1
|
Kurien BT, Ice JA, Wood RA, Pharaoh G, Cavett J, Lewis V, Bhaskaran S, Rasmussen A, Lessard CJ, Farris AD, Sivilis K, Koelsch KA, Van Remmen H, Scofield RH. Mitochondrial dysfunction and fatigue in Sjögren's disease. RMD Open 2025; 11:e005046. [PMID: 40274303 PMCID: PMC12020762 DOI: 10.1136/rmdopen-2024-005046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/03/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Sjögren's disease (SjD) is a chronic exocrine disorder typified by inflammation and dryness, but also profound fatigue, suggesting a pathological basis in cellular bioenergetics. In healthy states, dysfunctional mitochondria are recycled by mitophagic processes; when impaired, poorly functioning mitochondria persist and produce inflammatory reactive oxygen species. Employing a case-control study, we tested our hypothesis that mitochondrial dysregulation in T cells is associated with fatigue in SjD. METHODS We isolated pan T cells from peripheral blood mononuclear cells of 13 SjD and 4 non-Sjögren's sicca (NSS) subjects, who completed several fatigue questionnaires, along with 8 healthy subjects. Using Seahorse, we analysed T cells for mitochondrial oxygen consumption rate (OCR) and extracellular acidification rate, which we assessed for correlation with fatigue measures. Using public microarray data available for 190 SjD and 32 healthy subjects, we identified a mitophagic transcriptional signature that stratified SjD patients into 5 discrete clusters. Comparisons between the SjD subjects in these clusters to healthy individuals identified differentially expressed transcripts, which we subjected to bioinformatic interrogation. RESULTS Basal OCR, ATP-linked respiration, maximal respiration and reserve capacity were significantly lower in SjD and NSS subjects compared with healthy individuals, with no differences in non-mitochondrial respiration, basal glycolysis or glycolytic reserve. Scores related to a sleep questionnaire and Bowman's Profile of Fatigue and Discomfort showed correlation with altered OCR in SjD. Subgroup differential expression analysis revealed dynamic transcriptional activity between mitophagy subgroups, expanding the number of differentially expressed transcripts tenfold. CONCLUSIONS Mitochondrial dysfunction and fatigue are significant problems in SjD warranting further investigation.
Collapse
Affiliation(s)
- Biji T Kurien
- Research, US Department of Veterans Affairs, Oklahoma City, Oklahoma, USA
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - John Aubrey Ice
- Research, US Department of Veterans Affairs, Oklahoma City, Oklahoma, USA
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Rebecca A Wood
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Gavin Pharaoh
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Joshua Cavett
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Valerie Lewis
- Research, US Department of Veterans Affairs, Oklahoma City, Oklahoma, USA
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Astrid Rasmussen
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Christopher J Lessard
- Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - A Darise Farris
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kathy Sivilis
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kristi A Koelsch
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Holly Van Remmen
- Research, US Department of Veterans Affairs, Oklahoma City, Oklahoma, USA
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Robert Hal Scofield
- Research, US Department of Veterans Affairs, Oklahoma City, Oklahoma, USA
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
2
|
Celia AI, Galli M, Mancuso S, Alessandri C, Frati G, Sciarretta S, Conti F. Antiphospholipid Syndrome: Insights into Molecular Mechanisms and Clinical Manifestations. J Clin Med 2024; 13:4191. [PMID: 39064231 PMCID: PMC11277906 DOI: 10.3390/jcm13144191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Antiphospholipid syndrome (APS) is a complex systemic autoimmune disorder characterized by a hypercoagulable state, leading to severe vascular thrombosis and obstetric complications. The 2023 ACR/EULAR guidelines have revolutionized the classification and understanding of APS, introducing broader diagnostic criteria that encompass previously overlooked cardiac, renal, and hematologic manifestations. Despite these advancements, diagnosing APS remains particularly challenging in seronegative patients, where traditional tests fail, yet clinical symptoms persist. Emerging non-criteria antiphospholipid antibodies offer promising new diagnostic and management avenues for these patients. Managing APS involves a strategic balance of cardiovascular risk mitigation and long-term anticoagulation therapy, though the use of direct oral anticoagulants remains contentious due to varying efficacy and safety profiles. This article delves into the intricate pathogenesis of APS, explores the latest classification criteria, and evaluates cutting-edge diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandra Ida Celia
- Rheumatology, Department of Clinical Internal, Anesthesiological e Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.I.C.); (S.M.); (C.A.); (F.C.)
| | - Mattia Galli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.F.); (S.S.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| | - Silvia Mancuso
- Rheumatology, Department of Clinical Internal, Anesthesiological e Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.I.C.); (S.M.); (C.A.); (F.C.)
| | - Cristiano Alessandri
- Rheumatology, Department of Clinical Internal, Anesthesiological e Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.I.C.); (S.M.); (C.A.); (F.C.)
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.F.); (S.S.)
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.F.); (S.S.)
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Fabrizio Conti
- Rheumatology, Department of Clinical Internal, Anesthesiological e Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.I.C.); (S.M.); (C.A.); (F.C.)
| |
Collapse
|
3
|
Kurien BT, Ice JA, Wood R, Pharaoh G, Cavett J, Lewis V, Bhaskaran S, Rasmussen A, Lessard CJ, Farris AD, Sivils KL, Koelsch KA, Van Remmen H, Scofield RH. Mitochondrial Dysfunction and Fatigue in Sjögren's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.598269. [PMID: 38948768 PMCID: PMC11212898 DOI: 10.1101/2024.06.17.598269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Objectives Sjögren's disease (SjD) is a common exocrine disorder typified by chronic inflammation and dryness, but also profound fatigue, suggesting a pathological basis in cellular bioenergetics. In healthy states, damaged or dysfunctional mitochondrial components are broken down and recycled by mitophagy, a specialized form of autophagy. In many autoimmune disorders, however, evidence suggests that dysfunctional mitophagy allows poorly functioning mitochondria to persist and contribute to a cellular milieu with elevated reactive oxygen species. We hypothesized that mitophagic processes are dysregulated in SjD and that dysfunctional mitochondria contribute to overall fatigue. We sought to link fatigue with mitochondrial dysfunction directly in SjD, heretofore unexamined, and further sought to assess the pathogenic extent and implications of dysregulated mitophagy in SjD. Methods We isolated pan T cells via negative selection from the peripheral blood mononuclear cells of 17 SjD and 8 age-matched healthy subjects, all of whom completed fatigue questionnaires prior to phlebotomy. Isolated T cells were analyzed for mitochondrial oxygen consumption rate (OCR) and glycolysis using Seahorse, and linear correlations with fatigue measures were assessed. A mitophagy transcriptional signature in SjD was identified by reanalysis of whole-blood microarray data from 190 SjD and 32 healthy subjects. Differential expression analyses were performed by case/control and subgroup analyses comparing SjD patients by mitophagy transcriptional cluster against healthy subjects followed by bioinformatic interpretation using gene set enrichment analysis. Results Basal OCR, ATP-linked respiration, maximal respiration, and reserve capacity were significantly lower in SjD compared to healthy subjects with no observed differences in non-mitochondrial respiration, basal glycolysis, or glycolytic stress. SjD lymphocytic mitochondria show structural alterations compared to healthy subjects. Fatigue scores related to pain/discomfort in SjD correlated with the altered OCR. Results from subgroup analyses by mitophagic SjD clusters revealed highly variable inter-cluster differentially expressed genes (DEGs) and expanded the number of SjD-associated gene targets by tenfold within the same dataset. Conclusion Mitochondrial dysfunction, associated with fatigue, is a significant problem in SjD and warrants further investigation.
Collapse
|
4
|
Du X, Ma X, Tan Y, Shao F, Li C, Zhao Y, Miao Y, Han L, Dang G, Song Y, Yang D, Deng Z, Wang Y, Jiang C, Kong W, Feng J, Wang X. B cell-derived anti-beta 2 glycoprotein I antibody mediates hyperhomocysteinemia-aggravated hypertensive glomerular lesions by triggering ferroptosis. Signal Transduct Target Ther 2023; 8:103. [PMID: 36907919 PMCID: PMC10008839 DOI: 10.1038/s41392-023-01313-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/14/2022] [Accepted: 01/09/2023] [Indexed: 03/14/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) is a risk factor for chronic kidney diseases (CKDs) that affects about 85% CKD patients. HHcy stimulates B cells to secrete pathological antibodies, although it is unknown whether this pathway mediates kidney injury. In HHcy-treated 2-kidney, 1-clip (2K1C) hypertensive murine model, HHcy-activated B cells secreted anti-beta 2 glycoprotein I (β2GPI) antibodies that deposited in glomerular endothelial cells (GECs), exacerbating glomerulosclerosis and reducing renal function. Mechanistically, HHcy 2K1C mice increased phosphatidylethanolamine (PE) (18:0/20:4, 18:0/22:6, 16:0/20:4) in kidney tissue, as determined by lipidomics. GECs oxidative lipidomics validated the increase of oxidized phospholipids upon Hcy-activated B cells culture medium (Hcy-B CM) treatment, including PE (18:0/20:4 + 3[O], PE (18:0a/22:4 + 1[O], PE (18:0/22:4 + 2[O] and PE (18:0/22:4 + 3[O]). PE synthases ethanolamine kinase 2 (etnk2) and ethanolamine-phosphate cytidylyltransferase 2 (pcyt2) were increased in the kidney GECs of HHcy 2K1C mice and facilitated polyunsaturated PE synthesis to act as lipid peroxidation substrates. In HHcy 2K1C mice and Hcy-B CM-treated GECs, the oxidative environment induced by iron accumulation and the insufficient clearance of lipid peroxides caused by transferrin receptor (TFR) elevation and down-regulation of SLC7A11/glutathione peroxidase 4 (GPX4) contributed to GECs ferroptosis of the kidneys. In vivo, pharmacological depletion of B cells or inhibition of ferroptosis mitigated the HHcy-aggravated hypertensive renal injury. Consequently, our findings uncovered a novel mechanism by which B cell-derived pathogenic anti-β2GPI IgG generated by HHcy exacerbated hypertensive kidney damage by inducing GECs ferroptosis. Targeting B cells or ferroptosis may be viable therapeutic strategies for ameliorating lipid peroxidative renal injury in HHcy patients with hypertensive nephropathy.
Collapse
Affiliation(s)
- Xing Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Ying Tan
- Department of Nephrology, Peking University First Hospital, 100034, Beijing, P. R. China
| | - Fangyu Shao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, 100083, Beijing, P. R. China
| | - Yutong Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Lulu Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Guohui Dang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Yuwei Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Dongmin Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Zhenling Deng
- Department of Nephrology, Peking University Third Hospital, 100083, Beijing, P. R. China
| | - Yue Wang
- Department of Nephrology, Peking University Third Hospital, 100083, Beijing, P. R. China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China.
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 100191, Beijing, P. R. China.
| |
Collapse
|
5
|
Foulquier N, Le Dantec C, Bettacchioli E, Jamin C, Alarcón‐Riquelme ME, Pers J. Machine Learning for the Identification of a Common Signature for Anti-SSA/Ro 60 Antibody Expression Across Autoimmune Diseases. Arthritis Rheumatol 2022; 74:1706-1719. [PMID: 35635731 PMCID: PMC9804576 DOI: 10.1002/art.42243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Anti-Ro autoantibodies are among the most frequently detected extractable nuclear antigen autoantibodies, mainly associated with primary Sjögren's syndrome (SS), systemic lupus erythematosus (SLE), and undifferentiated connective tissue disease (UCTD). This study was undertaken to determine if there is a common signature for all patients expressing anti-Ro 60 autoantibodies regardless of their disease phenotype. METHODS Using high-throughput multiomics data collected from the cross-sectional cohort in the PRECISE Systemic Autoimmune Diseases (PRECISESADS) study Innovative Medicines Initiative (IMI) project (genetic, epigenomic, and transcriptomic data, combined with flow cytometry data, multiplexed cytokines, classic serology, and clinical data), we used machine learning to assess the integrated molecular profiling of 520 anti-Ro 60+ patients compared to 511 anti-Ro 60- patients with primary SS, patients with SLE, and patients with UCTD, and 279 healthy controls. RESULTS The selected clinical features for RNA-Seq, DNA methylation, and genome-wide association study data allowed for a clear distinction between anti-Ro 60+ and anti-Ro 60- patients. The different features selected using machine learning from the anti-Ro 60+ patients constituted specific signatures when compared to anti-Ro 60- patients and healthy controls. Remarkably, the transcript Z score of 3 genes (ATP10A, MX1, and PARP14), presenting with overexpression associated with hypomethylation and genetic variation and independently identified using the Boruta algorithm, was clearly higher in anti-Ro 60+ patients compared to anti-Ro 60- patients regardless of disease type. Our findings demonstrated that these signatures, enriched in interferon-stimulated genes, were also found in anti-Ro 60+ patients with rheumatoid arthritis and those with systemic sclerosis and remained stable over time and were not affected by treatment. CONCLUSION Anti-Ro 60+ patients present with a specific inflammatory signature regardless of their disease type, suggesting that a dual therapeutic approach targeting both Ro-associated RNAs and anti-Ro 60 autoantibodies should be considered.
Collapse
Affiliation(s)
- Nathan Foulquier
- B Lymphocytes, Autoimmunity and Immunotherapies laboratory, UMR 1227Université de Brest, INSERMBrestFrance
| | - Christelle Le Dantec
- B Lymphocytes, Autoimmunity and Immunotherapies laboratory, UMR 1227Université de Brest, INSERMBrestFrance
| | - Eleonore Bettacchioli
- B Lymphocytes, Autoimmunity and Immunotherapies laboratory, UMR 1227Université de Brest, INSERMBrestFrance
| | - Christophe Jamin
- B Lymphocytes, Autoimmunity and Immunotherapies laboratory, UMR 1227Université de Brest, INSERM, and University Hospital of BrestBrestFrance
| | | | - Jacques‐Olivier Pers
- B Lymphocytes, Autoimmunity and Immunotherapies laboratory, UMR 1227Université de Brest, INSERM, and University Hospital of BrestBrestFrance
| |
Collapse
|
6
|
Linge CP, Jern A, Tydén H, Gullstrand B, Yan H, Welinder C, Kahn R, Jönssen A, Semple JW, Bengtsson AA. Enrichment of complement, immunoglobulins and autoantibody targets in the proteome of platelets from patients with Systemic Lupus Erythematosus (SLE). Thromb Haemost 2022; 122:1486-1501. [PMID: 35419777 PMCID: PMC9420555 DOI: 10.1055/a-1825-2915] [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] [Indexed: 11/04/2022]
Abstract
Background
Systemic lupus erythematosus (SLE) is a complex disease characterized by autoimmunity toward apoptotic cells, excessive amounts of circulating immune complexes, and complement activation. A decreased platelet size has been observed in SLE and their nonhemostatic functions may play an active role in the disease. The main objective of this study was to find clues that could explain their decreased size and functional role, analyzing the entire platelet proteome.
Methods
Platelets were isolated from 23 patients with SLE. The five individuals with the highest and lowest average platelet forward scatter were selected for further analysis. Platelet protein content was analyzed using liquid chromatography with tandem mass spectrometry (LC-MS/MS) and compared with platelets from five healthy controls. Data are available via ProteomeXchange with identifier PXD031202.
Results
Out of 2,572 proteins identified, 396 had significantly different levels (ANOVA
q
-value ≤ 0.01). Forty proteins, including immunoglobulin-, complement- and phosphatidylserine-binding proteins had higher abundance in platelets from SLE patients, largely independent of size (fold difference of ≥1.5 and a
t
-test
p
-value of ≤0.05 as cut-off). Functional characterization revealed increased degranulation and skewed hemostatic balance in platelets from SLE patients. In the SLE proteome, immunoglobulin proteins were negatively correlated to serum complement C3 and C4 and the highest relative levels were detected in platelets of normal size.
Conclusion
Platelets from SLE patients shared a specific protein profile, including immunoglobulins, complement proteins, and autoantigens, largely independent of the platelet size and in agreement with an integrated role for platelets in SLE.
Collapse
Affiliation(s)
- Carl Petrus Linge
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Andreas Jern
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Helena Tydén
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Hong Yan
- BioMS, Swedish National Infrastructure for Biological Mass Spectrometry, Lund, Sweden
| | - Charlotte Welinder
- Department of Clinical Sciences Lund, Lund University Department of Oncology and Pathology, Lund, Sweden
| | - Robin Kahn
- Wallenberg Center for Molecular Medicin, Lund University Faculty of Medicine, Lund, Sweden.,Paediatrics, Lund University Faculty of Medicine, Lund, Sweden
| | - Andreas Jönssen
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - John W Semple
- Transfusion Medicine, Lunds Universitet, Lund, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| |
Collapse
|
7
|
Abisror N, Nguyen Y, Marozio L, Esteve Valverde E, Udry S, Pleguezuelo DE, Billoir P, Mayer-Pickel K, Urbanski G, Zigon P, De Moreuil C, Hoxha A, Bezanahary H, Carbillon L, Kayem G, Bornes M, Yelnik C, Johanet C, Nicaise-Roland P, Lambert M, Salle V, Latino OJ, Hachulla E, Benedetto C, Bourrienne MC, Benhamou Y, Alijotas-Reig J, Fain O, Mekinian A. Obstetrical outcome and treatments in seronegative primary APS: data from European retrospective study. RMD Open 2021; 6:0. [PMID: 32848089 PMCID: PMC7507995 DOI: 10.1136/rmdopen-2020-001340] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/27/2020] [Accepted: 07/23/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE To compare characteristics, pregnancies and treatments during pregnancies of seronegative and seropositive antiphospholipid syndrome (APS), to analyse factors associated with obstetrical outcome. PATIENTS AND METHODS Inclusion criteria were: (1) thrombotic and/or obstetrical APS (Sydney criteria); (2) absence of conventional antiphospholipid antibodies (APL); (3) at least one persistent non-conventional APL among IgA anticardiolipin antibodies, IgA anti-B2GPI, anti-vimentin G/M, anti-annexin V G/M, anti-phosphatidylethanolamine G/M and anti-phosphatidylserine/prothrombin G/M antibodies. The exclusion criteria were: (1) systemic lupus erythematosus ( SLE) or SLE-like disease; and (2) other connective tissue disease. RESULTS A total of 187 women (mean 33±5 years) with seronegative APS were included from 14 centres in Austria, Spain, Italy, Slovenia and France and compared with 285 patients with seropositive APS. Seronegative APS has more obstetrical rather than thrombotic phenotypes, with only 6% of venous thrombosis in comparison to seropositive APS. Cumulative incidence of adverse obstetrical events was similar in seronegative and seropositive APS patients, although higher rates of intrauterine deaths (15% vs 5%; p=0.03), of preeclampsia (7% vs 16%, p=0.048) and lower live birth term (36±3 vs 38±3 weeks of gestation; p=0.04) were noted in seropositive APS. The cumulative incidence of adverse obstetrical events was significantly improved in treated versus untreated seronegative APS (log rank<0.05), whereas there was no difference between patients who received aspirin or aspirin-low-molecular weighted heparin combination. CONCLUSION Several non-criteria APL can be detected in patients with clinical APS features without any conventional APL, with various rates. The detection of non-criteria APL and thus the diagnosis of seronegative APS could discuss the therapeutic management similar to seropositive APS, but well-designed controlled studies are necessary.
Collapse
Affiliation(s)
- Noemie Abisror
- Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France
| | - Yann Nguyen
- Department of Internal Medicine, AP-HP Nord, Beaujon Hospital, Paris University,Clichy, France
| | - Luca Marozio
- Department of Surgical Sciences, Obstetrics and Gynecology, University of Torino, Turin, Italy
| | | | - Sebastian Udry
- 5Autoimmune, Thrombophilic Diseases and Pregnancy Section, Acute Hospital "Dr Carlos G Durand", Buenos Aires, Argentina
| | | | - Paul Billoir
- Department of Internal Medicine, Vascular and Thrombosis Unit, Rouen University Hospital, Normandie University, UNIROUEN, INSERM, Rouen, France
| | | | - Geoffrey Urbanski
- Service Department of internal medicine, CHU d'Angers, Angers, France
| | - Polona Zigon
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Claire De Moreuil
- Département de Médecine Vasculaire, Médecine Interne et Pneumologie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France.,EA 3878, GETBO, Université Bretagne Loire, Brest Cedex, France
| | - Ariela Hoxha
- Department of Medicine-DIMED, Rheumatology Unit, University of Padua, Padua, Italy
| | - Holy Bezanahary
- Department of Internal Medicine, University Hospital of Limoges, Limoges, France
| | - Lionel Carbillon
- Department of Obstetrics, Gynecology and Reproductive Medicine Centers, Hôpitaux Universitaires Paris Seine Saint-Denis, Assistance Publique-Hôpitaux de Paris, Bondy, France.,University of Paris 13, Sorbonne University,Bobigny, France
| | - Gilles Kayem
- Service de Gynécologie Obstétrique, Hôpital Trousseau, AP-HP, Paris, France.,Sorbonne Université,Paris, France
| | - Marie Bornes
- Department of Gynaecology and Obstetrics and Reproductive Medicine, Tenon Hospital, Assistance Publique - Paris Hospitals,Paris, France
| | - Cecile Yelnik
- Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest (CeRAINO), Univ. Lille, CHU Lille, Lille, France
| | | | | | - Marc Lambert
- Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest (CeRAINO), Univ. Lille, CHU Lille, Lille, France
| | - Valéry Salle
- Department of Internal Medicine, University Hospital of Amiens, Amiens, France
| | - Omar Jose Latino
- 5Autoimmune, Thrombophilic Diseases and Pregnancy Section, Acute Hospital "Dr Carlos G Durand", Buenos Aires, Argentina
| | - Eric Hachulla
- Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest (CeRAINO), Univ. Lille, CHU Lille, Lille, France
| | - Chiara Benedetto
- Department of Surgical Sciences, Obstetrics and Gynecology, University of Torino, Turin, Italy
| | - Marie Charlotte Bourrienne
- Université de Paris, INSERM UMR_S1148, Paris cedex 18, France.,Laboratoire d'Hématologie, AP-HP, Hôpital Bichat, Paris cedex 18, France
| | - Ygal Benhamou
- Department of Internal Medicine, Vascular and Thrombosis Unit, Rouen University Hospital, Normandie University, UNIROUEN, INSERM, Rouen, France
| | - Jaume Alijotas-Reig
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine-1, Vall d'Hebron University Hospital, Barcelona, Spain.,Department of Medicine, Faculty of Medicine, Universitat Autönoma de Barcelona, Barcelona, Spain
| | - Olivier Fain
- Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France
| | - Arsène Mekinian
- Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France
| | | |
Collapse
|
8
|
Shao F, Miao Y, Zhang Y, Han L, Ma X, Deng J, Jiang C, Kong W, Xu Q, Feng J, Wang X. B cell-derived anti-beta 2 glycoprotein I antibody contributes to hyperhomocysteinaemia-aggravated abdominal aortic aneurysm. Cardiovasc Res 2021; 116:1897-1909. [PMID: 31782769 DOI: 10.1093/cvr/cvz288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/09/2019] [Accepted: 11/27/2019] [Indexed: 01/29/2023] Open
Abstract
AIMS Overactivated B cells secrete pathological antibodies, which in turn accelerate the formation of abdominal aortic aneurysms (AAAs). Hyperhomocysteinaemia (HHcy) aggravates AAA in mice; however, the underlying mechanisms remain largely elusive. In this study, we further investigated whether homocysteine (Hcy)-activated B cells produce antigen-specific antibodies that ultimately contribute to AAA formation. METHODS AND RESULTS ELISA assays showed that HHcy induced the secretion of anti-beta 2 glycoprotein I (anti-β2GPI) antibody from B cells both in vitro and in vivo. Mechanistically, Hcy increased the accumulation of various lipid metabolites in B cells tested by liquid chromatography-tandem mass spectrometry, which contributed to elevated anti-β2GPI IgG secretion. By using the toll-like receptor 4 (TLR4)-specific inhibitor TAK-242 or TLR4-deficient macrophages, we found that culture supernatants from Hcy-activated B cells and HHcy plasma IgG polarized inflammatory macrophages in a TLR4-dependent manner. In addition, HHcy markedly increased the incidence of elastase- and CaPO4-induced AAA in male BALB/c mice, which was prevented in μMT mice. To further determine the importance of IgG in HHcy-aggravated AAA formation, we purified plasma IgG from HHcy or control mice and then transferred the IgG into μMT mice, which were subsequently subjected to elastase- or CaPO4-induced AAA. Compared with μMT mice that received plasma IgG from control mice, μMT mice that received HHcy plasma IgG developed significantly exacerbated elastase- or CaPO4-induced AAA accompanied by increased elastin degradation, MMP2/9 expression, and anti-β2GPI IgG deposition in vascular lesions, as shown by immunofluorescence histochemical staining. CONCLUSION Our findings reveal a novel mechanism by which Hcy-induced B cell-derived pathogenic anti-β2GPI IgG might, at least in part, contribute to HHcy-aggravated chronic vascular inflammation and AAA formation.
Collapse
Affiliation(s)
- Fangyu Shao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Yutong Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Lulu Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Jiacheng Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Qingbo Xu
- Cardiovascular Division, Cardiology Department, BHF Center for Vascular Regeneration, King's College London, London, UK
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
9
|
Barrera MJ, Aguilera S, Castro I, Carvajal P, Jara D, Molina C, González S, González MJ. Dysfunctional mitochondria as critical players in the inflammation of autoimmune diseases: Potential role in Sjögren's syndrome. Autoimmun Rev 2021; 20:102867. [PMID: 34118452 DOI: 10.1016/j.autrev.2021.102867] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/11/2021] [Indexed: 12/13/2022]
Abstract
Relevant reviews highlight the association between dysfunctional mitochondria and inflammation, but few studies address the contribution of mitochondria and mitochondria-endoplasmic reticulum (ER) contact sites (MERCs) to cellular homeostasis and inflammatory signaling. The present review outlines the important role of mitochondria in cellular homeostasis and how dysfunctional mitochondrion can release and misplace mitochondrial components (cardiolipin, mitochondrial DNA (mtDNA), and mitochondrial formylated peptides) through multiple mechanisms. These components can act as damage-associated molecular patterns (DAMPs) and induce an inflammatory response via pattern recognition receptors (PRRs). Accumulation of damaged ROS-generating mitochondria, accompanied by the release of mitochondrial DAMPs, can activate PRRs such as the NLRP3 inflammasome, TLR9, cGAS/STING, and ZBP1. This process would explain the chronic inflammation that is observed in autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), type I diabetes (T1D), and Sjögren's syndrome. This review also provides a comprehensive overview of the importance of MERCs to mitochondrial function and morphology, cellular homeostasis, and the inflammatory response. MERCs play an important role in calcium homeostasis by mediating the transfer of calcium from the ER to the mitochondria and thereby facilitating the production of ATP. They also contribute to the synthesis and transfer of phospholipids, protein folding in the ER, mitochondrial fission, mitochondrial fusion, initiation of autophagosome formation, regulation of cell death/survival signaling, and regulation of immune responses. Therefore, alterations within MERCs could increase inflammatory signaling, modulate ER stress responses, cell homeostasis, and ultimately, the cell fate. This study shows severe ultrastructural alterations of mitochondria in salivary gland cells from Sjögren's syndrome patients for the first time, which could trigger alterations in cellular bioenergetics. This finding could explain symptoms such as fatigue and malfunction of the salivary glands in Sjögren's syndrome patients, which would contribute to the chronic inflammatory pathology of the disease. However, this is only a first step in solving this complex puzzle, and several other important factors such as changes in mitochondrial morphology, functionality, and their important contacts with other organelles require further in-depth study. Future work should focus on detecting the key milestones that are related to inflammation in patients with autoimmune diseases, such as Sjögren´s syndrome.
Collapse
Affiliation(s)
- María-José Barrera
- Facultad de Odontología, Universidad San Sebastián, Bellavista 7, Santiago, 8420524, Chile
| | | | - Isabel Castro
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Patricia Carvajal
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Jara
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudio Molina
- Facultad de Odontología, Universidad San Sebastián, Bellavista 7, Santiago, 8420524, Chile
| | - Sergio González
- Escuela de Odontología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - María-Julieta González
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Avidity of anti-phospholipid antibodies in relation to their levels. Cent Eur J Immunol 2021; 45:136-143. [PMID: 33456323 PMCID: PMC7792445 DOI: 10.5114/ceji.2020.97901] [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: 01/14/2019] [Accepted: 05/07/2019] [Indexed: 11/30/2022] Open
Abstract
Introduction The heterogeneity of anti-phospholipid antibodies can be manifested not only in different antigenic specificities, but also in their avidities. The aim of the study was to investigate the relationship between anti-cardiolipin antibody (aCL) IgG avidities and levels within the range of their titres, from very low to high ones. Material and methods We analyzed 78 serum samples from 60 patients by ELISA with chaotropic agents, using urea concentration of 6 and 8 mol/l and single diluted serum samples. The changes of aCL levels and avidities were explored during a long-term follow-up in 14 patients. Results The avidities of aCLs did not differ in the groups of patients classified according to aCL levels. The higher avidity antibodies predominated in our patients and the fluctuation of avidities in the longitudinal follow-up did not show significant differences. No relationship between aCL levels and their avidities was found. Conclusions aCL avidities seem to have no relationship with aCL levels and high-avidity aCLs; the potentially deleterious effects might be present also in patients with low and extremely low aCL levels. Avidity of aCLs belongs to stable characteristics with insignificant changes in time.
Collapse
|
11
|
Salle V. [Seronegative antiphospholipid syndrome: Myth or reality?]. Rev Med Interne 2020; 41:265-274. [PMID: 32115196 DOI: 10.1016/j.revmed.2020.02.005] [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: 07/05/2019] [Revised: 12/23/2019] [Accepted: 02/02/2020] [Indexed: 10/24/2022]
Abstract
The antiphospholipid syndrome (APS) is a systemic autoimmune disease characterized by thrombosis and/or obstetrical manifestations and the persistent presence, at least 12 weeks apart, of antiphospholipid antibodies (aPL) such as lupus anticoagulant (LA) and/or anticardiolipin antibodies (ACL) and/or anti-β2 glycoprotein I antibodies (aβ2GPI). The finding of patients with clinical profile highly suggestive of APS but who are negative for conventional biological criteria has led to the concept of seronegative APS. In the last few years, new antigen targets and methodological approaches have been employed to more clearly identify this syndrome in patients with thrombosis or obstetrical complications without conventional aPL. Although seronegative APS is still controversial, there is increasing recognition of the existence of this subgroup. However, clinical relevance of non conventional aPL need to be confirmed by efforts toward standardizing new biological tools and longitudinal studies involving large cohort of patients.
Collapse
Affiliation(s)
- V Salle
- Service de médecine interne, CHU Amiens-Picardie, 1, place Victor-Pauchet, 80054 Amiens cedex 1, France; Laboratoire de biochimie recherche, CHU Amiens-Picardie, Amiens, France.
| |
Collapse
|
12
|
|
13
|
Conti F, Andreoli L, Crisafulli F, Mancuso S, Truglia S, Tektonidou MG. Does seronegative obstetric APS exist? "pro" and "cons". Autoimmun Rev 2019; 18:102407. [PMID: 31639518 DOI: 10.1016/j.autrev.2019.102407] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 12/27/2022]
Abstract
Antiphospholipid Syndrome (APS) is the commonest treatable cause of recurrent miscarriage and pharmacological treatment of pregnant patients with antiphospholipid antibodies (aPL) should aim at preventing obstetric complications and maternal thrombotic events. Conventional treatment for patients with an established diagnosis of obstetric APS (OAPS), generally resulting in over 70-80% successful pregnancies. Since seropositive (SP)-APS and seronegative (SN)-APS patients had shown similar clinical profiles, patients with SN- OAPS, as well as SP-OAPS, should receive combined treatment in order to improve the pregnancy prognosis; indeed, current standard of care increased good pregnancy outcome in SN-APS, with similar effect to confirmed APS. The above data suggest that there are patients with the clinical manifestations of OAPS but persistently negative to conventional aPL that need to be identified to ensure adequate therapy and therefore a better prognosis. The clinical utility of non-criteria aPL in the diagnosis of SN-APS is still a matter of debate. In the last decade more and more studies have reported the presence of patients suffering from SN-APS in which non-conventional ("non-criteria") aPL might be present or antibodies may be detected using methodological approaches different from the traditional assays. To improve test standardization large prospective, multicenter, and multinational studies are needed. Therefore, when assessing a patient with clinical manifestations consistent with OAPS but aPL negative using the conventional available assays, the clinician should consider the possibility that the patient is affected with SN-APS.
Collapse
Affiliation(s)
- Fabrizio Conti
- Lupus Clinic, Rheumatology Unit, Dipartimento Di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Laura Andreoli
- Rheumatology and Clinical Immunology, Spedali Civili, Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesca Crisafulli
- Rheumatology and Clinical Immunology, Spedali Civili, Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Mancuso
- Lupus Clinic, Rheumatology Unit, Dipartimento Di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Simona Truglia
- Lupus Clinic, Rheumatology Unit, Dipartimento Di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy.
| | - Maria G Tektonidou
- Head of Rheumatology Unit, First Department of Propaedeutic Internal Medicine, School of Medicine, National University of Athens, Greece
| |
Collapse
|
14
|
Heikal N, Martins TB, White SK, Willis R, Ware Branch D, Schmidt RL, Tebo AE. Laboratory Evaluation of Antiphospholipid Syndrome. Am J Clin Pathol 2019; 152:638-646. [PMID: 31305881 DOI: 10.1093/ajcp/aqz085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Anti-β2 glycoprotein I domain I (anti-domain I) and anti-phosphatidylserine/prothrombin (aPS/PT) antibodies are present in patients with antiphospholipid syndrome (APS); however, their use in evaluation remains unclear. METHODS Diagnostic attributes of lupus anticoagulant (LAC), anti-domain I IgG, anti-cardiolipin, anti-β2 glycoprotein I (anti-β2GPI), and aPS/PT IgG and IgM antibodies were assessed in 216 patients evaluated for APS. RESULTS LAC had the best odds ratio (OR, 14.2) while that for anti-domain 1 IgG was comparable to anti-β2GPI IgG (OR, 8.3 vs 9.4) but higher than all others. Significant correlations were observed for thrombosis (P = .03) and pregnancy-related morbidity (P = .001) with anti-domain IgG and for any thrombosis with aPS/PT IgG (P = .006). Use of noncriteria antiphospholipid with or without criteria markers did not significantly increase the probability to diagnose APS. CONCLUSIONS Noncriteria tests can contribute to diagnosis and stratification of APS but do not improve diagnostic yield. Optimal strategies for implementation require prospective investigation.
Collapse
Affiliation(s)
- Nahla Heikal
- ARUP Institute of Clinical and Experimental Pathology, University of Utah School of Medicine, Salt Lake City
- Department of Pathology, University of Utah School of Medicine, Salt Lake City
| | - Thomas B Martins
- ARUP Institute of Clinical and Experimental Pathology, University of Utah School of Medicine, Salt Lake City
| | - Sandra K White
- Department of Pathology, University of Utah School of Medicine, Salt Lake City
| | - Rohan Willis
- Division of Rheumatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston
| | - D Ware Branch
- Division of Maternal-Fetal Medicine, University of Utah School of Medicine, Salt Lake City
- Intermountain Healthcare, Salt Lake City, UT
| | - Robert L Schmidt
- ARUP Institute of Clinical and Experimental Pathology, University of Utah School of Medicine, Salt Lake City
- Department of Pathology, University of Utah School of Medicine, Salt Lake City
| | - Anne E Tebo
- ARUP Institute of Clinical and Experimental Pathology, University of Utah School of Medicine, Salt Lake City
- Department of Pathology, University of Utah School of Medicine, Salt Lake City
| |
Collapse
|
15
|
Hollingsworth TJ, Radic MZ, Beranova-Giorgianni S, Giorgianni F, Wang Y, Iannaccone A. Murine Retinal Citrullination Declines With Age and is Mainly Dependent on Peptidyl Arginine Deiminase 4 (PAD4). Invest Ophthalmol Vis Sci 2019; 59:3808-3815. [PMID: 30073354 PMCID: PMC6074612 DOI: 10.1167/iovs.18-24118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Citrullination is a post-translational modification (PTM) that serves many normal physiological functions. Studies have shown that this PTM—along with expression of the catalyzing enzymes, peptidyl arginine deiminases (PADs)—are increased in autoimmune and age-related pathologies. PAD2 retinal expression has been previously documented in rat and human. Herein, we report on the expression levels and patterns of PAD2, PAD4, and retinal citrullination in the murine retina with age. Methods Wild-type (WT) and Pad4-/- (PAD4KO) mice ages 0.5, 0.75, 1, 3, 6, and 9 months were investigated after euthanasia and eye enucleation. Retinal lysates from 3-month-old mice were probed for PAD4 by western blot. Whole eyes were fixed, cryosectioned, and probed using an anti-PAD2/4 antibody (Ab), a specific anti-PAD4 Ab, and F95 anti-citrullinated peptide Ab. Fluorescent intensities were quantified with ImageJ. Results WT retinas show different levels of PAD4 expression in distinct retinal layers, with the most intense labeling in inner retinal layers, while PAD4KO mice lacked retinal PAD4. Using a nonspecific anti-PAD2/4 Ab, PAD reactivity observed in PAD4KO mice was attributed to PAD2. In WT, both PAD2 and PAD4 expression levels decrease significantly with age while low-level residual PAD2 expression was seen in PAD4KO mice. Citrullination levels in WT retinas paralleled PAD4 expression, with PAD4KO mice exhibiting consistently minimal citrullination. Conclusions Both PAD2 and PAD4 expression and citrullination decrease with age in the murine retina. However, in the absence of PAD4, retinal citrullination is nearly abolished, indicating that PAD4 is a main effector for retinal citrullination under physiological conditions.
Collapse
Affiliation(s)
- T J Hollingsworth
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Marko Z Radic
- Department of Microbiology, Biochemistry and Immunology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Sarka Beranova-Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Francesco Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Yanming Wang
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States
| | - Alessandro Iannaccone
- Duke University School of Medicine, Duke Eye Center, Department of Ophthalmology, Durham, North Carolina, United States
| |
Collapse
|
16
|
Litvinova E, Darnige L, Kirilovsky A, Burnel Y, de Luna G, Dragon-Durey MA. Prevalence and Significance of Non-conventional Antiphospholipid Antibodies in Patients With Clinical APS Criteria. Front Immunol 2018; 9:2971. [PMID: 30619328 PMCID: PMC6302212 DOI: 10.3389/fimmu.2018.02971] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
Background: The biological diagnostics of antiphospholipid syndrome (APS) takes into account the persistent positivity for anticardiolipin and/or anti-β2GP1 antibodies and/or presence of lupus anticoagulant (LA). However, some non-conventional antiphospholipid antibodies have emerged that could help in the diagnosis of APS. Objectives: To study the potential usefulness of non-conventional antiphospholipid antibodies in clinical practice. Methods: Eighty-seven patients, aged from 15 to 92 years were included and classified in following groups: 41 patients positive for the conventional antibodies with clinical criterion of APS (31 with primary APS and 10 secondary), 17 seronegative APS (SNAPS) patients (i.e., persistent negativity for the conventional antibodies with a strong clinical suspicion of APS), 11 asymptomatic antiphospholipid antibodies carriers (i.e., persistent positivity for the conventional antibodies without clinical evidence of APS), and 18 patients presenting with a first thrombotic or obstetrical event. IgG and IgM were detected to the following antigens: phosphatidylserine/prothrombin (PS/PT) by ELISA, and phosphatidic acid, phosphatidyl-ethanolamine, phosphatidyl-glycerol, phosphatidyl-inositol, phosphatidylserine, annexin V, prothrombin by immunodot. Anti-β2GP1 IgA, and anti-β2GP1 domain 1 IgG were detected by chemiluminescence. Results: Positivity for the non-conventional antibodies was correlated with APS severity; patients with catastrophic APS (CAPS) being positive for 10.7 (Median, Range: 5–14) non-conventional antibodies. 9/17 seronegative patients were positive for at least one of non-conventional antibodies. A study of non-supervised hierarchical clustering of all markers revealed that anti-PS/PT antibodies showed high correlation with the presence of LA. All patients with APS triple positivity (highest risk profile) exhibited also persistent positivity for anti-PS/PT antibodies. Conclusions: Our data obtained from a prospective cohort constituted mainly by patients with primary APS, suggest that non-conventional APS antibodies may be useful for patients classified as SNAPS. They demonstrate the potential value of aPS/PT antibodies as a strong marker of APS. We propose that anti-PS/PT antibodies could be a surrogate APS biological marker of LA to classify in high-risk profile patients treated by direct oral anticoagulants (DOACs), in whom LA detection cannot be achieved.
Collapse
Affiliation(s)
| | - Luc Darnige
- Department of Biological Haematology, HEGP, APHP, Paris, France.,Inserm UMR-S1140, Paris, France.,Paris Descartes University, Paris, France
| | | | - Yann Burnel
- Department of Biological Haematology, HEGP, APHP, Paris, France
| | | | | |
Collapse
|
17
|
Alessandri C, Agmon-Levin N, Conti F, Perricone C, Ortona E, Pendolino M, Capozzi A, Delunardo F, Mancini R, Truglia S, Spinelli FR, Ceccarelli F, Sorice M, Shoenfeld Y, Valesini G. Anti-mutated citrullinated vimentin antibodies in antiphospholipid syndrome: diagnostic value and relationship with clinical features. Immunol Res 2018; 65:524-531. [PMID: 28215033 DOI: 10.1007/s12026-017-8899-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Antiphospholipid antibodies (aPLs) are a heterogeneous group of autoantibodies essential for the diagnosis of antiphospholipid syndrome (APS) but do not predict clinical manifestations or disease progression. Hence, the co-presence of other antibodies may prove useful. Autoimmunity directed toward vimentin and other citrullinated peptides was established in rheumatoid arthritis (RA) and in other autoimmune conditions including systemic lupus erythematosus (SLE). We have previously described the presence of autoantibodies directed against vimentin/cardiolipin complex in patients with antiphospholipid syndrome (APS), but there are no data on the role of citrullinated vimentin in APS. Thus, we evaluated the prevalence and clinical significance of anti-MCV in APS patients. The study group consisted of 79 unselected outpatients with APS. Control groups included 25 patients with SLE, 30 patients with RA, and 20 healthy subjects age- and sex-matched. To detect anti-MCV, anti-vimentin, anti-vimentin/cardiolipin, and anti-CCP2 antibodies, commercial or homemade enzyme-linked immunosorbent assays (ELISA) were performed. Anti-MCV antibodies were found in a high percentage of APS patients (26.6%). A significant correlation between anti-MCV and anti-vimentin/cardiolipin serum levels was observed (p = 0.029). Moreover, vimentin reactivity was increased by its citrullination or conjugation with cardiolipin (p = 0.01 and p < 0.001, respectively). Interestingly, anti-MCV was found associated with the presence of arthritis (p = 0.011) and anti-vimentin/cardiolipin was highly specific for the presence of arterial or venous thrombosis in APS (p = 0.003 and p = 0.002, respectively). The detection of additional autoantibodies may contribute to clinical assessment of APS patients. Citrullination may occur in APS and play a role in the pathogenesis of this condition. KEY POINTS •Anti-MCV antibodies can be found in APS patients and are associated with the presence of arthritis. •Anti-vimentin/cardiolipin is strongly associated with the presence of thrombosis (both arterial and venous). •Citrullination occurs in APS, participate in disease pathogenesis, and influence clinical picture.
Collapse
Affiliation(s)
- Cristiano Alessandri
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Nancy Agmon-Levin
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Fabrizio Conti
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Carlo Perricone
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy.
| | - Elena Ortona
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Pendolino
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Antonella Capozzi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Federica Delunardo
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Rome, Italy
| | - Riccardo Mancini
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Simona Truglia
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Francesca Romana Spinelli
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Maurizio Sorice
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Guido Valesini
- Lupus Clinic, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
18
|
Zohoury N, Bertolaccini ML, Rodriguez-Garcia JL, Shums Z, Ateka-Barrutia O, Sorice M, Norman GL, Khamashta M. Closing the Serological Gap in the Antiphospholipid Syndrome: The Value of "Non-criteria" Antiphospholipid Antibodies. J Rheumatol 2017; 44:1597-1602. [PMID: 28864642 DOI: 10.3899/jrheum.170044] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Most clinicians use the 2006 Sydney classification criteria to evaluate patients suspected of having antiphospholipid syndrome (APS). Although sensitive and specific for APS, many patients fulfilling clinical criteria for the syndrome are persistently negative for the specific serological tests ("laboratory criteria"). These "seronegative APS" (SN-APS) patients can go undiagnosed and untreated until they experience serious clinical events. This study's objective was to describe antibody profiles of SN-APS patients using non-criteria markers, assess the clinical utility of these markers separately and in combination, and suggest incorporation into guidelines for patients suspected of APS. METHODS We categorized 175 consecutive patients suspected of APS into 2 subgroups: 107 fulfilling Sydney APS classification for seropositive APS (SP-APS) and 68 with clinical manifestations suggestive of APS but having negative serology, on 2 occasions, for criteria markers (SN-APS). On study inclusion, samples were retested for criteria and 11 non-criteria markers, including antiphosphatidylserine/prothrombin antibodies. RESULTS Using 4 of 11 non-criteria tests, a cumulative 30.9% of SN-APS patients were detected. Combining results of all 11 non-criteria tests, 25 SN-APS (36.8%) and 89 SP-APS (83.2%) were positive for 1 or more non-criteria antibodies. CONCLUSION Failure to diagnose APS can result in severe clinical consequences. Patients displaying clinical features of APS, but negative for conventional criteria markers, should undergo additional testing for non-criteria biomarkers. In our cohort, around one-third of SN-APS patients showed reactivity to 1 or more non-criteria markers. An update to the current classification criteria incorporating new serological markers should be considered to identify and stratify patients with APS for more effective treatment and management.
Collapse
Affiliation(s)
- Navid Zohoury
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy. .,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics. .,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital.
| | - Maria Laura Bertolaccini
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Jose Luis Rodriguez-Garcia
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Zakera Shums
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Oier Ateka-Barrutia
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Maurizio Sorice
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Gary L Norman
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| | - Munther Khamashta
- From Inova Diagnostics Inc., San Diego, California, USA; Academic Department of Vascular Surgery, King's College London; Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital, London, UK; Department of Internal Medicine, University of Castilla-La Mancha, Albacete, Spain; Department of Experimental Medicine, La Sapienza University, Rome, Italy.,Navid Zohoury, Zakera Shums, and Gary L. Norman are employees of Inova Diagnostics.,N. Zohoury, BS, Inova Diagnostics Inc.; M.L. Bertolaccini, PhD, Academic Department of Vascular Surgery, King's College London; J.L. Rodriguez-Garcia, PhD, Department of Internal Medicine, University of Castilla-La Mancha; Z. Shums, MSc, Inova Diagnostics Inc.; O. Ateka-Barrutia, MD, Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital; M. Sorice, MD, Department of Experimental Medicine, La Sapienza University; G.L. Norman, PhD (AGAF, FAASLD), Inova Diagnostics Inc.; M. Khamashta, MD, PhD (FRCP), Graham Hughes Lupus Research Laboratory, Lupus Research Unit, The Rayne Institute, Division of Women's Health, King's College London, St. Thomas' Hospital
| |
Collapse
|
19
|
Giacomelli R, Afeltra A, Alunno A, Baldini C, Bartoloni-Bocci E, Berardicurti O, Carubbi F, Cauli A, Cervera R, Ciccia F, Cipriani P, Conti F, De Vita S, Di Benedetto P, Doria A, Drosos AA, Favalli EG, Gandolfo S, Gatto M, Grembiale RD, Liakouli V, Lories R, Lubrano E, Lunardi C, Margiotta DPE, Massaro L, Meroni P, Minniti A, Navarini L, Pendolino M, Perosa F, Pers JO, Prete M, Priori R, Puppo F, Quartuccio L, Ruffatti A, Ruscitti P, Russo B, Sarzi-Puttini P, Shoenfeld Y, Somarakis GA, Spinelli FR, Tinazzi E, Triolo G, Ursini F, Valentini G, Valesini G, Vettori S, Vitali C, Tzioufas AG. International consensus: What else can we do to improve diagnosis and therapeutic strategies in patients affected by autoimmune rheumatic diseases (rheumatoid arthritis, spondyloarthritides, systemic sclerosis, systemic lupus erythematosus, antiphospholipid syndrome and Sjogren's syndrome)? Autoimmun Rev 2017; 16:911-924. [DOI: 10.1016/j.autrev.2017.07.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/20/2017] [Indexed: 02/06/2023]
|
20
|
Controversias del síndrome de anticuerpos antifosfolipídicos en obstetricia. ACTA ACUST UNITED AC 2017; 13:30-36. [DOI: 10.1016/j.reuma.2016.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/25/2016] [Accepted: 04/28/2016] [Indexed: 11/17/2022]
|
21
|
Known unknowns of cardiolipin signaling: The best is yet to come. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:8-24. [PMID: 27498292 PMCID: PMC5323096 DOI: 10.1016/j.bbalip.2016.08.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/27/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
Since its discovery 75years ago, a wealth of knowledge has accumulated on the role of cardiolipin, the hallmark phospholipid of mitochondria, in bioenergetics and particularly on the structural organization of the inner mitochondrial membrane. A surge of interest in this anionic doubly-charged tetra-acylated lipid found in both prokaryotes and mitochondria has emerged based on its newly discovered signaling functions. Cardiolipin displays organ, tissue, cellular and transmembrane distribution asymmetries. A collapse of the membrane asymmetry represents a pro-mitophageal mechanism whereby externalized cardiolipin acts as an "eat-me" signal. Oxidation of cardiolipin's polyunsaturated acyl chains - catalyzed by cardiolipin complexes with cytochrome c. - is a pro-apoptotic signal. The messaging functions of myriads of cardiolipin species and their oxidation products are now being recognized as important intracellular and extracellular signals for innate and adaptive immune systems. This newly developing field of research exploring cardiolipin signaling is the main subject of this review. This article is part of a Special Issue entitled: Lipids of Mitochondria edited by Guenther Daum.
Collapse
|
22
|
Thrombotic risk assessment in antiphospholipid syndrome: the role of new antibody specificities and thrombin generation assay. Clin Mol Allergy 2016; 14:6. [PMID: 27429595 PMCID: PMC4947367 DOI: 10.1186/s12948-016-0043-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/07/2016] [Indexed: 11/28/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune condition characterized by the presence of antiphospholipid antibodies (aPL) in subjects presenting with thrombosis and/or pregnancy loss. The currently used classification criteria were updated in the international consensus held in Sidney in 2005. Vascular events seem to result of local procoagulative alterations upon triggers influence (the so called “second-hit theory”), while placental thrombosis and complement activation seem to lead to pregnancy morbidity. The laboratory tests suggested by the current classification criteria include lupus anticoagulant, a functional coagulation assay, and anticardiolipin and anti-β2-glycoprotein-I antibodies, generally detected by solid phase enzyme-linked immunosorbent assay. The real challenge for treating physicians is understanding what is the actual weight of aPL in provoking clinical manifestations in each case. As thrombosis has a multi-factorial cause, each patient needs a risk-stratified approach. In this review we discuss the role of thrombotic risk assessment in primary and secondary prevention of venous and arterial thromboembolic disease in patients with APS, focusing on new antibody specificities, available risk scoring models and new coagulation assays.
Collapse
|
23
|
Conti F, Ceccarelli F, Perricone C, Leccese I, Massaro L, Pacucci VA, Truglia S, Miranda F, Spinelli FR, Alessandri C, Valesini G. The chronic damage in systemic lupus erythematosus is driven by flares, glucocorticoids and antiphospholipid antibodies: results from a monocentric cohort. Lupus 2016; 25:719-26. [PMID: 26821965 DOI: 10.1177/0961203315627199] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 12/21/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Literature data suggest a significantly higher mortality in patients affected by systemic lupus erythematosus (SLE) developing chronic damage. Therefore, damage prevention is a major goal in the management of SLE patients. In the present study, we assessed damage by means of the Systemic Lupus International Collaborative Clinics/American College of Rheumatology (SLICC/ACR) damage index (SDI), in a large cohort of SLE patients. Additionally, we aimed at evaluating its association with demographic and clinical features as well as with disease activity and laboratory findings. PATIENTS AND METHODS We enrolled consecutive patients affected by SLE diagnosed according to the American College of Rheumatology (ACR) 1997 revised criteria. Chronic damage was determined by SDI calculated at the last examination in all patients with at least six months of follow-up. Disease activity was assessed by the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K); flare was defined as an increase of SLEDAI-2K ≥ 4 compared with the previous visit. RESULTS We evaluated 349 SLE patients (M/F 25/324, mean age ± SD 42.7 ± 12.4 years, mean disease duration ± SD 164.9 ± 105.2 months). Among the enrolled patients, 125 (35.8%) showed a SDI ≥ 1 (mean SDI ± SD 1.7 ± 0.9, range 0-5). The musculo-skeletal was the most frequently involved organ/system in SDI score (41/349 patients, 11.7%), with deforming/erosive arthritis in 21/349 (6.0%). The presence of chronic damage was associated with age (P < 0.001), disease duration (P < 0.001), number of flares (P = 0.02) and with the use of glucocorticoids (P = 0.02). The logistic regression analysis revealed the association between neuropsychiatric damage and antiphospholipid syndrome (P = 0.01, OR = 3.9) and between the presence of cardiovascular damage and anti-β2GPI antibodies (P = 0.01, OR 6.2). CONCLUSIONS In the present study chronic damage was identified in about one third of SLE patients. The association between SDI and the number of flares claim for a thigh-control of the disease activity in order to prevent the chronic damage. The possible role of antiphospholipid antibodies (aPL) in the development of neuropsychiatric and cardiovascular damage may suggest a more careful assessment of such aPL positive patients.
Collapse
Affiliation(s)
- F Conti
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - F Ceccarelli
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - C Perricone
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - I Leccese
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - L Massaro
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - V A Pacucci
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - S Truglia
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - F Miranda
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - F R Spinelli
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - C Alessandri
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - G Valesini
- Lupus Clinic, Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
24
|
Iannaccone A, Giorgianni F, New DD, Hollingsworth TJ, Umfress A, Alhatem AH, Neeli I, Lenchik NI, Jennings BJ, Calzada JI, Satterfield S, Mathews D, Diaz RI, Harris T, Johnson KC, Charles S, Kritchevsky SB, Gerling IC, Beranova-Giorgianni S, Radic MZ. Circulating Autoantibodies in Age-Related Macular Degeneration Recognize Human Macular Tissue Antigens Implicated in Autophagy, Immunomodulation, and Protection from Oxidative Stress and Apoptosis. PLoS One 2015; 10:e0145323. [PMID: 26717306 PMCID: PMC4696815 DOI: 10.1371/journal.pone.0145323] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 12/01/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We investigated sera from elderly subjects with and without age-related macular degeneration (AMD) for presence of autoantibodies (AAbs) against human macular antigens and characterized their identity. METHODS Sera were collected from participants in the Age-Related Maculopathy Ancillary (ARMA) Study, a cross-sectional investigation ancillary to the Health ABC Study, enriched with participants from the general population. The resulting sample (mean age: 79.2±3.9 years old) included subjects with early to advanced AMD (n = 131) and controls (n = 231). Sera were tested by Western blots for immunoreactive bands against human donor macular tissue homogenates. Immunoreactive bands were identified and graded, and odds ratios (OR) calculated. Based on these findings, sera were immunoprecipitated, and subjected to 2D gel electrophoresis (GE). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify the targets recognized by circulating AAbs seen on 2D-GE, followed by ELISAs with recombinant proteins to confirm LC-MS/MS results, and quantify autoreactivities. RESULTS In AMD, 11 immunoreactive bands were significantly more frequent and 13 were significantly stronger than in controls. Nine of the more frequent bands also showed stronger reactivity. OR estimates ranged between 4.06 and 1.93, and all clearly excluded the null value. Following immunoprecipitation, 2D-GE and LC-MS/MS, five of the possible autoreactivity targets were conclusively identified: two members of the heat shock protein 70 (HSP70) family, HSPA8 and HSPA9; another member of the HSP family, HSPB4, also known as alpha-crystallin A chain (CRYAA); Annexin A5 (ANXA5); and Protein S100-A9, also known as calgranulin B that, when complexed with S100A8, forms calprotectin. ELISA testing with recombinant proteins confirmed, on average, significantly higher reactivities against all targets in AMD samples compared to controls. CONCLUSIONS Consistent with other evidence supporting the role of inflammation and the immune system in AMD pathogenesis, AAbs were identified in AMD sera, including early-stage disease. Identified targets may be mechanistically linked to AMD pathogenesis because the identified proteins are implicated in autophagy, immunomodulation, and protection from oxidative stress and apoptosis. In particular, a role in autophagy activation is shared by all five autoantigens, raising the possibility that the detected AAbs may play a role in AMD via autophagy compromise and downstream activation of the inflammasome. Thus, we propose that the detected AAbs provide further insight into AMD pathogenesis and have the potential to contribute to disease biogenesis and progression.
Collapse
Affiliation(s)
- Alessandro Iannaccone
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- * E-mail:
| | - Francesco Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - David D. New
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - T. J. Hollingsworth
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Allison Umfress
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Albert H. Alhatem
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Indira Neeli
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Nataliya I. Lenchik
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Barbara J. Jennings
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Jorge I. Calzada
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Suzanne Satterfield
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Dennis Mathews
- Eye Specialty Group, Memphis, TN, United States of America
- Southern College of Optometry, Memphis, TN, United States of America
| | - Rocio I. Diaz
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Tamara Harris
- National Institute on Aging, NIH, Bethesda, MD, United States of America
| | - Karen C. Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Steve Charles
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Stephen B. Kritchevsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Sticht Center on Aging, Wake Forest University, Winston-Salem, NC, United States of America
| | - Ivan C. Gerling
- Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Sarka Beranova-Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Marko Z. Radic
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | | |
Collapse
|
25
|
Fukui S, Hirota S, Iwamoto N, Karata H, Kawakami A. Takayasu Arteritis With Antiphosphatidylserine/Prothrombin Antibody-Positive Antiphospholipid Syndrome: Case Report and Literature Review. Medicine (Baltimore) 2015; 94:e2345. [PMID: 26705229 PMCID: PMC4697995 DOI: 10.1097/md.0000000000002345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A relationship between Takayasu arteritis (TA) and positive antiphospholipid antibody states has been pointed out, but patients with TA complicated with antiphospholipid antibody syndrome (APS) are rare. Here we report the case of a 17-year-old Japanese man diagnosed with TA based on pulselessness of the left brachial artery, discrepancy of blood pressure between the upper extremities, and arterial wall thickening and narrowing of artery in contrast computed tomography. He was also diagnosed with provisional APS based on a pulmonary infarction without narrowing of the pulmonary artery and positive antiphosphatidylserine/prothrombin antibody. The patient also had concurrent Crohn's disease (CD) based on histopathological findings, which may have been associated with TA. We started high-dose corticosteroid therapy and anticoagulation therapy, and his symptoms including fever, dizziness, chest pain, and lower-right uncomfortable abdomen improved.We reviewed 9 cases of TA with APS including our patient by conducting a PubMed search. Based on past reports, we considered the relationship among TA, APS, and CD.Clinicians should bear in mind that many etiologies can exist in 1 patient, and differential diagnoses are essential.
Collapse
Affiliation(s)
- Shoichi Fukui
- From the Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki (SF, SH, NI, AK); and Department of Pathology, Nagasaki University Hospital, Nagasaki, Japan (HK)
| | | | | | | | | |
Collapse
|
26
|
McDonnell T, Pericleous C, Laurine E, Tommasi R, Garza-Garcia A, Giles I, Ioannou Y, Rahman A. Development of a high yield expression and purification system for Domain I of Beta-2-glycoprotein I for the treatment of APS. BMC Biotechnol 2015; 15:104. [PMID: 26576675 PMCID: PMC4650279 DOI: 10.1186/s12896-015-0222-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 11/03/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this paper we describe a novel method to achieve high yield bacterial expression of a small protein domain with considerable therapeutic potential; Domain I of Beta-2-glycoprotein I (β2GPI). β2GPI is intrinsic to the pathological progression of the Antiphospholipid Syndrome (APS). Patients develop autoantibodies targeting an epitope located on the N-terminal Domain I of β2GPI rendering this domain of interest as a possible therapeutic. RESULTS This new method of production of Domain I of β2GPI has increased the production yield by ~20 fold compared to previous methods in E.coli. This largely scalable, partially automated method produces 50-75 mg of pure, folded, active Domain I of β2GPI per litre of expression media. CONCLUSION The application of this method may enable production of Domain I on sufficient scale to allow its use as a therapeutic.
Collapse
Affiliation(s)
- Thomas McDonnell
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Charis Pericleous
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Emmanuelle Laurine
- PolyTherics, Babraham Research Campus, Babraham, CB22 3AT, Cambridge, UK.
| | - Rita Tommasi
- PolyTherics, Babraham Research Campus, Babraham, CB22 3AT, Cambridge, UK.
| | - Acely Garza-Garcia
- Structural Biology, Medical Research Council National Institute for Medical Research, London, UK.
| | - Ian Giles
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Yiannis Ioannou
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK. .,Arthritis Research UK Centre for Adolescent Rheumatology, University College London, London, UK.
| | - Anisur Rahman
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
27
|
Slone EA, Pope MR, Fleming SD. Phospholipid scramblase 1 is required for β2-glycoprotein I binding in hypoxia and reoxygenation-induced endothelial inflammation. J Leukoc Biol 2015. [PMID: 26216936 DOI: 10.1189/jlb.3a1014-480r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multiple pathologic conditions, including hemorrhage, tumor angiogenesis, and ischemia-reperfusion events, will result in hypoxia and subsequent reperfusion. Previous studies have analyzed the lipid changes within whole tissues and indicated that ischemia-reperfusion altered tissue and cellular phospholipids. Using an in vitro cell culture model of hypoxia and reoxygenation, we examined the endothelial lipid changes. We hypothesized that phospholipid scramblase 1, a protein that regulates bilayer asymmetry, is involved in altering the phospholipids of endothelial cells during hypoxia, a component of ischemia, leading to β2-glycoprotein I and IgM binding and subsequent lipid-mediated, inflammatory responses. We have completed the first comprehensive study of steady-state phospholipid scramblase 1 mRNA levels, protein expression, and activity under conditions of hypoxia and reoxygenation. Phospholipid scramblase 1 regulates phosphatidylserine exposure in response to oxygen stress, leading to β2-glycoprotein I and IgM binding and lipid-mediated, inflammatory responses.
Collapse
Affiliation(s)
| | - Michael R Pope
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
28
|
Conti F, Ceccarelli F, Gigante A, Perricone C, Barbano B, Massaro L, Spinelli FR, Alessandri C, Valesini G, Cianci R. Ultrasonographic Evaluation of Resistive Index and Renal Artery Stenosis in Patients with Anti-Phospholipid Syndrome: Two Distinct Mechanisms? ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:1814-1820. [PMID: 25800790 DOI: 10.1016/j.ultrasmedbio.2015.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 06/04/2023]
Abstract
Renal involvement in anti-phospholipid syndrome (APS) is still relatively unknown and probably underestimated. The described lesions consist of renal artery stenosis (RAS), venous renal thrombosis and glomerular lesions. The resistive index (RI) of intra-renal arteries, expression of the degree of vascular resistance, has been analyzed in different nephropathies and observed to be associated with functional parameters and some histologic features. In contrast, there are no studies on patients with APS. We evaluated the presence of a pathologic RI and RAS in a cohort of patients with APS. The study protocol included ultrasonographic assessment to measure the RI (RIs >0.7 were considered pathologic) and to determine the presence of RAS. We enrolled 36 patients with APS, 13 with primary APS and 23 with the form associated with systemic lupus erythematosus (SLE, secondary APS). As controls, we enrolled 10 anti-phospholipid antibody carriers, 10 patients with SLE without renal involvement and 14 age- and sex-matched healthy patients. A pathologic RI was identified in five patients with APS (13.9%) and in none of the anti-phospholipid antibody carriers (p = 0.00007). Four of the five (80%) patients with a pathologic RI had secondary APS. Three patients, all with primary APS, had RAS. The almost exclusive association of a pathologic RI with secondary APS and of RAS with primary APS suggests the involvement of two pathogenic pathways in the development of these different manifestations. The hypercoagulability status driven by APS could play a central role in the determination of RAS in patients with primary APS, whereas the activation of mTORC (mammalian target of rapamycin complex) pathways could be the pathogenic mechanism inducing development of a pathologic RI.
Collapse
Affiliation(s)
- Fabrizio Conti
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy.
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Antonietta Gigante
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Carlo Perricone
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Biagio Barbano
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Laura Massaro
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Francesca Romana Spinelli
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Lupus Clinic, Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Rosario Cianci
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
29
|
Study of the fibrinolytic process in a patient with antiphospholipid syndrome. Blood Coagul Fibrinolysis 2015; 26:239-45. [DOI: 10.1097/mbc.0000000000000062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Abstract
Systemic lupus erythematosus is a remarkable and challenging disorder. Its diversity of clinical features is matched by the complexity of the factors (genetic, hormonal, and environmental) that cause it, and the array of autoantibodies with which it is associated. In this Seminar we reflect on changes in its classification criteria; consider aspects of its more serious clinical expression; and provide a brief review of its aetiopathogenesis, major complications, coping strategies, and conventional treatment. Increased understanding of the cells and molecules involved in the development of the diseases has encouraged the identification of new, better targeted biological approaches to its treatment. The precise role of these newer therapies remains to be established.
Collapse
Affiliation(s)
| | - Grainne Murphy
- Centre for Rheumatology, Department of Medicine, University College London Hospital, London, UK
| | - David Isenberg
- Centre for Rheumatology, Department of Medicine, University College London Hospital, London, UK.
| |
Collapse
|
31
|
Plavsic A, Miskovic R, Raskovic S, Bogic M, Bonaci Nikolic B. Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Antiphospholipid syndrome is an autoimmune disorder defined as association of vascular thrombosis and/or pregnancy complications with presence of antiphospholipid antibodies (lupus anticoagulant, anticardiolipin and anti-β2 glycoprotein I). It is the most common cause of acquired thrombophilia, and can occur as an independent entity or in relation with other diseases, especially systemic lupus erythematosus. Presence of antiphospholipid syndrome in systemic lupus erythematosus is additional vaso occlusive factor in already present inflammation, bringing further risk for thrombotic events. Clinical and serological manifestations of antiphospholipid syndrome and systemic lupus erythematosus are very similar, so possible connection for these two autoimmune disorders is assumed.
Collapse
|
32
|
Risk of venous and arterial thrombosis according to type of antiphospholipid antibodies in adults without systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2014; 13:595-608. [DOI: 10.1016/j.autrev.2013.11.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 11/08/2013] [Indexed: 11/20/2022]
|
33
|
Obstetric antiphospholipid syndrome: a recent classification for an old defined disorder. Autoimmun Rev 2014; 13:901-8. [PMID: 24820522 DOI: 10.1016/j.autrev.2014.05.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 04/18/2014] [Indexed: 12/11/2022]
Abstract
Obstetric antiphospholipid syndrome (APS) is now being recognized as a distinct entity from vascular APS. Pregnancy morbidity includes >3 consecutive and spontaneous early miscarriages before 10weeks of gestation; at least one unexplained fetal death after the 10th week of gestation of a morphologically normal fetus; a premature birth before the 34th week of gestation of a normal neonate due to eclampsia or severe pre-eclampsia or placental insufficiency. It is not well understood how antiphospholipid antibodies (aPLs), beyond their diagnostic and prognostic role, contribute to pregnancy manifestations. Indeed aPL-mediated thrombotic events cannot explain the obstetric manifestations and additional pathogenic mechanisms, such as a placental aPL mediated complement activation and a direct effect of aPLs on placental development, have been reported. Still debated is the possible association between aPLs and infertility and the effect of maternal autoantibodies on non-vascular manifestations in the babies. Combination of low dose aspirin and unfractionated or low molecular weight heparin is the effective treatment in most of the cases. However, pregnancy complications, in spite of this therapy, can occur in up to 20% of the patients. Novel alternative therapies able to abrogate the aPL pathogenic action either by interfering with aPL binding at the placental level or by inhibiting the aPL-mediated detrimental effect are under active investigation.
Collapse
|
34
|
Matta BN, Uthman I, Taher AT, Khamashta MA. The current standing of diagnosis of antiphospholipid syndrome associated with systemic lupus erythematosus. Expert Rev Clin Immunol 2014; 9:659-68. [PMID: 23899236 DOI: 10.1586/1744666x.2013.811183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The antiphospholipid syndrome was first described in the early 1980s. The term was first coined to describe patients presenting with recurrent arterial and venous thrombosis or pregnancy complications. Antiphospholipid syndrome was first reported in systemic lupus erythematosus patients, but later on it became obvious that systemic lupus erythematosus is not a necessary condition for its occurrence. It has been shown that antibodies to phospholipids are the main causative agents of the disease, hence its name. The diagnosis of the disease has witnessed a remarkable evolution over the course of the past 25 years. With the observation that clinical parameters would not be enough to accurately diagnose the disease, antiphospholipid antibodies were recognized to play a central role in this regard. The main hindrance to an accurate diagnosis was the lack of standardization between different laboratory parameters that tested for the antiphospholipid antibodies. Lately, a combination of tests has been acknowledged to play a crucial role in diagnosis.
Collapse
Affiliation(s)
- Bassem N Matta
- Department of Internal Medicine, Division of Hematology & Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | | | | | | |
Collapse
|
35
|
|
36
|
Diagnose eines primären Antiphospholipidsyndroms bei Ulcus cruris unter Marcumar-Therapie. Hautarzt 2013; 64:666-70. [DOI: 10.1007/s00105-013-2601-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
37
|
Nayfe R, Uthman I, Aoun J, Saad Aldin E, Merashli M, Khamashta MA. Seronegative antiphospholipid syndrome. Rheumatology (Oxford) 2013; 52:1358-67. [PMID: 23502076 DOI: 10.1093/rheumatology/ket126] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
APS is an autoimmune disease that leads to arterial and/or venous thrombosis, recurrent pregnancy loss and persistently positive aPLs. Patients with clinical manifestations highly suggestive of APS but persistently negative conventional aPLs are classified as having seronegative APS. Ongoing research has revealed the existence of non-criteria antibodies proposed to be relevant to APS and that can be potentially included in the disease's classification criteria. We present a literature review on the most promising antibodies of this heterogeneous aPL family, which includes antibodies to a zwitterionic phospholipid, namely phosphatidylethanolamine, phospholipid-binding plasma proteins, phospholipid-protein complexes and anionic phospholipids other than cardiolipin. Although these molecules can increase the diagnostic yield of APS, their clinical relevance is still debatable and needs to be confirmed by interlaboratory efforts toward standardizing diagnostic tools, in addition to experimental data and larger longitudinal studies.
Collapse
Affiliation(s)
- Rabih Nayfe
- Department of Internal Medicine, American University of Beirut Medical Center, PO Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon
| | | | | | | | | | | |
Collapse
|
38
|
Avrameas S, Selmi C. Natural autoantibodies in the physiology and pathophysiology of the immune system. J Autoimmun 2013; 41:46-9. [PMID: 23384670 DOI: 10.1016/j.jaut.2013.01.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 01/23/2023]
Abstract
Natural autoantibodies (autoNAb) recognize self antigens and are an important component of the immune system, as species ranging from invertebrates to vertebrates have polyreactive IgM NAbs. In higher vertebrates, different polyreactive autoNAbs isotypes are also frequently encountered and autopolyreactive IgG NAbs are largely predominant compared to low-titer monoreactive IgG NAbs specific for either self or non-self antigens. Autopolyreactive NAbs manifest the capacity to recognize three-dimensional structures and thus represent a fundamental feature of the immune system that has long been preserved during evolution. NAbs are produced in a continuum of functional and phenotypic tiers of B cells and are likely to derive from proteins initially selected to build the organism that were adapted through evolution to recognize environmental constituents, while preserving their capacity to recognize self antigens. The clonal selection is considered the predominant mechanism of the regulation of the immune system complexity but growing evidence suggests that autoNAbs are also actively implicated. In all species NAbs reacting with either self or non-self antigens constitute a vast network of infinite interactions providing high complexity, stability and plasticity. This evolutionary process was intended to allow the effective recognition of environmental antigens, immune memory, immunoregulatory phenomena, as well as tissue homeostasis. The present article is intended to illustrate the history and the current and future developments in our understanding of self and non-self recognizing NAbs to ultimately enlighten the complexity of the immune system regulation.
Collapse
Affiliation(s)
- Stratis Avrameas
- School of Medicine, University of Athens, Mikras Asias 75, 115 27 Athens, Greece.
| | | |
Collapse
|
39
|
Abstract
The mechanisms leading to the onset and perpetuation of systemic and tissue-specific autoimmune diseases are complex, and numerous hypotheses have been proposed or confirmed over the past 12 months. It is particularly of note that the number of articles published during 2011 in the major immunology and autoimmunity journals increased by 3 % compared to the previous year. The present article is dedicated to a brief review of the reported data and, albeit not comprehensive of all articles, is aimed at identifying common and future themes. First, clinical researchers were particularly dedicated to defining refractory forms of diseases and to discuss the use and switch of therapeutic monoclonal antibodies in everyday practice. Second, following the plethora of genome-wide association studies reported in most multifactorial diseases, it became clear that genomics cannot fully explain the individual susceptibility and additional environmental or epigenetic factors are necessary. Both these components were widely investigated, both in organ-specific (i.e., type 1 diabetes) and systemic (i.e., systemic lupus erythematosus) diseases. Third, a large number of 2011 works published in the autoimmunity area are dedicated to dissect pathogenetic mechanisms of tolerance breakdown in general or in specific conditions. While our understanding of T regulatory and Th17 cells has significantly increased in 2011, it is of note that most of the proposed lines of evidence identify potential targets for future treatments and should not be overlooked.
Collapse
|
40
|
Anti-phosphatidylethanolamine antibody, thromboembolic events and the antiphospholipid syndrome. Autoimmun Rev 2012; 12:230-4. [PMID: 22796282 DOI: 10.1016/j.autrev.2012.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 07/04/2012] [Indexed: 11/21/2022]
|
41
|
Venous thromboembolic disease in systemic autoimmune diseases: An association to keep in mind. Autoimmun Rev 2012; 12:289-94. [DOI: 10.1016/j.autrev.2012.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/01/2012] [Indexed: 11/18/2022]
|
42
|
Mahler M, Norman GL, Meroni PL, Khamashta M. Autoantibodies to domain 1 of beta 2 glycoprotein 1: A promising candidate biomarker for risk management in antiphospholipid syndrome. Autoimmun Rev 2012; 12:313-7. [DOI: 10.1016/j.autrev.2012.05.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 11/29/2022]
|
43
|
Paroli M, Polidoro A, Romano S, Accapezzato D. Churg-Strauss syndrome associated with antiphospholipid antibodies in a patient with recurrent myocardial and cerebral ischemia. Int Med Case Rep J 2012; 5:79-81. [PMID: 23754929 PMCID: PMC3658260 DOI: 10.2147/imcrj.s37516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
We report on a case of Churg-Strauss syndrome (CSS) associated with the presence of antiphospholipid antibodies. The patient had a history of recurrent myocardial infarction and presented with acute ischemic cerebral disease. Eosinophilia with typical lung and skin lesions led us to diagnose the patient with CCS. We hypothesize that the presence of antiphospholipid antibodies significantly contributed to the ischemic events. We suggest that the search for antiphospholipid antibodies should be included in the laboratory work-up in CSS patients and patients affected by primary systemic vasculitides in general. Moreover, anticoagulant treatment appears to be warranted in all CSS patients and antiphospholipid antibodies to counteract this thrombosis-favoring association.
Collapse
Affiliation(s)
- Marino Paroli
- Department of Biotechnology and Medical-Surgical Sciences
| | | | | | | |
Collapse
|
44
|
Antiphospholipid antibodies as non-traditional risk factors in atherosclerosis based cardiovascular diseases without overt autoimmunity. A critical updated review. Autoimmun Rev 2012; 11:873-82. [DOI: 10.1016/j.autrev.2012.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Accepted: 03/06/2012] [Indexed: 11/23/2022]
|
45
|
Hasegawa M, Fujimoto M, Orito H, Matsushita T, Hamaguchi Y, Takehara K. Two cases of livedo vasculopathy with non-criteria antiphospholipid antibodies. J Dermatol 2012; 39:1026-30. [PMID: 22974060 DOI: 10.1111/j.1346-8138.2012.01664.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/25/2012] [Indexed: 11/30/2022]
Abstract
Livedo vasculopathy is characterized by reticular distribution of purpuric macules and papules of the lower legs, caused by intraluminal thrombosis of small vessels. Antiphospholipid antibodies are detected in a subset of these patients. We treated two cases (a 34-year-old man and a 46-year-old woman) with livedo vasculopathy. In both cases, thrombosis was seen only in the skin. The presence of immunoglobulin (Ig)G or IgM anticardiolipin antibody (Ab), IgG or IgM anti-β(2) -glycoprotein I Ab, or lupus anticoagulant are necessary for criteria-based diagnosis of antiphospholipid syndrome. However, our patients were negative for these Ab, and instead had either IgG antiphosphatidylethanolamine Ab or IgA anticardiolipin Ab. These Ab are suggestive of antiphospholipid syndrome but are not considered "criteria" Ab. This report demonstrates the existence of antiphosphatidylethanolamine Ab or IgA anticardiolipin Ab in patients with livedo vasculopathy. However, the frequency and significance of these Ab in livedo vasculopathy should be confirmed in larger longitudinal studies.
Collapse
Affiliation(s)
- Minoru Hasegawa
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Cattelan J, Racadot E, Di Martino V, Thevenot T. Antiannexin V antibodies: an underestimated source of noncirrhotic portal vein thrombosis? Hepatology 2012; 56:1182. [PMID: 22371219 DOI: 10.1002/hep.25669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
47
|
Antiphospholipid antibody profiling — Time for a new technical approach? Autoimmun Rev 2012; 11:821-6. [DOI: 10.1016/j.autrev.2012.02.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 02/19/2012] [Indexed: 02/01/2023]
|
48
|
Novel insights into pathogenesis, diagnosis and treatment of antiphospholipid syndrome. Curr Opin Rheumatol 2012; 24:473-81. [DOI: 10.1097/bor.0b013e328354ae8c] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
49
|
Ye Y, Hu Z, Liu J, Chen G, Zhou Y, Yu L. Detection of anticardiolipin antibody IgG by time-resolved fluoroimmunoassay. Clin Rheumatol 2012; 31:1339-45. [PMID: 22684164 DOI: 10.1007/s10067-012-2020-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/22/2012] [Accepted: 05/30/2012] [Indexed: 11/30/2022]
Abstract
In an effort to improve the quantitative detection of anticardiolipin antibodies (aCL) IgG so as to classify patients correctly as antiphospholipid syndrome (APS) positive, we developed a new immunoassay based on a sandwich time-resolved fluoroimmunoassay (TRFIA) using the complex of cardiolipin plus bovine β(2)GPI as antigen and Eu(3+)-labeled rabbit antihuman IgG as conjugate. The precision, sensitivity, specificity, and stability of the assay were evaluated, and comparison with the classical ELISA was also made. The aCL IgG TRFIA kit we established had a wider detectable range than three commercial ELISA ones from different manufacturers when a specimen was diluted, with strong positive result from 1:12.5 to 1:204,800. The average intra-assay and inter-assay CVs detected by the aCL IgG TRFIA was 3.14 and 3.70 %, respectively. The sensitivity was 0.1 GPL U/ml, and the clinical diagnostic specificity was 98 %. The established assay kit also behaved better in stability than the commercial ELISA ones. Additionally, the immunoassay we established correlated well with the ELISA, and the correlation coefficient was 0.975. We thus conclude that the TRFIA we developed for aCL IgG detection gives promise to a more sensitive and reliable diagnosis of APS and has potential value for large-scale screening programs.
Collapse
Affiliation(s)
- Yan Ye
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | | | | | | | | | | |
Collapse
|
50
|
Chang C. Neonatal autoimmune diseases: a critical review. J Autoimmun 2012; 38:J223-38. [PMID: 22402339 DOI: 10.1016/j.jaut.2011.11.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 12/21/2022]
Abstract
Neonatal autoimmune diseases are distinctly rare. Most neonatal autoimmune diseases result from the transplacental transfer of maternal antibodies directed against fetal or neonatal antigens in various tissues. In neonatal lupus, the heart seems to be particularly susceptible. Primary autoimmunity in newborns, with the exception of familial autoinflammatory diseases, is virtually non-existent. The pathophysiologic basis for the development of neonatal autoimmunity is not entirely clear, but differences in the neonatal immune system compared with the adult immune system, as well as unique characteristics of target antigens in the newborn period may be important factors. Neonatal lupus is the most common presentation of autoimmunity in the newborn. But the characteristics defining neonatal lupus are not well defined and the presentation of neonatal lupus differs from that of classical lupus. Other neonatal autoimmune diseases involving the interaction between maternal antibodies and fetal/neonatal antigens include neonatal anti-phospholipid syndrome, Behcet's disease, neonatal autoimmune thyroid disease, neonatal polymyositis and dermatomyositis, neonatal scleroderma and neonatal type I diabetes mellitus. While autoantibodies have been detected in patients with neonatal autoimmune disease, the pathogenic role of autoantibodies has not been well defined. Other mechanisms may play a role in the development of neonatal autoimmunity, including fetal/maternal microchimerism and aberrant apoptosis of fetal cells. The autoinflammatory syndromes are a completely different category, but are also included in discussion of neonatal autoimmune diseases. The autoinflammatory syndromes include the cryopyrin associated periodic syndromes (CAPS) - familial cold autoinflammatory syndrome (FCAS), neonatal onset multisystem inflammatory disease (NOMID) and Muckle-Wells syndrome, which all share a common pathophysiologic mechanism.
Collapse
Affiliation(s)
- Christopher Chang
- Division of Allergy, Asthma and Immunology, Thomas Jefferson University, Nemours/AI duPont Hospital for Children, Wilmington, DE 19803, USA.
| |
Collapse
|