151
|
Egri N, Bentow C, Rubio L, Norman GL, López-Sañudo S, Mahler M, Pérez-Isidro A, Cervera R, Viñas O, Espinosa G, Ruiz-Ortiz E. Anti-Phosphatidylserine/Prothrombin Antibodies at Two Points: Correlation With Lupus Anticoagulant and Thrombotic Risk. Front Immunol 2021; 12:754469. [PMID: 34790198 PMCID: PMC8592011 DOI: 10.3389/fimmu.2021.754469] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/29/2021] [Indexed: 01/18/2023] Open
Abstract
Antibodies to phospholipids (aPL) and associated proteins are a hallmark in the diagnosis of anti-phospholipid syndrome (APS). Those included in the classification criteria are the lupus anticoagulant (LA) and the IgG and IgM isotypes of anticardiolipin (aCL) and anti-beta-2 glycoprotein I (β2GPI) antibodies. Non-classification criteria markers such as autoantibodies that recognize the phosphatidylserine/prothrombin (aPS/PT) complex have been proposed as biomarkers for APS. Studies of aPS/PT antibodies have shown a strong correlation to clinical manifestations and LA. We aimed to study the value and the persistence of aPS/PT IgG and IgM antibodies in a cohort of consecutive patients with clinical suspicion of APS and their utility as thrombotic risk markers. Our study, with 103 patients, demonstrates that persistently positive results for aPS/PT IgG antibodies were significantly associated with APS classification, thrombosis, triple aPL positivity, LA positive result, and the Global APS Score (GAPSS) > than 9 points (p < 0.01, for each condition). On the other hand, no association was seen with pregnancy morbidity (p = 0.56) and SLE (p = 0.07). Persistence of aPS/PT antibodies, defined according to the current laboratory classification criteria, likely improves the diagnosis and clinical assessment of patients with APS.
Collapse
Affiliation(s)
- Natalia Egri
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Chelsea Bentow
- Headquarters & Technology Center, Autoimmunity, Werfen, San Diego, CA, United States
| | - Laura Rubio
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Gary L. Norman
- Headquarters & Technology Center, Autoimmunity, Werfen, San Diego, CA, United States
| | | | - Michael Mahler
- Headquarters & Technology Center, Autoimmunity, Werfen, San Diego, CA, United States
| | - Albert Pérez-Isidro
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ricard Cervera
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Autoimmune Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Odette Viñas
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Gerard Espinosa
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Autoimmune Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Estíbaliz Ruiz-Ortiz
- Institut de Recerca Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
152
|
Ricard L, Laurent C, Papo M, Deriaz S, Catano J, Alamowitch S, Kayem G, Chasset F, De Moreuil C, Boffa JJ, Gerotziafas G, Elalamy I, Bornes M, Maillot F, Audemard-Verger A, Planche V, Ballot E, Fain O, Mekinian A. Clinical and prognostic significance of antinuclear antibodies in primary antiphospholipid syndrome: A multicenter retrospective study. Joint Bone Spine 2021; 89:105297. [PMID: 34656751 DOI: 10.1016/j.jbspin.2021.105297] [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: 07/08/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The antiphospholipid syndrome (APS) (1) is defined by the development of vascular thrombosis, or pregnancy morbidity in the presence of persistent antiphospholipid antibodies (aPL). Antinuclear antibodies (ANA) can be detected in primary APS patients without any clinical systemic autoimmune disease. The presence of ANA antibodies could confer a specific phenotype in primary APS. OBJECTIVE To evaluate the characteristics of APS patients with antinuclear antibodies without other autoimmune disease (ANA positive APS patients) in comparison with primary APS without ANA or secondary APS patients with associated systemic lupus erythematosus (SLE). METHODS Clinical and biologic data from 195 APS were retrospectively collected and patients were classified as primary APS with positive ANA (ANA-positive APS), primary APS without any ANA (ANA-negative APS), and SLE-associated APS (SLE-APS). RESULTS Fourty patients (21%) were classified into ANA-positive APS group, 77 (39%) in ANA-negative APS and 78 (40%) in SLE-APS. In ANA-positive APS patients, 20 patients (51%) had arterial thrombosis, 14 (41%) had veinous thrombosis and 19% had obstetrical complications. There was no difference between the three groups for the frequency of thrombotic manifestations and obstetrical complications. ANA-positive APS patients had more non-criteria manifestations than ANA-negative APS (48% versus 25%; P≤0.01). ANA-positive APS had more triple aPL positivity (59% versus 18%; P<0.001) and more thrombosis and obstetrical recurrences (63% versus 36%; P<0.01) in comparison with ANA-negative APS patients. ANA-positive APS had more triple aPL positivity than SLE-APS patients (54% versus 33%; P<0.05). ANA-positive APS and SLE-APS patients had similar clinical manifestations, and recurrences. Despite a limited follow-up (28 months (11-50)) none of the ANA-positive APS develop SLE. Antiplatelet and anticoagulant therapies were similar for the three groups. SLE-APS patients received more immunomodulatory therapies. CONCLUSION ANA positivity in patients with APS enables to individualize a subset of patients with a more severe phenotype. Whereas the ANA positivity does not seem to be associated with the risk to develop SLE, prospective studies with a longer follow-up are necessary, in particular to evaluate the effect of additional therapies in this subset of APS.
Collapse
Affiliation(s)
- Laure Ricard
- Service de médecine interne et Inflammation-Immunopathology-Biotherapy Department (DMU i3), hôpital Saint-Antoine, Sorbonne université, AP-HP, 75012 Paris, France
| | - Charlotte Laurent
- Service de médecine interne et Inflammation-Immunopathology-Biotherapy Department (DMU i3), hôpital Saint-Antoine, Sorbonne université, AP-HP, 75012 Paris, France
| | - Matthias Papo
- Médecine interne institut e3m, groupe hospitalier Pitié-Salpêtrière, Paris, France
| | - Sophie Deriaz
- Médecine interne, CHRU Hôpitaux de Tours, Tours, France
| | - Jennifer Catano
- Service de médecine interne et Inflammation-Immunopathology-Biotherapy Department (DMU i3), hôpital Saint-Antoine, Sorbonne université, AP-HP, 75012 Paris, France
| | - Sonia Alamowitch
- Service de neurologie et d'urgences neurovasculaires, hôpital Saint-Antoine, Paris, France
| | - Gilles Kayem
- Obstétrique, hôpital Armand-Trousseau, AP-HP, Paris, France
| | - François Chasset
- Service de dermatologie, faculté de médecine, hôpital Tenon, Sorbonne université, AP-HP, Paris, France
| | | | | | - Grigorios Gerotziafas
- Inserm UMR-S938 Cancer Biology and Therapeutics, Research Group "Cancer-Hemostasis-Angiogenesis" CRSA, institut universitaire de Cancérologie, Sorbonne université, Paris, France; Department of thrombosis and haemostasis, service d'hématologie Biologique, faculté de médecine, hôpital Tenon, hôpitaux universitaires de l'Est Parisien, Assistance publique-Hôpitaux de Paris, Sorbonne université, Paris, France
| | - Ismail Elalamy
- Inserm UMR-S938 Cancer Biology and Therapeutics, Research Group "Cancer-Hemostasis-Angiogenesis" CRSA, institut universitaire de Cancérologie, Sorbonne université, Paris, France; I M Sechenov First Moscow State Medical University, Department of Obstetrics and Gynecology, Moscow, Russia
| | - Marie Bornes
- Department of thrombosis and haemostasis, service d'hématologie Biologique, faculté de médecine, hôpital Tenon, hôpitaux universitaires de l'Est Parisien, Assistance publique-Hôpitaux de Paris, Sorbonne université, Paris, France
| | | | | | | | - Eric Ballot
- Immunologie biologique, hôpital Saint-Antoine, Paris, France
| | - Olivier Fain
- Service de médecine interne et Inflammation-Immunopathology-Biotherapy Department (DMU i3), hôpital Saint-Antoine, Sorbonne université, AP-HP, 75012 Paris, France
| | - Arsène Mekinian
- Service de médecine interne et Inflammation-Immunopathology-Biotherapy Department (DMU i3), hôpital Saint-Antoine, Sorbonne université, AP-HP, 75012 Paris, France.
| |
Collapse
|
153
|
Hu C, Li S, Xie Z, You H, Jiang H, Shi Y, Qi W, Zhao J, Wang Q, Tian X, Li M, Zhao Y, Zeng X. Evaluation of the Diagnostic Value of Non-criteria Antibodies for Antiphospholipid Syndrome Patients in a Chinese Cohort. Front Immunol 2021; 12:741369. [PMID: 34567005 PMCID: PMC8461188 DOI: 10.3389/fimmu.2021.741369] [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: 07/14/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022] Open
Abstract
Objective Although specific anti-phospholipid antibodies (aPLs) have been used in the diagnosis of the antiphospholipid syndrome (APS) for years, new biomarkers are required to increase its diagnostic and risk-predictive power. This study aimed to explore the value of several non-criteria aPLs in a Chinese cohort. Methods A total of 312 subjects, namely, 100 patients diagnosed with primary APS, 51 with APS secondary to SLE, 71 with SLE, and 90 healthy controls, were recruited. Serum anticardiolipin (aCL) IgG/IgM/IgA, anti-β2-glycoprotein I (aβ2GPI) IgG/IgM/IgA, anti-phosphatidylserine/prothrombin antibodies (aPS/PT) IgG/IgM, and anti-annexin A5 antibodies (aAnxV) IgG/IgM were tested using ELISA kits. Results Of the total number of patients, 30.46% and 6.62% with APS were positive for aCL or aβ2GPI IgA, respectively, while 39.07% and 24.50% were positive for aAnxV or aPS/PT for at least one antibody (IgG or IgM). The addition test of aCL IgA and aAnxV IgM assists in identifying seronegative APS patients, and IgG aPS/PT was linked to stroke. Conclusion Detection of aCL IgA, aβ2GPI IgA, aAnxV IgG/M, and aPS/PT IgG/M as a biomarker provides additive value in APS diagnosis and would help in risk prediction for APS patients in medical practice.
Collapse
Affiliation(s)
- Chaojun Hu
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Siting Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Zhijuan Xie
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Hanxiao You
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Hui Jiang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yu Shi
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Wanting Qi
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Qian Wang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xinping Tian
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
154
|
Chinnaraj M, Pengo V, Pozzi N. A Novel ELISA Assay for the Detection of Anti-Prothrombin Antibodies in Antiphospholipid Syndrome Patients at High Risk of Thrombosis. Front Immunol 2021; 12:741589. [PMID: 34567006 PMCID: PMC8455811 DOI: 10.3389/fimmu.2021.741589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/03/2022] Open
Abstract
Autoantibodies targeting prothrombin (aPT) can be found in antiphospholipid syndrome (APS) patients. However, their detection has proven difficult to standardize. Here, we developed a new ELISA assay to improve the identification of aPT and compared its performance with currently available anti-phosphatidylserine/prothrombin antibodies (aPS/PT) and autoantibodies targeting prothrombin bound to the plastic plate (aPT-A) assays using a cohort of 27 APS patients at high risk of thrombosis. We generated a novel prothrombin variant, ProTS525A-Biot, carrying an artificial tag at the C-terminus suitable for site-specific biotinylation and added the mutation S525A to improve stability. ProTS525A-Biot was immobilized to neutravidin-coated plates at the desired density and with a defined orientation, i.e., pointing the N-terminal fragment-1 toward the solvent. Antibodies against ProTS525A-Biot (aPT-Bio) were found in 24 out of 27 triple-positive APS patients (88%). When compared to aPS/PT and aPT-A, aPT-Bio showed an excellent linear correlation with aPS/PT (R2 = 0.85) but not with aPT-A (R2 = 0.40). Since aPS/PT but not aPT-A are an emerging biomarker of thrombosis in APS, this method may find utility for detecting pathogenic aPT in APS but also other prothrombotic conditions such as COVID-19.
Collapse
Affiliation(s)
- Mathivanan Chinnaraj
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Vittorio Pengo
- Thrombosis Research Laboratory, Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padua, Italy.,Arianna Foundation on Anticoagulation, Bologna, Italy
| | - Nicola Pozzi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
155
|
Chu H, Sacharidou A, Nguyen A, Li C, Chambliss KL, Salmon JE, Shen YM, Lo J, Leone GW, Herz J, Hui DY, Marciano DK, Abrahams VM, Natale BV, Montalbano AP, Xiao X, Xu L, Natale DR, Shaul PW, Mineo C. Protein Phosphatase 2A Activation Via ApoER2 in Trophoblasts Drives Preeclampsia in a Mouse Model of the Antiphospholipid Syndrome. Circ Res 2021; 129:735-750. [PMID: 34404233 DOI: 10.1161/circresaha.120.318941] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Haiyan Chu
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - An Nguyen
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chun Li
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Jane E Salmon
- Medicine, Hospital for Special Surgery, Weill Cornell Medical College, New York (J.E.S.)
| | - Yu-Min Shen
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas
| | - Julie Lo
- Obstetrics and Gynecology (J.L.), University of Texas Southwestern Medical Center, Dallas
| | - Gustavo W Leone
- Froedtert-Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee (G.W.L.)
| | - Joachim Herz
- Molecular Genetics (J.H.), University of Texas Southwestern Medical Center, Dallas
| | - David Y Hui
- Pathology, University of Cincinnati College of Medicine (D.Y.H.)
| | - Denise K Marciano
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas.,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| | - Vikki M Abrahams
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT (V.M.A.)
| | - Bryony V Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Alina P Montalbano
- Biochemistry and Obstetrics and Gynecology (A.P.M.), University of Texas Southwestern Medical Center, Dallas
| | - Xue Xiao
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - Lin Xu
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - David R Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.).,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
156
|
A Novel Mutation in the VPS13B Gene in a Cohen Syndrome Patient with Positive Antiphospholipid Antibodies. Case Reports Immunol 2021; 2021:3143609. [PMID: 34484844 PMCID: PMC8413065 DOI: 10.1155/2021/3143609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
Cohen syndrome is an autosomal recessive disorder with the primary symptoms of mental deficiency, progressive retinopathy, hypotonia, microcephaly, obesity of midchildhood onset, intermittent neutropenia, and dysmorphic facial features. The syndrome has high phenotypic heterogeneity and is caused by loss-of-function mutations in the VPS13B gene. Here, we introduce a novel homozygous nonsense mutation (c.8698G > T, p.E2900X) in the VPS13B gene in an 11-year-old Iranian boy with major symptoms of Cohen syndrome. He also had mild anemia accompanied by positive antiphospholipid antibodies, the latter has never been previously reported in Cohen syndrome.
Collapse
|
157
|
Hollerbach A, Müller-Calleja N, Pedrosa D, Canisius A, Sprinzl MF, Falter T, Rossmann H, Bodenstein M, Werner C, Sagoschen I, Münzel T, Schreiner O, Sivanathan V, Reuter M, Niermann J, Galle PR, Teyton L, Ruf W, Lackner KJ. Pathogenic lipid-binding antiphospholipid antibodies are associated with severity of COVID-19. J Thromb Haemost 2021; 19:2335-2347. [PMID: 34242469 PMCID: PMC8420426 DOI: 10.1111/jth.15455] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Coronavirus disease 19 (COVID-19)-associated coagulopathy is a hallmark of disease severity and poor prognosis. The key manifestations of this prothrombotic syndrome-microvascular thrombosis, stroke, and venous and pulmonary clots-are also observed in severe and catastrophic antiphospholipid syndrome. Antiphospholipid antibodies (aPL) are detectable in COVID-19 patients, but their association with the clinical course of COVID-19 remains unproven. OBJECTIVES To analyze the presence and relevance of lipid-binding aPL in hospitalized COVID-19 patients. METHODS Two cohorts of 53 and 121 patients from a single center hospitalized for PCR-proven severe acute respiratory syndrome-coronavirus 2 infection were analyzed for the presence of aPL and clinical severity of COVID-19. RESULTS We here demonstrate that lipid-binding aPL are common in COVID-19. COVID-19 patients with lipid-binding aPL have higher median concentrations of C-reactive protein and D-dimer, and are more likely to have a critical clinical course and fatal outcome. Lipid-binding aPL isolated from COVID-19 patients target the recently described cell surface complex of lysobisphosphatidic acid (LBPA) with the protein C receptor (EPCR) to induce prothrombotic and inflammatory responses in monocytes and endothelial cells. We show that B1a cells producing lipid-reactive aPL of the IgG isotype circulate in the blood of COVID-19 patients. In vivo, COVID-19 aPL accelerate thrombus formation in an experimental mouse model dependent on the recently delineated signaling pathway involving EPCR-LBPA. CONCLUSIONS COVID-19 patients rapidly expand B1a cells secreting pathogenic lipid-binding aPL with broad thrombotic and inflammatory effects. The association with markers of inflammation and coagulation, clinical severity, and mortality suggests a causal role of aPL in COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Martin F Sprinzl
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Tanja Falter
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Heidi Rossmann
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| | - Marc Bodenstein
- Department of Anesthesiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Christian Werner
- Department of Anesthesiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Ingo Sagoschen
- Department of Cardiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Oliver Schreiner
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Visvakanth Sivanathan
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Michael Reuter
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Johannes Niermann
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Medicine I, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center Mainz, Germany
| |
Collapse
|
158
|
Hong JL, Hou YT, Lin PC, Chen YL, Chien DS, Yiang GT, Wu MY. Antiphospholipid Syndrome-Induced Leriche Syndrome in a Man with Lower Limbs Sensory and Motor Defect. J Cardiovasc Dev Dis 2021; 8:jcdd8090104. [PMID: 34564122 PMCID: PMC8472144 DOI: 10.3390/jcdd8090104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder with characteristics of arterial and/or venous thrombosis due to hypercoagulation status. Although deep vein thrombosis is common, the involvement of arterial thrombosis is more dangerous and poses a high risk of complications. Acute aorto-iliac occlusive disease (AIOD, known as Leriche syndrome) is severe arterial thrombosis that is associated with high morbidity and mortality rates. Severe acute occlusion may cause spinal cord ischemia, leading to neurological defects, such as acute onset of paraplegia. Co-occurrence of acute aorto-iliac occlusive disease and antiphospholipid syndrome is rare and may present with atypical symptoms mimicking other diseases, including chronic ulcers, musculoskeletal events, and pulmonary diseases. In patients with weak femoral pulses and recurrent thrombotic events, co-occurrence of APS and AIOD should be taken into consideration. Here, we describe a rare case of co-occurrence of APS and AIOD presenting with acute lower leg weakness and numbness. Timely thrombectomies and bilateral common iliac artery stentings rescued distal blood flow. We highlight the clinical features and early diagnosis of co-occurrence of APS and AIOD in order to prevent catastrophic complications. The detailed mechanism and pathogenesis of antiphospholipid syndrome-induced acute aorto-iliac occlusive disease are also discussed.
Collapse
Affiliation(s)
- Jeng-Luen Hong
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Yueh-Tseng Hou
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Po-Chen Lin
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Yu-Long Chen
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Da-Sen Chien
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan; (J.-L.H.); (Y.-T.H.); (P.-C.L.); (Y.-L.C.); (D.-S.C.); (G.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Correspondence: ; Tel.: +886-2-6628-9779; Fax: +886-2-6628-9009
| |
Collapse
|
159
|
Sobue T, Fukuda H, Matsumoto T, Lee B, Ito S, Iwata S. The background occurrence of selected clinical conditions prior to the start of an extensive national vaccination program in Japan. PLoS One 2021; 16:e0256379. [PMID: 34437567 PMCID: PMC8389412 DOI: 10.1371/journal.pone.0256379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/04/2021] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION The COVID-19 pandemic caused by SARS-CoV-2 has now affected tens of millions of people globally. It is the hope that vaccines against SARS-CoV-2 will deliver a comprehensive solution to this global pandemic; however, this will require extensive national vaccination programs. Ultimately, clinical conditions and even sudden unexplained death will occur around the time of vaccination, thus a distinction needs to be made between events that are causally related to the vaccine or temporally related to vaccination. This study aimed to estimate the background occurrence of 43 clinical conditions in the Japanese population. METHODS A retrospective cohort study was conducted from 2013 to 2019 using data from two large healthcare claims databases (MDV and JMDC) in Japan. The estimated number of new cases and incidence were calculated based on the actual number of new cases identified in the databases. The PubMed and Ichushi-web databases, as well as grey literature such as guidelines and government statistics, were also searched to identify any publications related to incidence of these conditions in Japan. RESULTS AND CONCLUSION The estimates of the number of total cases and incidence were similar for the MDV and JMDC databases for some diseases. In addition, some estimates were similar to those in the scientific literature. For example, from the MDV and JMDC databases, estimates of incidence of confirmed Bell's palsy in 2019 were 41.7 and 47.9 cases per 100,000 population per year, respectively. These estimates were of the same order from the scientific publication. Determining whether clinical conditions occurring around the time of vaccination are causally or only temporally related to vaccination will be critical for public health decision makers as well as for the general public. Comparison of background occurrence at the population level may provide some additional objective evidence for the evaluation of temporality or causality.
Collapse
Affiliation(s)
- Tomotaka Sobue
- Division of Environmental Medicine and Population Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University, Fukuoka, Japan
| | - Tetsuya Matsumoto
- Department of Infectious Diseases, International University of Health and Welfare, Narita, Japan
| | - Bennett Lee
- Vaccine Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | - Shuhei Ito
- Vaccine Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | - Satoshi Iwata
- Department of Infectious Diseases, National Cancer Center Hospital, Tokyo, Japan
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
160
|
Liu T, Dai J, Yang Z, Yu X, Xu Y, Shi X, Wei D, Tang Z, Xu G, Xu W, Liu Y, Shi C, Ni Q, Yang C, Zhang X, Wang X, Chen E, Qu J. Inactivated SARS-CoV-2 vaccine does not influence the profile of prothrombotic antibody nor increase the risk of thrombosis in a prospective Chinese cohort. Sci Bull (Beijing) 2021; 66:2312-2319. [PMID: 34336365 PMCID: PMC8313791 DOI: 10.1016/j.scib.2021.07.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 07/22/2021] [Indexed: 01/06/2023]
Abstract
The presence of antiphospholipid antibodies was shown to be associated with thrombosis in coronavirus disease 2019 (COVID-19) patients. Recently, according to reports from several studies, the vaccine-induced immune thrombotic thrombocytopenia is mediated by anti-platelet factor 4 (PF4)-polyanion complex in adenovirus-vectored COVID-19 vaccine recipients. It is impendent to explore whether inactivated COVID-19 vaccine widely used in China influences prothrombotic autoantibody production and induces thrombosis. In this prospective study, we recruited 406 healthcare workers who received two doses, 21 days apart, of inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine (BBIBP-CorV, Sinopharm). Paired blood samples taken before vaccination and four weeks after the second vaccination were used in detecting prothrombotic autoantibodies, including anticardiolipin (aCL), anti-β2 glycoprotein I (aβ2GP1), anti-phosphatidylserine/prothrombin (aPS/PT), and anti-PF4-heparin. The seroconversion rate of SARS-CoV-2 specific antibodies was 95.81% (389/406) four weeks after vaccination. None of the subjects had spontaneous thrombosis or thrombocytopenia over a minimum follow-up period of eight weeks. There was no significant difference in the presence of all ten autoantibodies between samples collected before and after vaccination: for aCL, IgG (7 vs. 8, P = 0.76), IgM (41 vs. 44, P = 0.73), IgA (4 vs. 4, P = 1.00); anti-β2GP1, IgG (7 vs. 6, P = 0.78), IgM (6 vs. 5, P = 0.76), IgA (3 vs. 5, P = 0.72); aPS/PT IgG (0 vs. 0, P = 1.00), IgM (6 vs. 5, P = 0.76); aPF4-heparin (2 vs. 7, P = 0.18), and antinuclear antibody (ANA) (18 vs. 21, P = 0.62). Notably, seven cases presented with anti-PF4-heparin antibodies (range: 1.18–1.79 U/mL) after vaccination, and none of them exhibited any sign of thrombotic disorder. In conclusion, inactivated SARS-CoV-2 vaccine does not influence the profile of antiphospholipid antibody and anti-PF4-heparin antibody nor increase the risk of thrombosis.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Dai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhitao Yang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoqi Yu
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanping Xu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Xinming Shi
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dong Wei
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zihan Tang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guanqun Xu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenxin Xu
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Liu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ce Shi
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Ni
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinxin Zhang
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| |
Collapse
|
161
|
Walter IJ, Klein Haneveld MJ, Lely AT, Bloemenkamp KWM, Limper M, Kooiman J. Pregnancy outcome predictors in antiphospholipid syndrome: A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102901. [PMID: 34280554 DOI: 10.1016/j.autrev.2021.102901] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 05/21/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To identify and assess the magnitude of effect of pregnancy outcome predictors in women with antiphospholipid syndrome (APS) by means of systematic review and meta-analysis. METHODS PubMed and Embase were searched (13th June 2020) for studies reporting on pre-pregnancy risk factors of pregnancy outcomes in APS patients. Literature screening and data extraction were conducted by two reviewers independently, in a blinded standardized manner. Pooled univariate odds ratios (OR) were computed using a random effects model. Heterogeneity was assessed by I2%. RESULTS The search yielded 3013 unique results; 27 records were included in this meta-analysis. Previous thrombosis was associated with a decreased live birth risk (OR 0.60, p < 0.01, I2 = 40%), increased neonatal mortality (OR 15.19, p < 0.01, I2 = 0%), an increased risk of antenatal or postpartum thrombosis (OR 6.26, p < 0.01, I2 = 0%) and an increased risk of delivering a small for gestational age neonate (SGA) (OR 2.60, p = 0.01, I2 = 0%). Patients with an APS laboratory category I (double or triple positivity) profile had a decreased live birth risk (OR 0.66, p < 0.01, I2 = 0%), an increased risk of SGA (OR 1.86, p = 0.01, I2 = 43%) and preterm birth (OR 1.35, p < 0.01, I2 = 49%). Triple positivity was associated with a decreased live birth risk (OR 0.33, p < 0.01, I2 = 68%), an increased risk of preeclampsia (OR 2.43, p = 0.02, I2 = 35%) and SGA (OR 2.47, p = 0.04, I2 = 61%). Patients with lupus anticoagulant positivity had an increased risk of preeclampsia (OR 2.10, p = 0.02, I2 = 48%), SGA (OR 1.78, p < 0.01, I2 = 0%) and preterm birth (OR 3.56, p = 0.01, I2 = 48%). Risk of bias assessment suggested considerable bias on study participation and statistical methods. CONCLUSIONS The results of this meta-analysis identified previous thrombosis, laboratory category I, triple positivity and lupus anticoagulant positivity as the most important predictors of adverse pregnancy outcomes. This up-to-date knowledge, can be used in preconception counseling and tailoring of obstetric care.
Collapse
Affiliation(s)
- Isabel Johanna Walter
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Obstetrics, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Mirthe Jasmijn Klein Haneveld
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Obstetrics, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Anne Titia Lely
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Obstetrics, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | | | - Maarten Limper
- University Medical Center Utrecht, Department of Rheumatology and Clinical Immunology, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Judith Kooiman
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Obstetrics, Lundlaan 6, 3584 EA Utrecht, the Netherlands.
| |
Collapse
|
162
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
163
|
Roccatello D, Sciascia S. A toggle switch linking coagulation and innate immunity in antiphospholipid antibody syndrome. Kidney Int 2021; 100:740-742. [PMID: 34216676 DOI: 10.1016/j.kint.2021.06.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 01/13/2023]
Affiliation(s)
- Dario Roccatello
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; CMID-Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Torino, Italy.
| | - Savino Sciascia
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; CMID-Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Torino, Italy
| |
Collapse
|
164
|
Weber BN, Stevens E, Barrett L, Bay C, Sinnette C, Brown JM, Divakaran S, Bibbo C, Hainer J, Dorbala S, Blankstein R, Liao K, Massarotti E, Costenbader K, Di Carli MF. Coronary Microvascular Dysfunction in Systemic Lupus Erythematosus. J Am Heart Assoc 2021; 10:e018555. [PMID: 34132099 PMCID: PMC8403317 DOI: 10.1161/jaha.120.018555] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Systemic lupus erythematosus (SLE) is a systemic autoimmune inflammatory disorder associated with premature atherosclerosis and increased cardiovascular risk. Systemic inflammation is an emerging risk factor for coronary microvascular dysfunction (CMD). We aimed to test whether CMD, defined as abnormal myocardial flow reserve (MFR) by positron emission tomography‐computed tomography, would be independently associated with SLE after adjusting for nonobstructive atherosclerotic burden and common cardiovascular risk factors. Methods and Results Consecutive patients with SLE who underwent symptom‐prompted stress cardiac positron emission tomography‐computed tomography were included (n=42). Obstructive coronary artery disease and systolic dysfunction were excluded. MFR was quantified by positron emission tomography‐computed tomography, and CMD was defined as MFR <2. We frequency matched patients who did not have SLE and had symptom‐prompted positron emission tomography studies on age, sex, and key cardiovascular risk factors (n=69). The attenuation correction computed tomography scans were reviewed for qualitative assessment of coronary artery calcium. Patients with SLE had a more severe reduction in global MFR compared with controls and a higher prevalence of CMD, despite a similar degree of nonobstructive atherosclerotic burden (1.91±0.5 versus 2.4±0.7, respectively, P<0.0001; CMD, 57.1% versus 33.3%, respectively, P=0.017). Conclusions We demonstrated that patients with SLE with cardiac symptoms without obstructive coronary artery disease have a high prevalence of coronary vasomotor abnormalities. In comparison with symptomatic matched controls, patients with SLE have a more severe reduction in MFR that is not accounted for by common cardiovascular factors or atherosclerotic burden.
Collapse
Affiliation(s)
- Brittany N Weber
- Division of Cardiovascular Medicine Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBoston MA.,Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Emma Stevens
- Division of Rheumatology, Inflammation, and Immunity Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Leanne Barrett
- Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Camden Bay
- Department of Radiology Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Corine Sinnette
- Division of Rheumatology, Inflammation, and Immunity Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Jenifer M Brown
- Division of Cardiovascular Medicine Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBoston MA.,Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Sanjay Divakaran
- Division of Cardiovascular Medicine Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBoston MA.,Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Courtney Bibbo
- Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Jon Hainer
- Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Sharmila Dorbala
- Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Ron Blankstein
- Division of Cardiovascular Medicine Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBoston MA.,Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| | - Katherine Liao
- Division of Rheumatology, Inflammation, and Immunity Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Elena Massarotti
- Division of Rheumatology, Inflammation, and Immunity Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Karen Costenbader
- Division of Rheumatology, Inflammation, and Immunity Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Marcelo F Di Carli
- Division of Cardiovascular Medicine Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBoston MA.,Cardiovascular Imaging Program Departments of Medicine and RadiologyBrigham and Women's HospitalHarvard Medical SchoolBoston MA
| |
Collapse
|
165
|
Impaired Autophagy Induced by oxLDL/ β2GPI/anti- β2GPI Complex through PI3K/AKT/mTOR and eNOS Signaling Pathways Contributes to Endothelial Cell Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6662225. [PMID: 34221236 PMCID: PMC8219424 DOI: 10.1155/2021/6662225] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/22/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022]
Abstract
Endothelial cell dysfunction plays a fundamental role in the pathogenesis of atherosclerosis (AS), and endothelial autophagy has protective effects on the development of AS. Our previous study had shown that oxidized low-density lipoprotein/β2-glycoprotein I/anti-β2-glycoprotein I antibody (oxLDL/β2GPI/anti-β2GPI) complex could promote the expressions of inflammatory cytokines and enhance the adhesion of leukocytes to endothelial cells. In the present study, we aimed to assess the effects of oxLDL/β2GPI/anti-β2GPI complex on endothelial autophagy and explore the associated potential mechanisms. Human umbilical vein endothelial cells (HUVECs) and mouse brain endothelial cell line (bEnd.3) were used as models of the vascular endothelial cells. Autophagy was evaluated by examining the expressions of autophagic proteins using western blotting analysis, autophagosome accumulation using transmission electron microscopy, and RFP-GFP-LC3 adenoviral transfection and autophagic flux using lysosome inhibitor chloroquine. The expressions of phospho-PI3K, phospho-AKT, phospho-mTOR, and phospho-eNOS were determined by western blotting analysis. 3-Methyladenine (3-MA) and rapamycin were used to determine the role of autophagy in oxLDL/β2GPI/anti-β2GPI complex-induced endothelial cell dysfunction. We showed that oxLDL/β2GPI/anti-β2GPI complex suppressed the autophagy, evidenced by an increase in p62 protein, a decrease in LC3-II and Beclin1, and a reduction of autophagosome generation in endothelial cells. Moreover, inhibition of autophagy was associated with PI3K/AKT/mTOR and eNOS signaling pathways. Rapamycin attenuated oxLDL/β2GPI/anti-β2GPI complex-induced endothelial inflammation, oxidative stress, and apoptosis, whereas 3-MA alone induced the endothelial injury. Our results suggested that oxLDL/β2GPI/anti-β2GPI complex inhibited endothelial autophagy via PI3K/AKT/mTOR and eNOS signaling pathways and further contributed to endothelial cell dysfunction. Collectively, our findings provided a novel mechanism for vascular endothelial injury in AS patients with an antiphospholipid syndrome (APS) background.
Collapse
|
166
|
Leal Rato M, Bandeira M, Romão VC, Aguiar de Sousa D. Neurologic Manifestations of the Antiphospholipid Syndrome - an Update. Curr Neurol Neurosci Rep 2021; 21:41. [PMID: 34125304 PMCID: PMC8200381 DOI: 10.1007/s11910-021-01124-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW In recent years, the spectrum of neurological manifestations of antiphospholipid syndrome (APS) has been growing. We provide a critical review of the literature with special emphasis on presentation, proposed mechanisms of disease, and treatment of neurological involvement in APS. RECENT FINDINGS Although stroke is the most common cause of neurological manifestations in patients with APS, other neurological disorders have been increasingly associated with the disease, including cognitive dysfunction, headache, and epilepsy. Direct oral anticoagulants have failed to show non-inferiority compared to vitamin K antagonists for the prevention of major thrombotic events. Antiphospholipid antibodies are often found in patients with acute COVID-19 but clear evidence supporting an association between these antibodies and the risk of thrombotic events, including stroke and cerebral venous thrombosis, is still lacking. APS patients may present with several distinct neurological manifestations. New criteria will facilitate the classification of patients presenting with increasingly recognized non-criteria neurological manifestations.
Collapse
Affiliation(s)
- Miguel Leal Rato
- Neurology, Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Matilde Bandeira
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Vasco C Romão
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Diana Aguiar de Sousa
- Neurology, Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal.
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal.
| |
Collapse
|
167
|
How I treat anticoagulant-refractory thrombotic antiphospholipid syndrome. Blood 2021; 137:299-309. [PMID: 32898856 DOI: 10.1182/blood.2020004942] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022] Open
Abstract
The standard treatment of thrombotic antiphospholipid syndrome (APS) is lifelong oral anticoagulation with a vitamin K antagonist (VKA), generally warfarin. A minority of patients with APS rethrombose despite seemingly adequate anticoagulation. These patients are deemed anticoagulant refractory. The management of anticoagulant-refractory APS is largely empirical and extrapolated from other clinically similar situations. Further options include increased VKA anticoagulation intensity or alternative antithrombotic strategies, including low-molecular-weight heparin, fondaparinux, the addition of antiplatelet therapy, and consideration of vascular options. Patients with anticoagulant-refractory thrombotic APS may have APS-associated thrombocytopenia, which necessitates balancing the risk of recurrent thrombosis vs bleeding to achieve adequate anticoagulation. The multiple mechanisms involved in the generation of the thrombotic phenotype in APS suggest that anticoagulation alone may not control thrombosis. Thus, other modalities, including adjunctive treatment (hydroxychloroquine, statins, and vitamin D) for APS-related thrombosis, merit consideration, as do immunomodulatory therapy and complement inhibition. Patients with APS may have coexistent systemic lupus erythematosus, which adds to the complexity of managing their thromboembolic disease. However, with attention to detail and judicious application of the limited data, it is possible to minimize the morbidity resulting from anticoagulant-refractory thrombotic APS. Multicenter studies are required to guide the sequence of interventions and their comparative efficacy in patients with anticoagulant-refractory thrombotic APS.
Collapse
|
168
|
Naranjo L, Ostos F, Gil-Etayo FJ, Hernández-Gallego J, Cabrera-Marante Ó, Pleguezuelo DE, Díaz-Simón R, Cerro M, Lora D, Martínez-Salio A, Serrano A. Presence of Extra-Criteria Antiphospholipid Antibodies Is an Independent Risk Factor for Ischemic Stroke. Front Cardiovasc Med 2021; 8:665741. [PMID: 34012984 PMCID: PMC8126615 DOI: 10.3389/fcvm.2021.665741] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/01/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Ischemic stroke is the most common and severe arterial thrombotic event in Antiphospholipid syndrome (APS). APS is an autoimmune disease characterized by the presence of thrombosis and antiphospholipid antibodies (aPL), which provide a pro-coagulant state. The aPL included in the classification criteria are lupus anticoagulant, anti-cardiolipin (aCL) and anti-β2-glycoprotein-I antibodies (aB2GPI) of IgG and IgM isotypes. Extra-criteria aPL, especially IgA aB2GPI and IgG/IgM anti-phosphatidylserine/prothrombin antibodies (aPS/PT), have been strongly associated with thrombosis. However, their role in the general population suffering from stroke is unknown. We aim (1) to evaluate the aPL prevalence in ischemic stroke patients, (2) to determine the role of aPL as a risk factor for stroke, and (3) to create an easy-to-use tool to stratify the risk of ischemic stroke occurrence considering the presence of aPL and other risk factors. Materials and Methods: A cohort of 245 consecutive ischemic stroke patients was evaluated in the first 24 h after the acute event for the presence of classic aPL, extra-criteria aPL (IgA aB2GPI, IgG, and IgM aPS/PT) and conventional cardiovascular risk factors. These patients were followed-up for 2-years. A group of 121 healthy volunteers of the same age range and representative of the general population was used as reference population. The study was approved by the Ethics Committee for Clinical Research (Reference numbers CEIC-14/354 and CEIC-18/182). Results: The overall aPL prevalence in stroke patients was 28% and IgA aB2GPI were the most prevalent (20%). In the multivariant analysis, the presence of IgA aB2GPI (OR 2.40, 95% CI: 1.03–5.53), dyslipidemia (OR 1.70, 95% CI: 1.01–2.84), arterial hypertension (OR 1.82, 95% CI: 1.03–3.22), atrial fibrillation (OR 4.31, 95% CI: 1.90–9.78), and active smoking (OR 3.47, 95% CI: 1.72–6.99) were identified as independent risk factors for ischemic stroke. A risk stratification tool for stroke was created based on these factors (AUC: 0.75). Conclusions: IgA aB2GPI are an important independent risk factor for ischemic stroke. Evaluation of aPL (including extra-criteria) in cardiovascular risk factor assessment for stroke can potentially increase the identification of patients at risk of thrombotic event, facilitating a decision on preventive treatments.
Collapse
Affiliation(s)
- Laura Naranjo
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Ostos
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Neurology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Francisco Javier Gil-Etayo
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jesús Hernández-Gallego
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Neurology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Óscar Cabrera-Marante
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Daniel Enrique Pleguezuelo
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Raquel Díaz-Simón
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Internal Medicine Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mercedes Cerro
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Nursing, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - David Lora
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Epidemiology Department, Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), Madrid, Spain.,Faculty of Statistical Studies, Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio Martínez-Salio
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Neurology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Antonio Serrano
- Healthcare Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
169
|
Abstract
LEARNING OBJECTIVES After studying this article, the participant should be able to: 1. Understand the available donor sites for autologous breast reconstruction. 2. Describe the advantages and limitations of each donor site. 3. Provide a rational, algorithmic preoperative evaluation and approach for patients seeking autologous breast reconstruction. 4. Develop an effective postoperative monitoring system to minimize complications and maximize salvage of microvascular thromboses. SUMMARY Breast reconstruction remains at the heart of the field of plastic and reconstructive surgery, and it is continuously evolving. Tremendous advances in breast implant technology and supplemental products, particularly acellular dermal matrices, have revolutionized breast reconstruction in the modern era. However, microvascular free flap breast reconstruction has also witnessed profound advancements with exceptionally high success rates, with the ability to provide the most durable and natural breast reconstruction. Although the pendulum oscillates between prosthesis-based reconstruction and autologous tissue, the present synopsis will focus on autologous free flap breast reconstruction from an historical perspective, recent advancements in microsurgery, and the future of autologous breast reconstruction.
Collapse
|
170
|
Gendron N, Dragon-Durey MA, Chocron R, Darnige L, Jourdi G, Philippe A, Chenevier-Gobeaux C, Hadjadj J, Duchemin J, Khider L, Yatim N, Goudot G, Krzisch D, Debuc B, Mauge L, Levasseur F, Pene F, Boussier J, Sourdeau E, Brichet J, Ochat N, Goulvestre C, Peronino C, Szwebel TA, Pages F, Gaussem P, Samama CM, Cheurfa C, Planquette B, Sanchez O, Diehl JL, Mirault T, Fontenay M, Terrier B, Smadja DM. Lupus anticoagulant single positivity at acute phase is not associated with venous thromboembolism or in-hospital mortality in COVID-19. Arthritis Rheumatol 2021; 73:1976-1985. [PMID: 33881229 PMCID: PMC8250965 DOI: 10.1002/art.41777] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/16/2021] [Indexed: 01/08/2023]
Abstract
Objective The clinical relevance of antiphospholipid antibodies (aPLs) in COVID‐19 is controversial. This study was undertaken to investigate the prevalence and prognostic value of conventional and nonconventional aPLs in patients with COVID‐19. Methods This was a multicenter, prospective observational study in a French cohort of patients hospitalized with suspected COVID‐19. Results Two hundred forty‐nine patients were hospitalized with suspected COVID‐19, in whom COVID‐19 was confirmed in 154 and not confirmed in 95. We found a significant increase in lupus anticoagulant (LAC) positivity among patients with COVID‐19 compared to patients without COVID‐19 (60.9% versus 23.7%; P < 0.001), while prevalence of conventional aPLs (IgG and IgM anti–β2‐glycoprotein I and IgG and IgM anticardiolipin isotypes) and nonconventional aPLs (IgA isotype of anticardiolipin, IgA isotype of anti‐β2‐glycoprotein I, IgG and IgM isotypes of anti–phosphatidylserine/prothrombin, and IgG and IgM isotypes of antiprothrombin) was low in both groups. Patients with COVID‐19 who were positive for LAC, as compared to patients with COVID‐19 who were negative for LAC, had higher levels of fibrinogen (median 6.0 gm/liter [interquartile range 5.0–7.0] versus 5.3 gm/liter [interquartile range 4.3–6.4]; P = 0.028) and C‐reactive protein (CRP) (median 115.5 mg/liter [interquartile range 66.0–204.8] versus 91.8 mg/liter [interquartile range 27.0–155.1]; P = 0.019). Univariate analysis did not show any association between LAC positivity and higher risks of venous thromboembolism (VTE) (odds ratio 1.02 [95% confidence interval 0.44–2.43], P = 0.95) or in‐hospital mortality (odds ratio 1.80 [95% confidence interval 0.70–5.05], P = 0.24). With and without adjustment for CRP level, age, and sex, Kaplan‐Meier survival curves according to LAC positivity confirmed the absence of an association with VTE or in‐hospital mortality (unadjusted P = 0.64 and P = 0.26, respectively; adjusted hazard ratio 1.13 [95% confidence interval 0.48–2.60] and 1.80 [95% confidence interval 0.67–5.01], respectively). Conclusion Patients with COVID‐19 have an increased prevalence of LAC positivity associated with biologic markers of inflammation. However, LAC positivity at the time of hospital admission is not associated with VTE risk and/or in‐hospital mortality.
Collapse
Affiliation(s)
- Nicolas Gendron
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Marie-Agnès Dragon-Durey
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France, Immunology department, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Richard Chocron
- Université de Paris, PARCC, INSERM, F-75015 Paris, France, Emergency department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Luc Darnige
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Georges Jourdi
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Aurélien Philippe
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Camille Chenevier-Gobeaux
- Department of Automated Diagnostic Biology, Hôpital Cochin, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Jérôme Hadjadj
- Université de Paris Imagine institute, laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, F-75015, Paris Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75014, Paris, France
| | - Jérôme Duchemin
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Lina Khider
- Université de Paris, Vascular Medicine department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Nader Yatim
- Translational Immunology lab, Department of Immunology, Institut Pasteur, and Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Guillaume Goudot
- Université de Paris, Vascular Medicine department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Daphné Krzisch
- Université de Paris, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Benjamin Debuc
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Plastic surgery department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Laetitia Mauge
- Université de Paris, PARCC, INSERM, F-75015 Paris, France, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Françoise Levasseur
- Université de Paris, Institut Cochin, INSERM, F-75014 Paris, France, Hematology department Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75014, Paris, France
| | - Frédéric Pene
- Intensive care medicine, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Jeremy Boussier
- Department of Automated Diagnostic Biology, Hôpital Cochin, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Elise Sourdeau
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Julie Brichet
- Université de Paris, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Nadège Ochat
- Université de Paris, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Claire Goulvestre
- logy department, Assistance Publique Hôpitaux de Paris. Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Christophe Peronino
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Tali-Anne Szwebel
- Translational Immunology lab, Department of Immunology, Institut Pasteur, and Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Franck Pages
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France, Immunology department, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Pascale Gaussem
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Charles-Marc Samama
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Anaesthesia, Intensive Care and Perioperative Medicine Department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Cherifa Cheurfa
- Anaesthesia, Intensive Care and Perioperative Medicine Department, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - Benjamin Planquette
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Respiratory Medicine Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France.,F-CRIN INNOVTE, Saint-Étienne, France
| | - Olivier Sanchez
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Respiratory Medicine Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France.,F-CRIN INNOVTE, Saint-Étienne, France
| | - Jean-Luc Diehl
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Intensive care unit and Biosurgical research lab (Carpentier Foundation), Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Tristan Mirault
- Université de Paris, Vascular Medicine department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75015, Paris, France
| | - Michaela Fontenay
- Université de Paris, Institut Cochin, INSERM, F-75014 Paris, France, Hematology department Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), F-75014, Paris, France
| | - Benjamin Terrier
- Department of Automated Diagnostic Biology, Hôpital Cochin, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75014, Paris, France
| | - David M Smadja
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006 Paris, France, Hematology department and Biosurgical research lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), F-75015, Paris, France.,F-CRIN INNOVTE, Saint-Étienne, France
| |
Collapse
|
171
|
Capecchi M, Ciavarella A, Artoni A, Abbattista M, Martinelli I. Thrombotic Complications in Patients with Immune-Mediated Hemolysis. J Clin Med 2021; 10:1764. [PMID: 33919638 PMCID: PMC8073900 DOI: 10.3390/jcm10081764] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
Autoimmune hemolytic anemias are rare and heterogeneous disorders characterized by hemolysis, which is a well-recognized risk factor for thrombosis. The most common immune-mediated anemias are represented by autoimmune hemolytic anemia and paroxysmal nocturnal hemoglobinuria, both associated with a high rate of thrombosis. Multiple pathophysiological mechanisms for thrombosis have been proposed, involving hemolysis itself and additional effects of the immune system. Despite the increasing awareness of the thrombotic risk in these conditions, evidence-based guidance on prevention and management of thrombotic events is lacking. We herein report available evidence on epidemiological data on thrombosis and thrombophilia in immune-mediated hemolysis, together with possible underlying pathophysiological mechanisms. In addition, we summarize current recommendations for treatment of thrombosis in immune-mediated hemolysis. In particular, we address the issue of thrombotic complications treatment and prophylaxis by proposing a therapeutic algorithm, focusing on specific situations such as splenectomy and pregnancy.
Collapse
Affiliation(s)
- Marco Capecchi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (M.A.); (I.M.)
| | - Alessandro Ciavarella
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (M.A.); (I.M.)
| | - Andrea Artoni
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (M.A.); (I.M.)
| | - Maria Abbattista
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (M.A.); (I.M.)
| | - Ida Martinelli
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (M.A.); (I.M.)
| |
Collapse
|
172
|
Benito-Garcia F, Pires I, Lima J. Aspirin Desensitization: Implications for Acetylsalicylic Acid-Sensitive Pregnant Women. ACTA ACUST UNITED AC 2021; 57:medicina57040390. [PMID: 33920569 PMCID: PMC8073510 DOI: 10.3390/medicina57040390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022]
Abstract
Low-dose acetylsalicylic acid (ASA) is widely used during pregnancy to prevent obstetric complications of placental dysfunction, such as preeclampsia, stillbirth and fetal growth restriction, and obstetric complications in pregnant women with antiphospholipid syndrome. ASA-sensitive pregnant women cannot benefit from the effects of ASA due to the possibility of severe or potentially life-threatening hypersensitivity reactions to ASA. ASA desensitization is a valuable and safe therapeutic option for these women, allowing them to start daily prophylaxis with ASA and prevent pregnancy complications. The authors discuss the recent advances in obstetric conditions preventable by ASA and the management of ASA hypersensitivity in pregnancy, including ASA desensitization. To encourage the implementation of ASA desensitization protocols in ASA-sensitive pregnant women, they also propose a practical approach for use in daily clinical practice.
Collapse
Affiliation(s)
| | - Inês Pires
- São Bernardo Hospital, Centro Hospitalar de Setúbal, 2910-549 Setúbal, Portugal;
| | - Jorge Lima
- Department of Obstetrics and Gynecology, CUF Descobertas Hospital, 1998-018 Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
- Correspondence: ; Tel.: +351-962-617-741
| |
Collapse
|
173
|
Müller-Calleja N, Hollerbach A, Royce J, Ritter S, Pedrosa D, Madhusudhan T, Teifel S, Meineck M, Häuser F, Canisius A, Nguyen TS, Braun J, Bruns K, Etzold A, Zechner U, Strand S, Radsak M, Strand D, Gu JM, Weinmann-Menke J, Esmon CT, Teyton L, Lackner KJ, Ruf W. Lipid presentation by the protein C receptor links coagulation with autoimmunity. Science 2021; 371:371/6534/eabc0956. [PMID: 33707237 DOI: 10.1126/science.abc0956] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/15/2020] [Accepted: 01/15/2021] [Indexed: 12/17/2022]
Abstract
Antiphospholipid antibodies (aPLs) cause severe autoimmune disease characterized by vascular pathologies and pregnancy complications. Here, we identify endosomal lysobisphosphatidic acid (LBPA) presented by the CD1d-like endothelial protein C receptor (EPCR) as a pathogenic cell surface antigen recognized by aPLs for induction of thrombosis and endosomal inflammatory signaling. The engagement of aPLs with EPCR-LBPA expressed on innate immune cells sustains interferon- and toll-like receptor 7-dependent B1a cell expansion and autoantibody production. Specific pharmacological interruption of EPCR-LBPA signaling attenuates major aPL-elicited pathologies and the development of autoimmunity in a mouse model of systemic lupus erythematosus. Thus, aPLs recognize a single cell surface lipid-protein receptor complex to perpetuate a self-amplifying autoimmune signaling loop dependent on the cooperation with the innate immune complement and coagulation pathways.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Anne Hollerbach
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Jennifer Royce
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Svenja Ritter
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Thati Madhusudhan
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Sina Teifel
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Myriam Meineck
- Department of Medicine I, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Friederike Häuser
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - T Son Nguyen
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Johannes Braun
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Kai Bruns
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Anna Etzold
- Institute of Human Genetics, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Senckenberg Zentrum für Humangenetik, 60314 Frankfurt, Germany
| | - Ulrich Zechner
- Institute of Human Genetics, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Senckenberg Zentrum für Humangenetik, 60314 Frankfurt, Germany
| | - Susanne Strand
- Department of Medicine I, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Markus Radsak
- Department of Medicine III, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Dennis Strand
- Department of Medicine I, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Jian-Ming Gu
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Julia Weinmann-Menke
- Department of Medicine I, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Charles T Esmon
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany. .,Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| |
Collapse
|
174
|
Dong RJ, Lei SY, Li J, Yang XP, Li YY, Zhang YG. Thrombotic antiphospholipid syndrome in a child with human immunodeficiency virus: a rare case report. Thromb J 2021; 19:20. [PMID: 33766024 PMCID: PMC7992315 DOI: 10.1186/s12959-021-00273-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antiphospholipid syndrome (APS) is a non-inflammatory autoimmune disorder induced by antiphospholipid antibodies, which occurs exceedingly rarely in pediatric population and even more rarely reported in HIV positive children. A case of 11 years old boy had a sudden onset of swelling in his left lower leg along with pain which were worsening gradually. Initially, topical ointment was applied for 1 month which were ineffective in reducing pain and swelling. Instead, the symptoms were aggravated and suddenly spread to the proximal thigh, accompanied by dyskinesia of left lower leg. Both color doppler ultrasonography and vascular CT scan of left lower leg revealed deep venous thrombosis. His serum anti-phospholipid antibodies (aPLs) were tested positive. He was a known case of HIV virological failure with substantial HIV viral load (VL) despite receiving regular antiretroviral therapy (ART). His symptoms improved after giving aggressive antithrombotic and high dose corticosteroid treatments. CONCLUSION When pediatric patients develop thrombotic disease, APS also needs to be ruled out. The autoantibodies levels should be routinely tested to look for recurrent thrombosis in children with HIV/AIDS.
Collapse
Affiliation(s)
- Rong-Jing Dong
- Yunnan Provincial Hospital of Infectious Disease/Yunnan AIDS Care Center, Kunming, 650301, China.,Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Su-Yun Lei
- Yunnan Provincial Hospital of Infectious Disease/Yunnan AIDS Care Center, Kunming, 650301, China
| | - Jun Li
- College of Pharmaceutical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, 650500, China
| | - Xin-Ping Yang
- Yunnan Provincial Hospital of Infectious Disease/Yunnan AIDS Care Center, Kunming, 650301, China
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Yun-Gui Zhang
- Yunnan Provincial Hospital of Infectious Disease/Yunnan AIDS Care Center, Kunming, 650301, China.
| |
Collapse
|
175
|
Obermayer G, Afonyushkin T, Göderle L, Puhm F, Schrottmaier W, Taqi S, Schwameis M, Ay C, Pabinger I, Jilma B, Assinger A, Mackman N, Binder CJ. Natural IgM antibodies inhibit microvesicle-driven coagulation and thrombosis. Blood 2021; 137:1406-1415. [PMID: 33512411 DOI: 10.1182/blood.2020007155] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
Thrombosis and its associated complications are a major cause of morbidity and mortality worldwide. Microvesicles (MVs), a class of extracellular vesicles, are increasingly recognized as mediators of coagulation and biomarkers of thrombotic risk. Thus, identifying factors targeting MV-driven coagulation may help in the development of novel antithrombotic treatments. We have previously identified a subset of circulating MVs that is characterized by the presence of oxidation-specific epitopes and bound by natural immunoglobulin M (IgM) antibodies targeting these structures. This study investigated whether natural IgM antibodies, which are known to have important anti-inflammatory housekeeping functions, inhibit the procoagulatory properties of MVs. We found that the extent of plasma coagulation is inversely associated with the levels of both free and MV-bound endogenous IgM. Moreover, the oxidation epitope-specific natural IgM antibody LR04, which recognizes malondialdehyde adducts, reduced MV-dependent plasmatic coagulation and whole blood clotting without affecting thrombocyte aggregation. Intravenous injection of LR04 protected mice from MV-induced pulmonary thrombosis. Of note, LR04 competed the binding of coagulation factor X/Xa to MVs, providing a mechanistic explanation for its anticoagulatory effect. Thus, our data identify natural IgM antibodies as hitherto unknown modulators of MV-induced coagulation in vitro and in vivo and their prognostic and therapeutic potential in the management of thrombosis.
Collapse
Affiliation(s)
- Georg Obermayer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Taras Afonyushkin
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Laura Göderle
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Florian Puhm
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Soreen Taqi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Schwameis
- Department of Clinical Pharmacology
- Department of Emergency Medicine, and
| | - Cihan Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Ingrid Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | | | | | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
176
|
Sang Y, Roest M, de Laat B, de Groot PG, Huskens D. Interplay between platelets and coagulation. Blood Rev 2021; 46:100733. [PMID: 32682574 PMCID: PMC7354275 DOI: 10.1016/j.blre.2020.100733] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/12/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Haemostasis stops bleeding at the site of vascular injury and maintains the integrity of blood vessels through clot formation. This regulated physiological process consists of complex interactions between endothelial cells, platelets, von Willebrand factor and coagulation factors. Haemostasis is initiated by a damaged vessel wall, followed with a rapid adhesion, activation and aggregation of platelets to the exposed subendothelial extracellular matrix. At the same time, coagulation factors aggregate on the procoagulant surface of activated platelets to consolidate the platelet plug by forming a mesh of cross-linked fibrin. Platelets and coagulation mutually influence each other and there are strong indications that, thanks to the interplay between platelets and coagulation, haemostasis is far more effective than the two processes separately. Clinically this is relevant because impaired interaction between platelets and coagulation may result in bleeding complications, while excessive platelet-coagulation interaction induces a high thrombotic risk. In this review, platelets, coagulation factors and the complex interaction between them will be discussed in detail.
Collapse
Affiliation(s)
- Yaqiu Sang
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | - Mark Roest
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | - Bas de Laat
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | | | - Dana Huskens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands.
| |
Collapse
|
177
|
Abstract
Rhodomyrtone (Rom) is a plant-derived broad-spectrum antibiotic active against many Gram-positive pathogens. A single point mutation in the regulatory farR gene (farR*) confers resistance to Rom in Staphylococcus aureus (RomR). The mutation in farR* alters the activity of the regulator, FarR*, in such a way that not only its own gene, farR*, but also the divergently transcribed farE gene and genes controlled by the global regulator, agr, are highly upregulated. Here, we show that mainly the upregulation of the fatty acid efflux pump FarE causes the RomR phenotype, as farE deletion in either the parent or the RomR strain (RomR ΔfarE) yielded hypersensitivity to Rom. Comparative lipidome analysis of the supernatant (exolipidomics) and the pellet fraction revealed that the RomR strain excreted about 10 times more phospholipids (PGs) than the parent strain or the ΔfarE mutants. Since the PG content in the supernatant (2,244 ng/optical density [OD]) was more than 100-fold higher than that of fatty acids (FA), we assumed that PG interacts with Rom, thereby abrogating its antimicrobial activity. Indeed, by static and dynamic light scattering (SLS and DLS) and isothermal titration calorimetry (ITC) analyses, we could demonstrate that both PG and Rom were vesicular and reacted with each other in milliseconds to form a 1:1.49 [Rom-PG(32:0), where PG(32:0) is PG with C32:0 lipids] complex. The binding is entropically driven and hence hydrophobic and of low specificity in nature. Our results indicate that the cytoplasmic membrane is the actual target of Rom, which is also in agreement with Rom's induced rapid collapse of the membrane potential and decreased membrane integrity. IMPORTANCE Antibiotic resistance is a growing public health problem, and alternative antibiotics are urgently needed. Rhodomyrtone (Rom), an antimicrobial compound originally isolated from Rhodomyrtus tomentosa, is active against multidrug-resistant Gram-positive pathogens. However, Rom-resistant (RomR) mutants occur with low frequency. In this study, we unraveled the underlying resistance mechanism, which is based on a point mutation in the farR regulator gene, causing overexpression of FarE, which most likely acts as a phospholipid (PG) efflux pump, as large amounts of PG were found in the supernatant and the pellet fraction. We show that PG can bind to Rom, thereby abrogating its antimicrobial activity. The direct interaction of Rom with PG suggests that Rom's actual target is the cytoplasmic membrane. Antibiotics that interact with PG are rare. Since Rom can be chemically synthesized, it serves as a lead compound for synthesis of improved variants.
Collapse
|
178
|
Current Promising Biomarkers and Methods in the Diagnostics of Antiphospholipid Syndrome: A Review. Biomedicines 2021; 9:biomedicines9020166. [PMID: 33567576 PMCID: PMC7914732 DOI: 10.3390/biomedicines9020166] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022] Open
Abstract
Antiphospholipid syndrome (APS) is a hypercoagulation condition associated with the incidence of heterogenic antiphospholipid antibodies (aPLs), which non-specifically affect hemostasis processes. APS is clinically manifested by recurrent arterial and venous thromboses and reproduction losses. The aPL antibodies, which may induce clinical manifestations of APS, include criteria antibodies anti-cardiolipin, anti-β2-glycoprotein-I, and lupus anticoagulant, but also non-criteria antibodies, for example anti-β2-glycoprotein-I domain I, anti-phosphatidylserine/prothrombin, anti-annexin V, and many others. APS occurs mostly in patients of younger and middle age, most frequently in females. Laboratory diagnostics of APS are quite difficult, as they include a wide spectrum of examining methods, which are based on various principles of detection and are performed using various laboratory techniques. The objective of the review is to describe the current state of potentially examined biomarkers and methods in APS diagnostics. The aforementioned biomarkers are lupus anticoagulant, anti-β2-glycoprotein-I, anti-cardiolipin, anti-β2-glycoprotein-I domain I, anti-phosphatidylserine/prothrombin, anti-β2-glycoprotein-I IgA, anti-cardiolipin IgA, anti-annexin V and II, anti-prothrombin, anti-cardiolipin/vimentin, anti-protein S/protein C, and antibodies against phospholipid antigens for whose diagnostics we may use some of the methods established for a long time and some of the modern methods—the coagulation method for the determination of lupus anticoagulant (LA), enzyme-linked imunosorbent assay (ELISA), chemiluminescence analysis (CLIA), multiplex fluorescence flow immunoassay (MFFIA), fluorescence enzyme immunoassay (EliA), line immunoassay (LIA), multiline dot assay (MLDA), and thin-layer chromatography (TLC). Conclusion: Antibodies against phosphatidylethanolamine, phosphatidic acid, phosphatidylserine, phosphatidylinositol, cardiolipin/vimentin complex, and annexin V are currently the most studied new markers. However, these assays have not been standardized until now, both from the laboratory and clinical point of view. In this review we summarize the evidence of the most studied aPL markers and their potential clinical significance in seronegative APS (SN-APS).
Collapse
|
179
|
Abstract
Antiphospholipid syndrome (APS) is a thromboinflammatory disease with a variety of clinical phenotypes. Primary thrombosis prophylaxis should take an individualized risk stratification approach. Moderate-intensity vitamin K antagonist such as warfarin remains the primary strategy for secondary thrombosis prophylaxis among APS patients, especially for patients with predominantly venous disease. For now, direct oral anti-coagulants should be avoided in most APS patients, especially those with history of arterial manifestations. Obstetric APS management should be tailored based on an individual patient's antiphospholipid antibody profile, and obstetric and thrombotic history. Pharmacological agents beyond anticoagulants may be considered for the management of microthrombotic and nonthrombotic manifestations of APS, although more data are needed. A relatively recent discovery in the area of APS pathogenesis is the implication of neutrophil extracellular traps in thrombin generation and initiation of inflammatory cascades. APS is a complex thromboinflammatory disease with a broad clinical spectrum. Personalized therapy according to an individual's unique thrombosis and obstetric risk should be advocated.
Collapse
|
180
|
Sayar Z, Moll R, Isenberg D, Cohen H. Thrombotic antiphospholipid syndrome: A practical guide to diagnosis and management. Thromb Res 2021; 198:213-221. [PMID: 33485122 PMCID: PMC7560059 DOI: 10.1016/j.thromres.2020.10.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/12/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
Thrombotic antiphospholipid syndrome (APS) is characterised by venous, arterial and/or small vessel thrombosis in the context of persistently positive antiphospholipid antibodies (aPL). The diagnosis and management of thrombotic APS continues to prove challenging for clinicians. We provide a practical guide to the diagnosis of APS including who to test for aPL and which tests to do. We also consider clinical practice points on the management of venous, arterial and small vessel thrombosis, in the context of first and recurrent thrombotic events. Non-criteria manifestations of APS are reviewed. An approach to recurrent thrombosis and anticoagulant-refractory APS is discussed, with options including increasing the anticoagulation intensity of vitamin K antagonists, switching to low-molecular-weight-heparin, the use of fondaparinux and/or the addition of antiplatelet treatment. Adjunctive options such as vitamin D, hydroxychloroquine and statins are also addressed.
Collapse
Affiliation(s)
- Zara Sayar
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK; Department of Haematology, Whittington Health NHS Trust, London, UK.
| | - Rachel Moll
- Haemostasis Research Unit, Department of Haematology, University College London, London, UK
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, London, UK; Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hannah Cohen
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK; Haemostasis Research Unit, Department of Haematology, University College London, London, UK
| |
Collapse
|
181
|
Lee EE, Jun JK, Lee EB. Management of Women with Antiphospholipid Antibodies or Antiphospholipid Syndrome during Pregnancy. J Korean Med Sci 2021; 36:e24. [PMID: 33496084 PMCID: PMC7834901 DOI: 10.3346/jkms.2021.36.e24] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/12/2020] [Indexed: 01/10/2023] Open
Abstract
Antiphospholipid syndrome (APS), which is characterized by the presence of antiphospholipid antibodies (aPL), is associated with increased risk of thrombosis and obstetric complications, including preterm delivery and recurrent pregnancy losses. APS shows diverse clinical manifestations and the risk of complications varies among clinical subtypes. Although these patients are usually treated with aspirin and anticoagulants, the optimal treatment in various clinical settings is unclear, as the risk of complications vary among clinical subtypes and the management strategy depends on whether the patient is pregnant or not. Also, there are unmet needs for the evidence-based, pregnancy-related treatment of asymptomatic women positive for aPL. This review focuses on the management of positive aPL or APS in pregnant and postpartum women, and in women attempting to become pregnant. For asymptomatic aPL positive women, no treatment, low dose aspirin (LDA) or LDA plus anticoagulants can be considered during antepartum and postpartum. In obstetric APS patients, preconceptional LDA is recommended. LDA plus low molecular weight heparin is administered after confirmation of pregnancy. Vascular APS patients should take frequent pregnancy test and receive heparin instead of warfarin after confirmation of pregnancy. During pregnancy, heparin plus LDA is recommended. Warfarin can be restarted 4 to 6 hours after vaginal delivery and 6 to 12 hours after cesarean delivery. Most importantly, a tailored approach and patient-oriented treatment are mandatory.
Collapse
Affiliation(s)
- Eunyoung Emily Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.
| |
Collapse
|
182
|
Abstract
PURPOSE OF REVIEW Antiphospholipid syndrome (APS), more common than once believed, is an autoimmune disease best known for its high risk of incident and recurrent thrombotic events. The approach to treatment potentially differs from treatment of thrombosis in the general population, and this article endeavors to review the latest updates on this topic. RECENT FINDINGS The epidemiology of APS is being increasingly elucidated by large population-based studies, with APS perhaps affecting as many as 1 in 2000 individuals. Vitamin K antagonists, aspirin, and heparinoids continue to have obvious roles in the management of patients with APS. There has recently been intensive study of direct oral anticoagulants in APS, with the most recent randomized studies raising concerns about their inferiority to vitamin K antagonists, at least in some subgroups. Other approaches to treating APS beyond anticoagulants and antiaggregants are also receiving increased attention in mechanistic and preclinical studies with an eye toward future roles in patients with refractory and/or microvascular disease. Pediatric APS is identified as an area in desperate need of additional prospective research. SUMMARY Progress continues to be made in pursuit of improving the lives of individuals afflicted with APS. The most important future directions would seem to involve leveraging modern molecular technologies in order to improve subphenotyping of antiphospholipid antibody-positive individuals. This will help personalize risk profiles and ideally define the optimal approach to therapy based on future risk, rather than past morbid events.
Collapse
|
183
|
Artemisinin analogue SM934 protects against lupus-associated antiphospholipid syndrome via activation of Nrf2 and its targets. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1702-1719. [PMID: 33481164 DOI: 10.1007/s11427-020-1840-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/15/2020] [Indexed: 01/24/2023]
Abstract
Kidney is a major target organ in both antiphospholipid syndrome (APS) and systemic lupus erythematosus (SLE). The etiology of antiphospholipid syndrome nephropathy associated lupus nephritis (APSN-LN) is intricate and remains largely unrevealed. We proposed in present work, that generation of antiphospholipid antibodies (aPLs), especially those directed towards the oxidized neoepitopes, are largely linked with the redox status along with disease progression. Moreover, we observed that compromised antioxidative capacity coincided with turbulence of inflammatory cytokine profile in the kidney of male NZW×BXSB F1 mice suffered from APSN-LN. SM934 is an artemisinin derivative that has been proved to have potent immunosuppressive properties. In current study, we elaborated the therapeutic benefits of SM934 in male NZW×BXSB F1 mice, a murine model develops syndrome resembled human APS associated with SLE, for the first time. SM934 treatment comprehensively impeded autoantibodies production, inflammatory cytokine accumulation and excessive oxidative stress in kidney. Among others, we interpreted in present work that both anti-inflammatory and antioxidative effects of SM934 is closely correlated with the enhancement of Nrf2 signaling and expression of its targets. Collectively, our finding confirmed that therapeutic strategy simultaneously exerting antioxidant and anti-inflammatory efficacy provide a novel feasible remedy for treating APSN-LN.
Collapse
|
184
|
Doyle AJ, Breen KA, Hunt BJ. Antiphospholipid Syndrome with Monoclonal Gammopathy-A Mechanism for Recurrent Thrombosis? Thromb Haemost 2021; 121:1387-1390. [PMID: 33472254 DOI: 10.1055/a-1366-9379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Andrew J Doyle
- Centre for Thrombosis and Haemostasis, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Karen A Breen
- Centre for Thrombosis and Haemostasis, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Beverley J Hunt
- Centre for Thrombosis and Haemostasis, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
185
|
Khan MM, Zafar A, Shahid S. Antiphospholipid Syndrome presenting as Chronic Venous Insufficiency in a Young Male. Arch Rheumatol 2021; 36:615-616. [PMID: 35382357 PMCID: PMC8957763 DOI: 10.46497/archrheumatol.2021.8515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/28/2020] [Indexed: 11/03/2022] Open
Affiliation(s)
- Marium Marufia Khan
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Amara Zafar
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Sana Shahid
- Department of General Surgery, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
186
|
Devreese KMJ. Testing for antiphospholipid antibodies: Advances and best practices. Int J Lab Hematol 2021; 42 Suppl 1:49-58. [PMID: 32543059 DOI: 10.1111/ijlh.13195] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/24/2020] [Accepted: 03/10/2020] [Indexed: 01/19/2023]
Abstract
The diagnosis of antiphospholipid syndrome (APS) relies on the detection of circulating antiphospholipid antibodies (aPL). Currently, lupus anticoagulant (LAC), anticardiolipin (aCL), and antibeta2-glycoprotein I antibodies (aβ2GPI) IgG or IgM are included as laboratory criteria if persistently present. Progress has been made on the standardization of tests as guidelines on LAC testing and immunological assays for aCL and aβ2GPI are published. However, LAC measurement remains a complicated procedure with many pitfalls and interfered by anticoagulant therapy. Solid-phase assays for aCL and aβ2GPI still show interassay differences. These methodological issues make the laboratory diagnosis of APS challenging. In the interpretation of aPL results, antibody profiles help in identifying patients at risk. Noncriteria aPL, such as antibodies against the domain I of beta2-glycoprotein (aDI) and antiphosphatidylserine-prothrombin (aPS/PT) antibodies have been studied in the last years and may be useful in risk stratification of APS patients. But, aDI and aPS/PT are not included in the current diagnostic criteria and testing in daily practice is not recommended as these antibodies have no added value in the diagnosis of APS. This review will focus on the technical aspects of the laboratory methods, the clinical relevance of assays and interpretation of aPL results in the diagnosis of APS.
Collapse
Affiliation(s)
- Katrien M J Devreese
- Department of Laboratory Medicine, Coagulation Laboratory, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
187
|
Yu X, He L. Aspirin and heparin in the treatment of recurrent spontaneous abortion associated with antiphospholipid antibody syndrome: A systematic review and meta-analysis. Exp Ther Med 2021; 21:57. [PMID: 33365057 PMCID: PMC7716630 DOI: 10.3892/etm.2020.9489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to review relevant, randomized, controlled trials in order to determine the effects of aspirin and heparin treatment on recurrent spontaneous abortion (RSA) in women with antiphospholipid syndrome (APS). Previous relevant studies were identified using PubMed, Cochrane, Embase, CNKI, VANFUN and VIP by retrieving appropriate key words. Additionally, key relevant sources in the literature were reviewed and articles published before May 2019 were included. The 22 selected studies included 1,515 patients in the treatment group and 1,531 patients in the control group. These previous studies showed that heparin and aspirin significantly improved live birth rate when compared with treatments using intravenous immunoglobulin, aspirin alone or aspirin combined with prednisone. Moreover, heparin and aspirin greatly increased the birth weight compared with placebo and improved vaginal delivery relative to intravenous immunoglobulin. The gestational age at birth was significantly higher in the heparin and aspirin group compared with the placebo group and the incidence of intrauterine growth restriction was lower in the heparin and aspirin group compared with the placebo group. Furthermore, heparin and aspirin markedly reduced the incidence of miscarriage compared with the aspirin group and the placebo group, and the incidence of pre-eclampsia was lower in the heparin and aspirin group than the placebo group. Thus, heparin and aspirin could be further examined for the treatment of RSA in women with APS.
Collapse
Affiliation(s)
- Xiaomei Yu
- Department of Obstetrics, Ward 1, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Li He
- Department of Women's Health Care, Chongqing Health Center for Women and Children, Yubei, Chongqing 401147, P.R. China
| |
Collapse
|
188
|
Kakkos SK, Gohel M, Baekgaard N, Bauersachs R, Bellmunt-Montoya S, Black SA, Ten Cate-Hoek AJ, Elalamy I, Enzmann FK, Geroulakos G, Gottsäter A, Hunt BJ, Mansilha A, Nicolaides AN, Sandset PM, Stansby G, Esvs Guidelines Committee, de Borst GJ, Bastos Gonçalves F, Chakfé N, Hinchliffe R, Kolh P, Koncar I, Lindholt JS, Tulamo R, Twine CP, Vermassen F, Wanhainen A, Document Reviewers, De Maeseneer MG, Comerota AJ, Gloviczki P, Kruip MJHA, Monreal M, Prandoni P, Vega de Ceniga M. Editor's Choice - European Society for Vascular Surgery (ESVS) 2021 Clinical Practice Guidelines on the Management of Venous Thrombosis. Eur J Vasc Endovasc Surg 2020; 61:9-82. [PMID: 33334670 DOI: 10.1016/j.ejvs.2020.09.023] [Citation(s) in RCA: 383] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
189
|
Abstract
Recurrent pregnancy loss is a distressing pregnancy disorder experienced by ~2.5% of women trying to conceive. Recurrent pregnancy loss is defined as the failure of two or more clinically recognized pregnancies before 20-24 weeks of gestation and includes embryonic and fetal losses. The diagnosis of an early pregnancy loss is relatively straightforward, although progress in predicting and preventing recurrent pregnancy loss has been hampered by a lack of standardized definitions, the uncertainties surrounding the pathogenesis and the highly variable clinical presentation. The prognosis for couples with recurrent pregnancy loss is generally good, although the likelihood of a successful pregnancy depends on maternal age and the number of previous losses. Recurrent pregnancy loss can be caused by chromosomal errors, anatomical uterine defects, autoimmune disorders and endometrial dysfunction. Available treatments target the putative risk factors of pregnancy loss, although the effectiveness of many medical interventions is controversial. Regardless of the underlying aetiology, couples require accurate information on their chances of having a baby and appropriate support should be offered to reduce the psychological burden associated with multiple miscarriages. Future research must investigate the pathogenesis of recurrent pregnancy loss and evaluate novel diagnostic tests and treatments in adequately powered clinical trials.
Collapse
|
190
|
Gimenez BT, Cezarette GN, Bomfim ADS, Monteiro WM, Russo EMDS, Frantz FG, Sampaio SV, Sartim MA. Role of crotoxin in coagulation: novel insights into anticoagulant mechanisms and impairment of inflammation-induced coagulation. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200076. [PMID: 33293940 PMCID: PMC7702976 DOI: 10.1590/1678-9199-jvatitd-2020-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Snake venom phospholipases A2 (svPLA2) are
biologically active toxins, capable of triggering and modulating a wide
range of biological functions. Among the svPLA2s, crotoxin (CTX)
has been in the spotlight of bioprospecting research due to its role in
modulating immune response and hemostasis. In the present study, novel
anticoagulant mechanisms of CTX, and the modulation of inflammation-induced
coagulation were investigated. Methods: CTX anticoagulant activity was evaluated using platelet poor plasma (PPP)
and whole blood (WB), and also using isolated coagulation factors and
complexes. The toxin modulation of procoagulant and pro-inflammatory effects
was evaluated using the expression of tissue factor (TF) and cytokines in
lipopolysaccharide (LPS)-treated peripheral blood mononuclear cells (PBMC)
and in WB. Results: The results showed that CTX impaired clot formation in both PPP and WB, and
was responsible for the inhibition of both intrinsic (TF/factor VIIa) and
extrinsic (factor IXa/factor VIIIa) tenase complexes, but not for factor Xa
and thrombin alone. In addition, the PLA2 mitigated the
prothrombinase complex by modulating the coagulation phospholipid role in
the complex. In regards to the inflammation-coagulation cross talk, the
toxin was capable of reducing the production of the pro-inflammatory
cytokines IL-1β, IL-6 and TNF-α, and was followed by decreased levels of TF
and procoagulant activity from LPS-treated PBMC either isolated or in
WB. Conclusion: The results obtained in the present study recognize the toxin as a novel
medicinal candidate to be applied in inflammatory diseases with coagulation
disorders.
Collapse
Affiliation(s)
- Bruna Terada Gimenez
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Gabriel Neves Cezarette
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Aline de Sousa Bomfim
- Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Wuelton Marcelo Monteiro
- Tropical Medicine Graduate Program, Amazonas State University, Manaus, AM, Brazil.,Carlos Borborema Clinical Research Institute, Doutor Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, AM, Brazil
| | - Elisa Maria de Sousa Russo
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Fabiani Gai Frantz
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Marco Aurelio Sartim
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.,Basic and Applied Immunology Graduate Program, Institute of Biological Sciences, Federal University of Amazonas, Manaus, AM, Brazil
| |
Collapse
|
191
|
The Weight of IgA Anti-β2glycoprotein I in the Antiphospholipid Syndrome Pathogenesis: Closing the Gap of Seronegative Antiphospholipid Syndrome. Int J Mol Sci 2020; 21:ijms21238972. [PMID: 33255963 PMCID: PMC7730063 DOI: 10.3390/ijms21238972] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
The specific value of IgA Anti-β2glycoprotein I antibodies (aB2GP1) in the diagnosis and management of antiphospholipid syndrome (APS) is still controversial and a matter of active debate. The relevance of the IgA aB2GP1 isotype in the pathophysiology of APS has been increasingly studied in the last years. There is well know that subjects with multiple positive APS tests are at increased risk of thrombosis and/or miscarriage. However, these antibodies are not included in the 2006 APS classification criteria. Since 2010 the task force of the Galveston International Congress on APS recommends testing IgA aB2GP1 isotype in patients with APS clinical criteria in the absence of criteria antibodies. In this review, we summarize the molecular and clinical “state of the art” of the IgA aB2GP in the context of APS. We also discuss some of the characteristics that may help to evaluate the real value of the IgA aB2GP1 determination in basic research and clinical practice. The scientific community should be aware of the importance of clarifying the role of IgA aB2GP1 in the APS diagnosis.
Collapse
|
192
|
Misasi R, Longo A, Recalchi S, Caissutti D, Riitano G, Manganelli V, Garofalo T, Sorice M, Capozzi A. Molecular Mechanisms of "Antiphospholipid Antibodies" and Their Paradoxical Role in the Pathogenesis of "Seronegative APS". Int J Mol Sci 2020; 21:ijms21218411. [PMID: 33182499 PMCID: PMC7665122 DOI: 10.3390/ijms21218411] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
Antiphospholipid Syndrome (APS) is an autoimmune disease characterized by arterial and/or venous thrombosis and/or pregnancy morbidity, associated with circulating antiphospholipid antibodies (aPL). In some cases, patients with a clinical profile indicative of APS (thrombosis, recurrent miscarriages or fetal loss), who are persistently negative for conventional laboratory diagnostic criteria, are classified as "seronegative" APS patients (SN-APS). Several findings suggest that aPL, which target phospholipids and/or phospholipid binding proteins, mainly β-glycoprotein I (β-GPI), may contribute to thrombotic diathesis by interfering with hemostasis. Despite the strong association between aPL and thrombosis, the exact pathogenic mechanisms underlying thrombotic events and pregnancy morbidity in APS have not yet been fully elucidated and multiple mechanisms may be involved. Furthermore, in many SN-APS patients, it is possible to demonstrate the presence of unconventional aPL ("non-criteria" aPL) or to detect aPL with alternative laboratory methods. These findings allowed the scientists to study the pathogenic mechanism of SN-APS. This review is focused on the evidence showing that these antibodies may play a functional role in the signal transduction pathway(s) leading to thrombosis and pregnancy morbidity in SN-APS. A better comprehension of the molecular mechanisms triggered by aPL may drive development of potential therapeutic strategies in APS patients.
Collapse
|
193
|
Velasco-Rodríguez D, Laso RV, García-Raso A, Mahíllo-Fernández I, Guzmán-López K, Martín-Herrero S, Barral EJ, Vegas-Sánchez MDC, Martínez-Becerra MJ, de la Plaza R, Romero LFL, Mínguez D, Alonso-Domínguez JM, López CB, López AG, Fernández MSS, Llamas-Sillero P. Thrombin generation in subjects with lupus anticoagulant without prior thrombosis or gestational morbidities. Thromb Res 2020; 196:425-431. [PMID: 33038586 DOI: 10.1016/j.thromres.2020.09.025] [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] [Received: 01/28/2020] [Revised: 04/27/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Lupus anticoagulant (LA) can be a cause of thrombosis and/or pregnancy morbidities, producing antiphospholipid syndrome (APS). An increase in thrombin generation (TG) is correlated with prothrombotic status. Several changes in TG-derived parameters have been reported in APS patients. OBJECTIVES Evaluate whether the TG phenotype of APS can also be described in LA subjects without clinical manifestations of APS, and to investigate the possible influence of both LA potency and antiphospholipid (aPL) profile on it. RESULTS TG was analyzed in 153 cases of LA and 41 healthy controls. We have observed prolongation of both lag time (3.7 min vs 2.32 min, p < 0.001) and time to peak (6.48 min vs 5.27 min, p < 0.001), increased peak height (221.7 nM vs 182.7 nM, p < 0.001), slightly higher ETP (221.7 nM·min vs 182.7 nM·min, p = 0.041), and higher velocity index (100.7 nM/min vs 74.53 nM/min, p = 0.001) in LA subjects compared to controls. After adding thrombomodulin (TM), ETP%inh was significantly lower in LA group (37.90% vs 59.90%, p < 0.001) showing resistance to TM/activated protein C (APC). Significant differences were found in lag time, time to peak and ETP%inh according to the potency and aPL profile. CONCLUSIONS Previously described differences in TG-derived parameters in APS patients have been confirmed in incidental LA subjects: prolonged lag time and time to peak, slightly higher ETP, higher peak height, and less sensitivity to TM/APC. High LA potency and triple-positive aPL profile enhance differences in lag time, time to peak and, especially, increase APC resistance, but no effect in ETP was observed.
Collapse
Affiliation(s)
- Diego Velasco-Rodríguez
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain.
| | - Rosa Vidal Laso
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | - Aránzazu García-Raso
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | | | - Karina Guzmán-López
- Department of Immunology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Martín-Herrero
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | - Elena Jiménez Barral
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | | | | | - Reyes de la Plaza
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | | | - Dolores Mínguez
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | | | - Carlos Blas López
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | - Amanda García López
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
| | | | - Pilar Llamas-Sillero
- Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain; Department of Hematology, Hospitales Quirón públicos, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
194
|
Cohen H, Cuadrado MJ, Erkan D, Duarte-Garcia A, Isenberg DA, Knight JS, Ortel TL, Rahman A, Salmon JE, Tektonidou MG, Williams DJ, Willis R, Woller SC, Andrade D. 16th International Congress on Antiphospholipid Antibodies Task Force Report on Antiphospholipid Syndrome Treatment Trends. Lupus 2020; 29:1571-1593. [PMID: 33100166 PMCID: PMC7658424 DOI: 10.1177/0961203320950461] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Antiphospholipid syndrome (APS), an acquired autoimmune thrombophilia, is characterised by thrombosis and/or pregnancy morbidity in association with persistent antiphospholipid antibodies. The 16th International Congress on Antiphospholipid Antibodies Task Force on APS Treatment Trends reviewed the current status with regard to existing and novel treatment trends for APS, which is the focus of this Task Force report. The report addresses current treatments and developments since the last report, on the use of direct oral anticoagulants in patients with APS, antiplatelet agents, adjunctive therapies (hydroxychloroquine, statins and vitamin D), targeted treatment including rituximab, belimumab, and anti-TNF agents, complement inhibition and drugs based on peptides of beta-2-glycoprotein I. In addition, the report summarises potential new players, including coenzyme Q10, adenosine receptor agonists and adenosine potentiation. In each case, the report provides recommendations for clinicians, based on the current state of the art, and suggests a clinical research agenda. The initiation and development of appropriate clinical studies requires a focus on devising suitable outcome measures, including a disease activity index, an optimal damage index, and a specific quality of life index.
Collapse
Affiliation(s)
- Hannah Cohen
- Haemostasis Research Unit, Department of Haematology, University
College London, London, UK
- University College London Hospitals NHS Foundation Trust,
London, UK
| | - Maria J Cuadrado
- Rheumatology Department, Clinica Universidad de Navarra, Madrid,
Spain
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Disease, Hospital
for Special Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Ali Duarte-Garcia
- Division of Rheumatology, Department of Internal Medicine, Mayo
Clinic, Rochester, Minnesota, USA
- Robert D. and Patricia E. Kern Center for the Science of Health
Care Delivery, Mayo Clinic, Rochester, Minnesota, USA
| | - David A Isenberg
- University College London Hospitals NHS Foundation Trust,
London, UK
- Centre for Rheumatology, Division of Medicine, University
College London, London, UK
| | - Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor,
Michigan, USA
| | - Thomas L Ortel
- Division of Hematology, Department of Medicine, and Department
of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Anisur Rahman
- Centre for Rheumatology, Division of Medicine, University
College London, London, UK
| | - Jane E Salmon
- Division of Rheumatology, Hospital for Special surgery, Weill
Cornell Medicine, New York, NY, USA
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, National
and Kapodistrian University of Athens, Athens, Greece
| | - David J Williams
- University College London Hospitals NHS Foundation Trust,
London, UK
- UCL EGA Institute for Women’s Health, University College
London, London, UK
| | - Rohan Willis
- Antiphospholipid Standardization Laboratory, University of
Texas Medical Branch, Galveston, TX, USA
| | - Scott C Woller
- Department of Medicine, Intermountain Medical Center, Murray
UT; Division of General Internal Medicine, University of Utah School of
Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
195
|
Hasan Ali O, Bomze D, Risch L, Brugger SD, Paprotny M, Weber M, Thiel S, Kern L, Albrich WC, Kohler P, Kahlert CR, Vernazza P, Bühler PK, Schüpbach RA, Gómez-Mejia A, Popa AM, Bergthaler A, Penninger JM, Flatz L. Severe COVID-19 is associated with elevated serum IgA and antiphospholipid IgA-antibodies. Clin Infect Dis 2020; 73:e2869-e2874. [PMID: 32997739 PMCID: PMC7543315 DOI: 10.1093/cid/ciaa1496] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/29/2020] [Indexed: 01/15/2023] Open
Abstract
Background Severe coronavirus disease 2019 (COVID-19) frequently entails complications that bear similarities to autoimmune diseases. To date, there is little data on possible IgA-mediated autoimmune responses. Here, we aim to determine whether COVID-19 is associated with a vigorous total IgA response and if IgA antibodies are associated with complications of severe illness. Since thrombotic events are frequent in severe COVID-19 and resemble hypercoagulation of antiphospholipid syndrome (APS), our approach focused on antiphospholipid antibodies (aPL). Methods In this retrospective cohort study clinical data and aPL from 64 patients with COVID-19 were compared from three independent tertiary hospitals (one in Liechtenstein, two in Switzerland). Samples were collected from April 9 th to May 1 st, 2020. Results Clinical records of 64 patients with COVID-19 were reviewed and divided into a cohort with mild illness (mCOVID) (41%), a discovery cohort with severe illness (sdCOVID) (22%) and a confirmation cohort with severe illness (scCOVID) (38%). Total IgA, IgG and aPL were measured with clinical diagnostic kits. Severe illness was significantly associated with increased total IgA (sdCOVID, P=0.01; scCOVID, p-value<0.001), but not total IgG. Among aPL, both cohorts with severe illness significantly correlated with elevated anti-Cardiolipin IgA (sdCOVID and scCOVID, p-value<0.001), anti-Cardiolipin IgM (sdCOVID, P=0.003; scCOVID, P<0.001), and anti-Beta2 Glycoprotein-1 IgA (sdCOVID and scCOVID, P<0.001). Systemic lupus erythematosus was excluded from all patients as a potential confounder. Conclusions Higher total IgA and IgA-aPL were consistently associated with severe illness. These novel data strongly suggest that a vigorous antiviral IgA-response, possibly triggered in the bronchial mucosa, induces systemic autoimmunity.
Collapse
Affiliation(s)
- Omar Hasan Ali
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.,Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - David Bomze
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lorenz Risch
- Labormedizinisches Zentrum Dr. Risch, Vaduz, Liechtenstein.,Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Bern, Switzerland
| | - Silvio D Brugger
- Department of Infectious Diseases and Hospital Hygiene, University Hospital Zurich, Zurich, Switzerland
| | - Matthias Paprotny
- Department of General Internal Medicine, Landesspital Liechtenstein, Vaduz, Liechtenstein
| | - Myriam Weber
- Department of General Internal Medicine, Landesspital Liechtenstein, Vaduz, Liechtenstein
| | - Sarah Thiel
- Department of General Internal Medicine, Landesspital Liechtenstein, Vaduz, Liechtenstein
| | - Lukas Kern
- Department of Pulmonology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Werner C Albrich
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Philipp Kohler
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Infectious Diseases and Hospital Epidemiology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Pietro Vernazza
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Philipp K Bühler
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Reto A Schüpbach
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Alejandro Gómez-Mejia
- Department of Infectious Diseases and Hospital Hygiene, University Hospital Zurich, Zurich, Switzerland
| | - Alexandra M Popa
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andreas Bergthaler
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Josef M Penninger
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada.,IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Lukas Flatz
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.,Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
196
|
Kostopoulou M, Nikolopoulos D, Parodis I, Bertsias G. Cardiovascular Disease in Systemic Lupus Erythematosus: Recent Data on Epidemiology, Risk Factors and Prevention. Curr Vasc Pharmacol 2020; 18:549-565. [DOI: 10.2174/1570161118666191227101636] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Systemic Lupus Erythematosus (SLE) is associated with increased risk for accelerated atherosclerosis
and cardiovascular (CV) events including coronary heart disease, cerebrovascular and peripheral
artery disease. CV events occur both early and late during the disease course, with younger
patients being at much higher risk than age-matched counterparts. The risk cannot be fully accounted for
by the increased prevalence of traditional atherosclerotic factors and may be due to pathophysiologic
intermediates such as type I interferons and other inflammatory cytokines, oxidative stress, activated
granulocytes and production of extracellular chromatin traps, antiphospholipid and other autoantibodies
causing dysfunction of lipoproteins, altogether resulting in endothelial injury and pro-atherogenic
dyslipidaemia. These mechanisms may be further aggravated by chronic intake of prednisone (even at
doses <7.5 mg/day), whereas immunomodulatory drugs, especially hydroxychloroquine, may exert antiatherogenic
properties. To date, there is a paucity of randomized studies regarding the effectiveness of
preventative strategies and pharmacological interventions specifically in patients with SLE. Nevertheless,
both the European League Against Rheumatism recommendations and extrapolated evidence from
the general population emphasize that SLE patients should undergo regular monitoring for atherosclerotic
risk factors and calculation of the 10-year CV risk. Risk stratification should include diseaserelated
factors and accordingly, general (lifestyle modifications/smoking cessation, antihypertensive and
statin treatment, low-dose aspirin in selected cases) and SLE-specific (control of disease activity, minimization
of glucocorticoids, use of hydroxychloroquine) preventive measures be applied as appropriate.
Further studies will be required regarding the use of non-invasive tools and biomarkers for CV assessment
and of risk-lowering strategies tailored to SLE.
Collapse
Affiliation(s)
- Myrto Kostopoulou
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dionysis Nikolopoulos
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - George Bertsias
- Department of Rheumatology, Clinical Rheumatology and Allergy, University of Crete Medical School, Iraklio, Greece
| |
Collapse
|
197
|
Cerdà P, Becattini C, Iriarte A, Hernández JC, Corbella X, Riera-Mestre A. Direct oral anticoagulants versus vitamin K antagonists in antiphospholipid syndrome: A meta-analysis. Eur J Intern Med 2020; 79:43-50. [PMID: 32482595 DOI: 10.1016/j.ejim.2020.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Anticoagulant treatment is recommended in patients with thrombosis and antiphospholipid syndrome (APS). Conflicting results have been reported on the role of direct oral anticoagulants (DOACs) in these patients. We performed a meta-analysis of randomized controlled trials (RCTs) focused on this issue. METHODS We searched MEDLINE and EMBASE for RCTs comparing DOACs and vitamin K antagonists (VKAs) for secondary thromboprophylaxis in patients with thrombotic APS. The primary objective was to assess the efficacy of DOACs compared to VKAs to prevent recurrence of thromboembolic events. Risk difference (RD) was reported as weighted RD according to Mantel-Haenszel random-effect method. RESULTS Three RCTs (426 patients) were included, all comparing rivaroxaban with VKAs. The proportion of recurrences (either arterial or venous) was higher among rivaroxaban patients when compared with those receiving VKAs (9.5% vs 2.8%; RD 6%, 95% CI, -0.05 - 0.18, p=0.29), although non-statistically significant. In patients with an arterial index event, thromboembolic recurrences were more frequent in those treated with rivaroxaban compared to those treated with VKAs (25% vs 6.2%; RD 19%, 95% CI, 0.04 - 0.33; p =0.01; I2 49%). In triple aPL positive patients, rivaroxaban showed higher rates of thromboembolic recurrences compared with VKAs (12% vs 3%; RD 9%, 95% CI, 0.02 - 0.15; p= 0.01; I2 13%). Non-statistically significant differences were observed in major bleeding events or mortality. CONCLUSIONS The use of rivaroxaban in APS patients is associated with an increased rate of thromboembolic recurrences compared to VKAs, at least in those with arterial index event or triple aPL positivity.
Collapse
Affiliation(s)
- Pau Cerdà
- Internal Medicine Department, Hospital Universitari Bellvitge - IDIBELL, Barcelona, Spain
| | - Cecilia Becattini
- Internal and Cardiovascular Medicine - Stroke Unit, University of Perugia, Perugia, Italy
| | - Adriana Iriarte
- Internal Medicine Department, Hospital Universitari Bellvitge - IDIBELL, Barcelona, Spain
| | - Josefina Cortés Hernández
- Internal Medicine Department. Rheumatology Research Group. Vall d´Hebrón University Hospital Research Institute, Barcelona, Spain
| | - Xavier Corbella
- Internal Medicine Department, Hospital Universitari Bellvitge - IDIBELL, Barcelona, Spain; Faculty of Medicine and Health Sciences. Universitat Internacional de Catalunya, Barcelona, Spain
| | - Antoni Riera-Mestre
- Internal Medicine Department, Hospital Universitari Bellvitge - IDIBELL, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
198
|
Mantovani Cardoso E, Hundal J, Feterman D, Magaldi J. Concomitant new diagnosis of systemic lupus erythematosus and COVID-19 with possible antiphospholipid syndrome. Just a coincidence? A case report and review of intertwining pathophysiology. Clin Rheumatol 2020; 39:2811-2815. [PMID: 32720260 PMCID: PMC7384868 DOI: 10.1007/s10067-020-05310-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/16/2022]
Abstract
In the midst of the COVID-19 pandemic, further understanding of its complications points towards dysregulated immune response as a major component. Systemic lupus erythematosus (SLE) is also a disease of immune dysregulation leading to multisystem compromise. We present a case of new-onset SLE concomitantly with COVID-19 and development of antiphospholipid antibodies. An 18-year-old female that presented with hemodynamic collapse and respiratory failure, progressed to cardiac arrest, and had a pericardial tamponade drained. She then progressed to severe acute respiratory distress syndrome, severe ventricular dysfunction, and worsening renal function with proteinuria and hematuria. Further studies showed bilateral pleural effusions, positive antinuclear and antidouble-stranded DNA antibodies, lupus anticoagulant, and anticardiolipin B. C3 and C4 levels were low. SARS-Cov-2 PCR was positive after 2 negative tests. She also developed multiple deep venous thrombosis, in the setting of positive antiphospholipid antibodies and lupus anticoagulant. In terms of pathophysiology, COVID-19 is believed to cause a dysregulated cytokine response which could potentially be exacerbated by the shift in Th1 to Th2 response seen in SLE. Also, it is well documented that viral infections are an environmental factor that contributes to the development of autoimmunity; however, COVID-19 is a new entity, and it is not known if it could trigger autoimmune conditions. Additionally, it is possible that SARS-CoV-2, as it happens with other viruses, might lead to the formation of antiphospholipid antibodies, potentially contributing to the increased rates of thrombosis seen in COVID-19.
Collapse
MESH Headings
- Adolescent
- Anemia/etiology
- Antibodies, Anticardiolipin/immunology
- Antibodies, Antinuclear/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antiphospholipid Syndrome/complications
- Antiphospholipid Syndrome/diagnosis
- Antiphospholipid Syndrome/immunology
- Antiphospholipid Syndrome/therapy
- Anuria/etiology
- Betacoronavirus
- COVID-19
- Cardiac Tamponade/diagnostic imaging
- Cardiac Tamponade/etiology
- Cardiac Tamponade/therapy
- Complement C3/immunology
- Complement C4/immunology
- Coronavirus Infections/complications
- Coronavirus Infections/diagnosis
- Coronavirus Infections/immunology
- Coronavirus Infections/therapy
- DNA/immunology
- Echocardiography
- Fatal Outcome
- Female
- Heart Arrest/etiology
- Hematuria/etiology
- Humans
- Lupus Coagulation Inhibitor/immunology
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/complications
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/immunology
- Pandemics
- Patient Positioning
- Pericardiocentesis
- Pneumonia, Viral/complications
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/immunology
- Pneumonia, Viral/therapy
- Prone Position
- Proteinuria/etiology
- Renal Dialysis
- Renal Insufficiency/etiology
- Renal Insufficiency/therapy
- Respiration, Artificial
- Respiratory Distress Syndrome/etiology
- Respiratory Distress Syndrome/therapy
- Respiratory Insufficiency/etiology
- Respiratory Insufficiency/therapy
- SARS-CoV-2
- Thrombocytopenia/etiology
- Venous Thrombosis/etiology
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
Collapse
Affiliation(s)
| | - Jasmin Hundal
- Internal Medicine Residency, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Dominique Feterman
- Internal Medicine Residency, University of Connecticut School of Medicine, Farmington, CT, USA
| | | |
Collapse
|
199
|
Bitsadze V, Nalli C, Khizroeva J, Lini D, Andreoli L, Lojacono A, Fazzi E, Shoenfeld Y, Tincani A, Makatsariya A. "APS pregnancy - The offspring". Lupus 2020; 29:1336-1345. [PMID: 32752918 DOI: 10.1177/0961203320947154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Antiphospholipid antibody syndrome (APS) is an autoimmune disease that affects women in childbearing age. In recent years, great improvements were achieved in the management of pregnancies in these women. Prematurity could be an issue in these pregnancies, mainly due to the direct pathogenic effect of antiphospholipid antibodies (aPL) on the placental surface. Maternal IgG aPL can cross the placenta and theoretically interact with the growing fetus; it could reach the fetal brain because of the incompleteness of the fetal blood-brain barrier: whether this can have an effect on brain development is still debated. Neonatal thrombosis episodes have been described in children positive for aPL, not always associated with maternal antibody positivity, suggesting the hypothesis of a possible aPL de novo synthesis in fetus and neonates. METHODS A keyword-based literature search was conducted. We also described a case of neonatal catastrophic antiphospholipid syndrome (CAPS). RESULTS Offspring of patients with APS are generally healthy but the occurrence of neonatal thrombosis or minor neurological disorders were reported. CONCLUSIONS The limited number of the available data on this sensitive issue supports the need for further studies. Clinical follow-up of children of mothers with APS seems to be important to exclude, in the neonatal period, the occurrence of aPL associated pathological events such as thrombosis, and in the long-term, impairment in learning skills or behavioral problems.
Collapse
Affiliation(s)
- Viktoria Bitsadze
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Cecilia Nalli
- Rheumatology and Immunology Unit, ASST Spedali Civili, Brescia, Italy
| | - Jamilya Khizroeva
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Daniele Lini
- Rheumatology and Immunology Unit, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Laura Andreoli
- Rheumatology and Immunology Unit, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Andrea Lojacono
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Obstetric and Gynecology Unit, ASST Spedali Civili, Brescia, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Child Neurology and Psychiatry Unit, ASST Spedali Civili, Brescia, Italy
| | - Yehuda Shoenfeld
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.,Department of Medicine 'B', The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Angela Tincani
- Rheumatology and Immunology Unit, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Alexander Makatsariya
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
200
|
Hillarp A, Strandberg K, Gustafsson KM, Lindahl TL. Unveiling the complex effects of direct oral anticoagulants on dilute Russell's viper venom time assays. J Thromb Haemost 2020; 18:1866-1873. [PMID: 32294291 DOI: 10.1111/jth.14829] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Dilute Russell viper venom time (dRVVT) assays can be affected by direct oral anticoagulants (DOACs), which may cause false-positive results. However, there are conflicting results indicating significant differences between different reagents and DOACs. OBJECTIVES To evaluate the effect of DOACs on dRVVT assays. MATERIAL AND METHODS Samples were prepared by adding DOAC (dabigatran, rivaroxaban, apixaban, or edoxaban) to pooled normal plasma in the concentration range 0 to 800 µg/L. Six integrated dRVVT reagents were used, all composed of a screen assay (low phospholipid content) and a confirm assay (high phospholipid content). The screen/confirm dRVVT results were expressed as normalized ratios. To further evaluate the observed differences between tests and DOACs, addition of synthetic phospholipids was used. RESULTS The dRVVT ratios increased dose dependently for all DOACs, with four of the six tests and the DOAC rivaroxaban having the greatest effect. With one test, the ratios were almost unaffected with increasing DOAC concentration, whereas another test revealed a negative dose dependency for all DOACs. Variable DOAC effects can be explained by different effects on dRVVT screen and confirm clotting time. Adding synthetic phospholipids to samples containing rivaroxaban resulted in greatly reduced screen clotting times and thereby lower calculated dRVVT ratios. CONCLUSIONS There is a great variability in the dRVVT test result with different DOACs. The dRVVT ratios are unaffected for some reagents and this can be explained by an equal dose-dependent effect on both screen and confirm assays. The phospholipid type and content of the different reagents may contribute to the observed differences.
Collapse
Affiliation(s)
- Andreas Hillarp
- Department of Clinical Chemistry and Transfusion Medicine, Halland County Hospital, Halmstad, Sweden
| | - Karin Strandberg
- Clinical Chemistry, University and Regional Laboratories Region Skåne, Malmö, Sweden
| | - Kerstin M Gustafsson
- Department of Biomedical and Clinical Sciences, Clinical Chemistry, Linköping University, Linköping, Sweden
| | - Tomas L Lindahl
- Department of Biomedical and Clinical Sciences, Clinical Chemistry, Linköping University, Linköping, Sweden
| |
Collapse
|