rare Diseases, University of Turin, Turin, Italy
| | - Vittorio Pengo
- Thrombosis Research Laboratory, Department of Cardiac Thoracic and Vascular Sciences, and Public Health, University of Padova; Arianna Foundation on Anticoagulation, Bologna, Italy
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Amaia Ugarte
- Autoimmune Diseases Research Unit, Department of Internal Medicine, Hospital Universitario Cruces, Barakaldo, Spain
| | - Maria Gerosa
- Clinical Rheumatology Unit, Research Center for Adult and Pediatric Diseases, Department of Clinical Sciences and Community Health, ASST Pini-CTO, University of Milan, Milan, Italy
| | | | | | - Paul Fortin
- CHU de Quebec - Université Laval, Quebec, Canada
| | - Lanlan Ji
- Rheumatology and Immunology Department, Peking University First Hospital, Beijing, PR China
| | - Maria Efthymiou
- Haemostasis Research Unit, Department of Haematology, University College London, London, UK
| | - Hannah Cohen
- Haemostasis Research Unit, Department of Haematology, University College London, London, UK
| | - D Ware Branch
- University of Utah and Intermountain Healthcare, Salt Lake City, USA
| | - Guilherme Ramires de Jesus
- Departamento de Obstetrícia, Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cecilia Nalli
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Michelle Petri
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Ricard Cervera
- Department of Autoimmune Diseases, Hospital Clínic Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, USA
| | | | - Rohan Willis
- Antiphospholipid Standardization Laboratory, University of Texas Medical Branch, Galveston, USA
| | | | - Joann Vega
- Barbara Volcker Centre for Women and Rheumatic Disease, Hospital for Special Surgery, Weill Cornell Medicine, New York, USA
| | - Denis Wahl
- Vascular Medicine Division and Regional Competence Centre for Systemic And Autoimmune Diseases, Nancy Academic Hospital, Nancy, France.,Inserm UMR_S 1116, Lorraine University, Nancy, France
| | - Doruk Erkan
- Barbara Volcker Centre for Women and Rheumatic Disease, Hospital for Special Surgery, Weill Cornell Medicine, New York, USA
| | | |
Collapse
|
21
|
McDonnell T, Wincup C, Buchholz I, Pericleous C, Giles I, Ripoll V, Cohen H, Delcea M, Rahman A. The role of beta-2-glycoprotein I in health and disease associating structure with function: More than just APS. Blood Rev 2020; 39:100610. [PMID: 31471128 PMCID: PMC7014586 DOI: 10.1016/j.blre.2019.100610] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/22/2022]
Abstract
Beta-2-Glycoprotein I (β2GPI) plays a number of essential roles throughout the body. β2GPI, C-reactive protein and thrombomodulin are the only three proteins that possess the dual capability to up and down regulate the complement and coagulation systems depending upon external stimulus. Clinically, β2GPI is the primary antigen in the autoimmune condition antiphospholipid syndrome (APS), which is typically characterised by pregnancy morbidity and vascular thrombosis. This protein is also capable of adopting at least two distinct structural forms, but it has been argued that several other intermediate forms may exist. Thus, β2GPI is a unique protein with a key role in haemostasis, homeostasis and immunity. In this review, we examine the genetics, structure and function of β2GPI in the body and how these factors may influence its contribution to disease pathogenesis. We also consider the clinical implications of β2GPI in the diagnosis of APS and as a potentially novel therapeutic target.
Collapse
Affiliation(s)
- Thomas McDonnell
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK.
| | - Chris Wincup
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Ina Buchholz
- Nanostructure Group, Institute of Biochemistry, University of Greifswald, Germany
| | - Charis Pericleous
- Imperial College London, Imperial College Vascular Sciences, National Heart & Lung Institute, ICTEM, Hammersmith Campus, Du Cane Road, London, UK
| | - Ian Giles
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Vera Ripoll
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Hannah Cohen
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Mihaela Delcea
- Nanostructure Group, Institute of Biochemistry, University of Greifswald, Germany
| | - Anisur Rahman
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| |
Collapse
|
22
|
Oxidative stress in endothelial cells induced by the serum of women with different clinical manifestations of the antiphospholipid syndrome. ACTA ACUST UNITED AC 2019; 39:673-688. [PMID: 31860179 PMCID: PMC7363350 DOI: 10.7705/biomedica.4701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Indexed: 12/27/2022]
Abstract
Introducción. El síndrome antifosfolípido se caracteriza por la presencia persistente de anticuerpos antifosfolípidos y manifestaciones clínicas de trombosis o morbilidad gestacional, las cuales se asocian con estrés oxidativo y disfunción endotelial. Objetivo. Evaluar los marcadores de estrés oxidativo en células endoteliales, inducidos por el suero de mujeres con diferentes manifestaciones clínicas del síndrome antifosfolípido y analizar la capacidad antioxidante de los sueros. Materiales y métodos. Se incluyeron 48 mujeres que fueron clasificadas así: presencia de anticuerpos antifosfolípidos y criterios clínicos de morbilidad gestacional, trombosis vascular o ambas. Como grupos control se incluyeron mujeres negativas para anticuerpos antifosfolípidos. En un modelo in vitro de células endoteliales estimuladas con los sueros de las mujeres del estudio, se determinaron algunos marcadores de estrés oxidativo por citometría de flujo. También, se analizó la capacidad antioxidante de los sueros incluidos. Resultados. Los sueros de los grupos de mujeres con síndrome antifosfolípido que presentaban trombosis, con morbilidad gestacional o sin ella, generaron un incremento significativo (p<0,05 y p<0,001) en los marcadores de estrés oxidativo endotelial, en contraste con el control de suero humano normal. No se observaron diferencias en el efecto de los sueros de los diferentes grupos de estudio sobre la lipoperoxidación endotelial. Tampoco se encontró diferencia en la actividad antioxidante de los sueros. Conclusión. El estrés oxidativo mitocondrial en el endotelio se asocia con la presencia de trombosis. Sin embargo, cuando esta se asocia con morbilidad gestacional, también se genera estrés oxidativo intracelular.
Collapse
|
23
|
Lopez-Pedrera C, Barbarroja N, Patiño-Trives AM, Collantes E, Aguirre MA, Perez-Sanchez C. New Biomarkers for Atherothrombosis in Antiphospholipid Syndrome: Genomics and Epigenetics Approaches. Front Immunol 2019; 10:764. [PMID: 31040845 PMCID: PMC6476988 DOI: 10.3389/fimmu.2019.00764] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/22/2019] [Indexed: 01/08/2023] Open
Abstract
Antiphospholipid Syndrome (APS) is an autoimmune disorder, characterized by pregnancy morbidity and/or a hyper coagulable state involving the venous or the arterial vasculature and associated with antiphospholipid antibodies (aPL), including anti-cardiolipin antibodies (aCL), anti-beta2-glycoprotein I (anti-ß2GPI), and Lupus anticoagulant (LA). In recent years there have been many advances in the understanding of the molecular basis of vascular involvement in APS. APS is of multifactorial origin and develops in genetically predisposed individuals. The susceptibility is determined by major histocompatibility complex (MHC). Different HLA-DR and HLA-DQ alleles have been reported in association with APS. Moreover, MHC II alleles may determine the autoantibody profile and, as such, the clinical phenotype of this disease. Besides, polymorphisms in genes related to the vascular system are considered relevant factors predisposing to clinical manifestations. Antiphospholipid antibodies (aPL) induce genomic and epigenetic alterations that support a pro- thrombotic state. Thus, a specific gene profile has been identified in monocytes from APS patients -related to aPL titres in vivo and promoted in vitro by aPL- explaining their cardiovascular involvement. Regarding epigenetic approaches, we previously recognized two miRNAs (miR-19b/miR-20a) as potential modulators of tissue factor, the main receptor involved in thrombosis development in APS. aPLs can further promote changes in the expression of miRNA biogenesis proteins in leukocytes of APS patients, which are translated into an altered miRNA profile and, consequently, in the altered expression of their protein targets related to thrombosis and atherosclerosis. MicroRNAs are further released into the circulation, acting as intercellular communicators. Accordingly, a specific signature of circulating miRNAs has been recently identified in APS patients as potential biomarkers of clinical features. Genomics and epigenetic biomarkers might also serve as indices for disease progression, clinical pharmacology, or safety, so that they might be used to individually predict disease outcome and guide therapeutic decisions. In that way, in the setting of a clinical trial, novel and specific microRNA–mRNA regulatory networks in APS, modified by effect of Ubiquinol treatment, have been identified. In this review, current and previous studies analyzing genomic/epigenetic changes related to the clinical profile of APS patients, and their modulation by effect of specific therapies, are discussed.
Collapse
Affiliation(s)
- Chary Lopez-Pedrera
- Instituto Maimonides de Investigación Biomédica de Cordoba, Reina Sofia Hospital, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| | - Nuria Barbarroja
- Hospital Universitario Reina Sofía, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| | - Alejandra Mª Patiño-Trives
- Instituto Maimonides de Investigación Biomédica de Cordoba, Reina Sofia Hospital, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| | - Eduardo Collantes
- Hospital Universitario Reina Sofía, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| | - Mª Angeles Aguirre
- Hospital Universitario Reina Sofía, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| | - Carlos Perez-Sanchez
- Hospital Universitario Reina Sofía, Córdoba, Spain.,Inflammatory and Systemic Autoimmune Diseases' Group, Instituto Maimonides de Investigacion Biomédica de Córdoba, Cordova, Spain.,Department of Medicine, Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
24
|
Lonati PA, Scavone M, Gerosa M, Borghi MO, Pregnolato F, Curreli D, Podda G, Femia EA, Barcellini W, Cattaneo M, Tedesco F, Meroni PL. Blood Cell-Bound C4d as a Marker of Complement Activation in Patients With the Antiphospholipid Syndrome. Front Immunol 2019; 10:773. [PMID: 31031764 PMCID: PMC6474283 DOI: 10.3389/fimmu.2019.00773] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/25/2019] [Indexed: 01/22/2023] Open
Abstract
Antiphospholipid syndrome (APS) is a chronic and disabling condition characterized by recurrent thrombosis and miscarriages mediated by antibodies against phospholipid-binding proteins (aPL), such as beta2glycoprotein I (β2GPI). Complement is involved in APS animal models and complement deposits have been documented in placenta and thrombotic vessels despite normal serum levels. Analysis of circulating blood cells coated with C4d displays higher sensitivity than the conventional assays that measure soluble native complement components and their unstable activation products in systemic lupus erythematosus (SLE). As C4d-coated blood cell count has been reported to be more sensitive than serum levels of complement components and their activation products in systemic lupus erythematosus (SLE) patients, we decided to evaluate the percentage of C4d positive B lymphocytes (BC4d), erythrocytes (EC4d), and platelets (PC4d) in primary APS patients and asymptomatic aPL positive carriers as marker of complement activation in APS. We assessed by flow cytometry the percentages of BC4d, EC4d, and PC4d in primary APS (PAPS; n. 23), 8 asymptomatic aPL positive carriers, 11 APS-associated SLE (SAPS), 17 aPL positive SLE, 16 aPL negative SLE, 8 aPL negative patients with previous thrombosis, 11 immune thrombocytopenia (ITP) patients, and 26 healthy subjects. In addition, we used an in vitro model to evaluate the ability of a monoclonal anti-β2GPI antibody (MBB2) to bind to normal resting or activated platelets and fix complement. EC4d and PC4d percentages were significantly higher in PAPS and aPL carriers as well as aPL positive SLE and SAPS than in aPL negative controls. The highest values were found in PAPS and in SAPS. The EC4d and PC4d percentages were significantly correlated with serum C3/C4 and anti-β2GPI/anti-cardiolipin IgG. In vitro studies showed that MBB2 bound to activated platelets only and induced C4d deposition. The detection of the activation product C4d on circulating erythrocytes and platelets supports the role of complement activation in APS. Complement may represent a new therapeutic target for better treatment and prevention of disability of APS patients.
Collapse
Affiliation(s)
- Paola Adele Lonati
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Mariangela Scavone
- Unità di Medicina II, ASST Santi Paolo e Carlo, Milan, Italy.,Dipartimento di Scienze Della Salute, University of Milan, Milan, Italy
| | - Maria Gerosa
- Dipartimento di Scienze Cliniche e di Comunità, University of Milan, Milan, Italy
| | - Maria Orietta Borghi
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Dipartimento di Scienze Cliniche e di Comunità, University of Milan, Milan, Italy
| | - Francesca Pregnolato
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Daniele Curreli
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Gianmarco Podda
- Unità di Medicina II, ASST Santi Paolo e Carlo, Milan, Italy.,Dipartimento di Scienze Della Salute, University of Milan, Milan, Italy
| | - Eti Alessandra Femia
- Unità di Medicina II, ASST Santi Paolo e Carlo, Milan, Italy.,Dipartimento di Scienze Della Salute, University of Milan, Milan, Italy
| | - Wilma Barcellini
- UOC Ematologia, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Cattaneo
- Unità di Medicina II, ASST Santi Paolo e Carlo, Milan, Italy.,Dipartimento di Scienze Della Salute, University of Milan, Milan, Italy
| | - Francesco Tedesco
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Pier Luigi Meroni
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| |
Collapse
|