1
|
Tang KT, Chen YS, Chen TT, Chao YH, Kung SP, Chen DY, Lin CC. Inhibiting Tyrosine Kinase 2 Ameliorates Antiphospholipid Syndrome Nephropathy. Mediators Inflamm 2024; 2024:5568822. [PMID: 39742289 PMCID: PMC11688129 DOI: 10.1155/mi/5568822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/07/2024] [Indexed: 01/03/2025] Open
Abstract
Objective: Antiphospholipid antibody syndrome (APS) is an autoimmune disease characterized by the presence of β2-glycoprotein I (β2-GPI)-targeting antiphospholipid antibodies (aPLs) and vascular thrombosis or obstetrical complications. One of its severe manifestations is nephropathy. Methods: To examine the role of type I interferon (IFN) and therapeutic potential of tyrosine kinase 2 (Tyk2) inhibition, we administered BMS-986202, a novel Tyk2 inhibitor, in a mouse model of APS nephropathy. We administered BMS-986202 to BALB/c mice at a dose of 2 mg/kg. Biochemical and histological characteristics of APS nephropathy were then determined. The type I IFN signature in the kidney was also evaluated by real-time polymerase chain reaction (PCR). Results: The Tyk2 inhibitor reversed the elevation of blood urea nitrogen (BUN) and microalbuminuria in the murine model of APS nephropathy. In addition, the Tyk2 inhibitor reversed the pathological vascular changes in the kidney as judged in electron microscopy (EM), and fibrin and C3 deposition as revealed in immunohistochemistry (IHC). An increased expression levels of IFN signature (IFN regulatory factor 7 (IRF7) and Mx1) in the kidneys of APS mice were found. Tyk2 inhibition reversed such an upregulation. Conclusion: Our results demonstrated the key role of type I IFN in the pathogenesis of APS nephropathy. Furthermore, the therapeutic efficacy of Tyk2 inhibition was demonstrated in a murine model of APS nephropathy. Our results could provide a new treatment strategy for this debilitating disease.
Collapse
Affiliation(s)
- Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Sin Chen
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| | - Tzu-Ting Chen
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hsuan Chao
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Shu-Ping Kung
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chi-Chien Lin
- Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Salter BM, Crowther MA. Catastrophic antiphospholipid syndrome: a CAPS-tivating hematologic disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:214-221. [PMID: 39644034 DOI: 10.1182/hematology.2024000544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Catastrophic antiphospholipid syndrome (CAPS) is a rare but life-threatening form of antiphospholipid syndrome (APS) defined by the rapid onset of large and small vessel thrombosis occurring simultaneously across multiple sites, resulting in multiorgan dysfunction. The presence of underlying immune dysfunction causing activation of coagulation and, in many cases, abnormal complement regulation predisposes these patients to thrombotic events. CAPS is often preceded by triggering factors such as infection, surgery, trauma, anticoagulation discontinuation, and malignancy. Given the high mortality rate, which may exceed 50%, prompt recognition and initiation of management is required. The detection of antiphospholipid antibodies and the histopathologic identification of microvascular ischemia via tissue biopsy are required to diagnose CAPS. However, these patients are often too unwell to obtain results and wait for them. As such, investigations should not delay CAPS therapy, especially if there is strong clinical suspicion. Management of CAPS requires "triple therapy" with glucocorticoids, intravenous heparin, therapeutic plasma exchange, and/or intravenous immunoglobulin. Treatment for refractory disease is based on poor-quality evidence but includes anti-CD20 (rituximab) or anticomplement (eculizumab) monoclonal antibodies and other immunosuppressant agents, either alone or in combination. The rarity of this syndrome and the subsequent lack of randomized clinical trials have led to a paucity of high-quality evidence to guide management. Continued international collaboration to expand ongoing CAPS registries will allow a better understanding of the response to newer targeted therapy.
Collapse
Affiliation(s)
- Brittany M Salter
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
3
|
Zouein J, Naim N, Spencer DM, Ortel TL. Genetic and genomic associations in antiphospholipid syndrome: A systematic review. Autoimmun Rev 2024; 24:103712. [PMID: 39617250 DOI: 10.1016/j.autrev.2024.103712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Numerous genes have been associated with APS in the literature. In recent years, microRNA (miRNA) and long non-coding RNA (lncRNA) have also been shown to modulate the expression of APS-related genes. OBJECTIVE We performed a systematic review to identify all studies reporting on genetic mechanisms that have been shown to be associated with APS. METHODS An extensive literature search was performed in the PubMed, Cochrane and Web of Science databases gathering all available articles through February 2024. We only selected case-control studies that met inclusion criteria and that focused on genetic contributors and modifiers related to primary APS. RESULTS Sixty studies were selected for data extraction. Selected studies were grouped into 8 broad categories for review and analysis: (1) gene expression studies; (2) thrombophilia genotypes; (3) single nucleotide polymorphisms (SNPs); (4) interferon-inducible genes; (5) microRNA studies; (6) long non-coding RNA (lncRNA) studies; (7) DNA methylation studies; and (8) differential gene expression studies. Several genes have been identified as associated with APS by more than one approach, including TF, complement associated genes, and interferon-inducible genes. It has been demonstrated that miRNA and lncRNA may alter the expression of important genes in patients with APS. CONCLUSION This systematic review has helped highlight important genes implicated in APS. Most importantly, pathways such as thrombosis/hemostasis, complement and interferon appear to be involved. Further studies are needed to help uncover important genes that could serve as biomarkers.
Collapse
Affiliation(s)
- Joseph Zouein
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Nabih Naim
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University of Beirut, Lebanon
| | - Diane M Spencer
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Thomas L Ortel
- Hemostasis and Thrombosis Center, Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
4
|
Dieudonné Y, Lorenzetti R, Rottura J, Janowska I, Frenger Q, Jacquel L, Vollmer O, Carbone F, Chengsong Z, Luka M, Depauw S, Wadier N, Giorgiutti S, Nespola B, Herb A, Voll RE, Guffroy A, Poindron V, Ménager M, Martin T, Soulas-Sprauel P, Rizzi M, Korganow AS, Gies V. Defective germinal center selection results in persistence of self-reactive B cells from the primary to the secondary repertoire in Primary Antiphospholipid Syndrome. Nat Commun 2024; 15:9921. [PMID: 39548093 PMCID: PMC11568317 DOI: 10.1038/s41467-024-54228-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Primary antiphospholipid syndrome (PAPS) is a life-threatening clotting disorder mediated by pathogenic autoantibodies. Here we dissect the origin of self-reactive B cells in human PAPS using peripheral blood and bone marrow of patients with triple-positive PAPS via combined single-cell RNA sequencing, B cell receptors (BCR) repertoire profiling, CITEseq analysis and single cell immortalization. We find that antiphospholipid (aPL)-specific B cells are present in the naive compartment, polyreactive, and derived from the natural repertoire. Furthermore, B cells with aPL specificities are not eliminated in patients with PAPS, persist until the memory and long-lived plasma cell stages, likely after defective germinal center selection, while becoming less polyreactive. Lastly, compared with the non-PAPS cells, PAPS B cells exhibit distinct IFN and APRIL signature as well as dysregulated mTORC1 and MYC pathways. Our findings may thus elucidate the survival mechanisms of these autoreactive B cells and suggest potential therapeutic targets for the treatment of PAPS.
Collapse
Affiliation(s)
- Yannick Dieudonné
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France.
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France.
| | - Raquel Lorenzetti
- Center for Chronic Immunodeficiency, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Rheumatology and Clinical Immunology, Medical University of Graz, Graz, Austria
| | - Julien Rottura
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Life Sciences, Strasbourg, France
| | - Iga Janowska
- Center for Chronic Immunodeficiency, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Quentin Frenger
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Life Sciences, Strasbourg, France
| | - Léa Jacquel
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Olivier Vollmer
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Francesco Carbone
- Université Paris Cité, Institut Imagine, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
| | - Zhu Chengsong
- Department of Immunology, Microarray and Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marine Luka
- Université Paris Cité, Institut Imagine, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
| | - Sabine Depauw
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nadège Wadier
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Stéphane Giorgiutti
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Benoît Nespola
- Laboratoire d'Immunologie, Plateau technique de Biologie, Strasbourg University Hospital, Strasbourg, France
| | - Agathe Herb
- Hematology laboratory, Strasbourg University Hospital, Strasbourg, France
| | - Reinhard Edmund Voll
- Center for Chronic Immunodeficiency, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aurélien Guffroy
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Vincent Poindron
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
| | - Mickaël Ménager
- Université Paris Cité, Institut Imagine, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
| | - Thierry Martin
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Pauline Soulas-Sprauel
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Pharmacy, Illkirch, France
| | - Marta Rizzi
- Center for Chronic Immunodeficiency, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- CIBSS - Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Anne-Sophie Korganow
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Vincent Gies
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France.
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Université de Strasbourg, Faculty of Pharmacy, Illkirch, France.
| |
Collapse
|
5
|
Taghavi M, Jacobs L, Demulder A, Jabrane A, Mesquita MDCF, Defawe C, Laureys M, Dernier Y, Pozdzik A, Collart F, Nortier J. Antiphospholipid antibody positivity is associated with maturation failure and thrombosis of native arteriovenous fistula: a retrospective study in HD patients. Clin Kidney J 2024; 17:sfae308. [PMID: 39512379 PMCID: PMC11540859 DOI: 10.1093/ckj/sfae308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Indexed: 11/15/2024] Open
Abstract
Background and hypothesis The prevalence of antiphospholipid antibody (aPL) is high among hemodialysis (HD) patients compared to the general population and is inconsistently associated with arteriovenous fistula (AVF) thrombosis or stenosis. The association with maturation failure has never been investigated. This study aims to evaluate native AVF complications (thrombosis, stenosis, and maturation failure) and primary patency in aPL positive HD patients. Methods We retrospectively identified 116 HD patients with native AVF. We collected the aPL profiles, the clinical and biological data potentially involved in AVF maturation failure, thrombosis, and stenosis, and investigated the association of these complications and aPL positivity. Kaplan-Meier survival analysis was performed. Results In our cohort, the prevalence of aPL persistent positivity was 32.7% and this was strongly associated with AVF maturation failure defined by ultrasound. aPL persistent positivity was a strong predictor in multivariate analysis and this association was independent of AVF stenosis or thrombosis during maturation process. There was no association with primary and functional primary patency, and stenosis. However, aPL persistent positivity according to ACR/EULAR classification criteria was associated with thrombosis when compared to strictly negative aPL patients. Conclusions In our cohort, aPL persistent positivity was significantly associated with AVF maturation failure and thrombosis but not with AVF stenosis. To our knowledge, we report for the first time, a statistically significant association between aPL positivity and delay or absence of native AVF maturation.
Collapse
Affiliation(s)
- Maxime Taghavi
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Lucas Jacobs
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Anne Demulder
- Laboratory of Hematology and Hemostasis, Brugmann University Hospital, ULB, Brussels, Belgium
| | - Abla Jabrane
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Catherine Defawe
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marc Laureys
- Radiology Department, Brugmann University Hospital, ULB, Brussels, Belgium
| | - Yves Dernier
- Vascular Surgery Department, Brugmann University Hospital, ULB, Brussels, Belgium
| | - Agnieszka Pozdzik
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frédéric Collart
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Joëlle Nortier
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
6
|
Bucci T, Merashli M, Pignatelli P, Pastori D, Delgado-Alves J, Lip GYH, Ames PRJ. Intima media thickness of the carotid artery in primary antiphospholipid syndrome: A systematic review and meta-analysis. Autoimmun Rev 2024; 23:103657. [PMID: 39366515 DOI: 10.1016/j.autrev.2024.103657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Primary antiphospholipid syndrome (PAPS) has been associated with an increase in clinical events associated with atherosclerotic vascular disease. Intima media thickness (IMT) of carotid arteries is a surrogate marker of atherosclerotic vascular disease. OBJECTIVES To conduct a systematic review and meta-analysis of studies evaluating IMT and their clinical correlates in PAPS. METHODS Systematic search of EMBASE and PubMed databases from January 2000 to December 2023; we employed random effect meta-analyses for continuous outcomes and Peto's odds ratio for rare events. RESULTS The meta-analysis included 21 studies (20 case control and 1 cohort) showing that PAPS patients (n = 1103) had thicker IM than controls (n = 832) (p < 0.0001) with wide heterogeneity (I2 = 86.9 %); PAPS patients (n = 782) also had a greater pooled prevalence of carotid plaques than controls (n = 537) (13.1 % vs 2.97 %, p < 0.0001). A sensitivity analysis by meta-regression indicated that mean age, gender, disease duration, lipid profile, blood pressures, smoking and statin use all explained the heterogeneity variance; a sensitivity analysis by subgroups confirmed smoking status and statin use as explanatory variables with the addition of ethnicity. CONCLUSION Atherosclerosis of the carotid artery represents a clinical feature of PAPS in relation to the traditional risk factors and to statin use. Minimising the atherogenic risk with statins could reduce the late arterial atherothrombotic risks of PAPS.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Mira Merashli
- Department of Rheumatology, American University of Beirut, Beirut, Lebanon
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Daniele Pastori
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; IRCCS Neuromed, Localita' Camerelle, Pozzilli, Italy
| | - Jose' Delgado-Alves
- Immune Response & Vascular Disease, iNOVA,4Health, Nova Medical School, Nova University Lisbon, Lisbon, Portugal
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University & Liverpool Heart and Chest Hospital, Liverpool, UK; Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Denmark
| | - Paul R J Ames
- Immune Response & Vascular Disease, iNOVA,4Health, Nova Medical School, Nova University Lisbon, Lisbon, Portugal; Department of Haematology, Dumfries Royal Infirmary, Cargenbridge, Dumfries, UK.
| |
Collapse
|
7
|
Richter P, Badescu MC, Rezus C, Ouatu A, Dima N, Popescu D, Burlui AM, Bratoiu I, Mihai IR, Rezus E. Antiphospholipid Antibodies as Key Players in Systemic Lupus Erythematosus: The Relationship with Cytokines and Immune Dysregulation. Int J Mol Sci 2024; 25:11281. [PMID: 39457063 PMCID: PMC11509045 DOI: 10.3390/ijms252011281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by an overproduction of cytokines, such as interleukins and interferons, contributing to systemic inflammation and tissue damage. Antiphospholipid syndrome is a thrombo-inflammatory autoimmune disease affecting a third of SLE patients. We performed an in-depth analysis of the available literature, and we highlighted the complex interplay between immunity, inflammation, and thrombosis, the three major pathogenic pathways that are trapped in a mutually reinforcing destructive loop.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
| | - Alexandra Maria Burlui
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| |
Collapse
|
8
|
Aguirre Del-Pino R, Monahan RC, Huizinga TWJ, Eikenboom J, Steup-Beekman GM. Risk Factors for Antiphospholipid Antibodies and Antiphospholipid Syndrome. Semin Thromb Hemost 2024; 50:817-828. [PMID: 38228166 DOI: 10.1055/s-0043-1776910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Persistence of serum antiphospholipid antibodies (aPL) is associated with a high thrombotic risk, both arterial and venous, and with pregnancy complications. Due to the potential morbidity and mortality associated with the presence of aPL, identifying and recognizing risk factors for the development of aPL and thrombosis in aPL carriers may help to prevent and reduce the burden of disease. Multiple elements are involved in the pathomechanism of aPL development and aPL-related thrombosis such as genetics, malignancy, and infections. This review will address the role of both well-known risk factors and their evolution, and of emerging risk factors, including COVID-19, in the development of aPL and thrombosis in aPL carriers.
Collapse
Affiliation(s)
- Rodrigo Aguirre Del-Pino
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Division of Rheumatology, A Coruña University Hospital (CHUAC), Galicia, Spain
| | - Rory C Monahan
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jeroen Eikenboom
- Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Gerda M Steup-Beekman
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Rheumatology, Haaglanden Medical Center, The Hague, The Netherlands
| |
Collapse
|
9
|
Del Giudice C, Mahe G, Thony F, Zuily S, Goyault G, Diard A, Loffroy R, Galanaud JP, Thouveny F, Quere I, Menez C, Jurus C, Pernod G, Pernes JM, Sapoval M. Venous recanalisation in the setting of post-thrombotic syndrome: An expert consensus from the French Society of Vascular Medicine (SFMV) and the French Society of Cardiovascular Imaging and Interventional Radiology (SFICV). JOURNAL DE MEDECINE VASCULAIRE 2024; 49:141-161. [PMID: 39278694 DOI: 10.1016/j.jdmv.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/04/2024] [Indexed: 09/18/2024]
Abstract
Several aspects of the management of post-thrombotic syndrome (PTS) are still a matter of debate, or not yet addressed in international guidelines. The objective of this expert consensus from the French Society of Vascular Medicine (SFMV) and the French Society of Cardiovascular Imaging (SFICV) was to define the main elements of diagnosis and treatment of this syndrome, and to develop a proposal for its preoperative, procedural and follow-up management. In this consensus, the following issues were addressed: clinical and ultrasound diagnosis; pre-procedural workup; indications and contraindications to venous recanalisation; procedures; clinical and duplex ultrasound reports; follow-up; long-term treatment; management of great saphenous vein incompetency; anticoagulant and antiplatelet therapy after venous stenting.
Collapse
Affiliation(s)
- Costantino Del Giudice
- Radiology Department, Institut Mutualiste Montsouris, 42, boulevard Jourdan, 75014 Paris, France
| | - Guillaume Mahe
- Vascular Medicine Unit, Rennes University Hospital, Rennes, France.
| | - Frederic Thony
- Grenoble-Alpes University, Department of Imaging and Interventional Radiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Stephane Zuily
- Vascular Medicine Division and Regional Competence Centre For Rare Vascular And Systemic Autoimmune Diseases, Nancy Regional University Hospital, Nancy, France
| | - Gilles Goyault
- Department of Vascular and Oncological Interventional Radiology, Institut Cardiovasculaire de Strasbourg (ICS), Clinique Rhena, Strasbourg, France
| | | | - Romaric Loffroy
- Department of Vascular and Interventional Radiology, Image-Guided Therapy Centre, François-Mitterrand University Hospital, 14, rue Paul-Gaffarel, BP 77908, 21079 Dijon, France
| | - Jean-Philippe Galanaud
- Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Francine Thouveny
- Vascular Radiology Department, Angers University Hospital, Angers, France
| | - Isabelle Quere
- Department of Vascular Medicine, Montpellier University Hospital, University of Montpellier, Montpellier, France
| | - Caroline Menez
- Department of Vascular Medicine, Grenoble-Alpes University Hospital, Grenoble, France
| | | | - Gilles Pernod
- Department of Vascular Medicine, Grenoble-Alpes University Hospital, Grenoble, France
| | - Jean-Marc Pernes
- Radiology Department, Institut Mutualiste Montsouris, 42, boulevard Jourdan, 75014 Paris, France
| | - Marc Sapoval
- Department of Interventional Radiology, Georges-Pompidou European Hospital, AP-HP, Inserm U970, Université Paris Cité, Faculté de Médecine, 75006 Paris, France
| |
Collapse
|
10
|
Nikolopoulos D, Loukogiannaki C, Sentis G, Garantziotis P, Manolakou T, Kapsala N, Nikoloudaki M, Pieta A, Flouda S, Parodis I, Bertsias G, Fanouriakis A, Filia A, Boumpas DT. Disentangling the riddle of systemic lupus erythematosus with antiphospholipid syndrome: blood transcriptome analysis reveals a less-pronounced IFN-signature and distinct molecular profiles in venous versus arterial events. Ann Rheum Dis 2024; 83:1132-1143. [PMID: 38609158 PMCID: PMC11420729 DOI: 10.1136/ard-2024-225664] [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: 02/12/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
INTRODUCTION Systemic lupus erythematosus with antiphospholipid syndrome (SLE-APS) represents a challenging SLE endotype whose molecular basis remains unknown. METHODS We analysed whole-blood RNA-sequencing data from 299 patients with SLE (108 SLE-antiphospholipid antibodies (aPL)-positive, including 67 SLE-APS; 191 SLE-aPL-negative) and 72 matched healthy controls (HC). Pathway enrichment analysis, unsupervised weighted gene coexpression network analysis and machine learning were applied to distinguish disease endotypes. RESULTS Patients with SLE-APS demonstrated upregulated type I and II interferon (IFN) pathways compared with HC. Using a 100-gene random forests model, we achieved a cross-validated accuracy of 75.6% in distinguishing these two states. Additionally, the comparison between SLE-APS and SLE-aPL-negative revealed 227 differentially expressed genes, indicating downregulation of IFN-α and IFN-γ signatures, coupled with dysregulation of the complement cascade, B-cell activation and neutrophil degranulation. Unsupervised analysis of SLE transcriptome identified 21 gene modules, with SLE-APS strongly linked to upregulation of the 'neutrophilic/myeloid' module. Within SLE-APS, venous thromboses positively correlated with 'neutrophilic/myeloid' and 'B cell' modules, while arterial thromboses were associated with dysregulation of 'DNA damage response (DDR)' and 'metabolism' modules. Anticardiolipin and anti-β2GPI positivity-irrespective of APS status-were associated with the 'neutrophilic/myeloid' and 'protein-binding' module, respectively. CONCLUSIONS There is a hierarchical upregulation and-likely-dependence on IFN in SLE with the highest IFN signature observed in SLE-aPL-negative patients. Venous thrombotic events are associated with neutrophils and B cells while arterial events with DDR and impaired metabolism. This may account for their differential requirements for anticoagulation and provide rationale for the potential use of mTOR inhibitors such as sirolimus and the direct fIIa inhibitor dabigatran in SLE-APS.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Catherine Loukogiannaki
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh, AG Groningen, Τhe Netherlands
| | - George Sentis
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Panagiotis Garantziotis
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Theodora Manolakou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Noemin Kapsala
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Myrto Nikoloudaki
- Rheumatology, University of Crete School of Medicine, Iraklio, Crete, Greece
| | - Antigone Pieta
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Flouda
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - George Bertsias
- Rheumatology, University of Crete School of Medicine, Iraklio, Crete, Greece
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Antonis Fanouriakis
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Filia
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios T Boumpas
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
11
|
Varotto L, Spigolon L, Dotto A, Leonardi D, Bragantini G, Cerrito LF, Deluca C, Hoxha A. Myocardial Involvement in Catastrophic Antiphospholipid Syndrome during Pregnancy or Puerperium: A Case of a Young Breastfeeding Woman and Literature Review. J Clin Med 2024; 13:4732. [PMID: 39200874 PMCID: PMC11355751 DOI: 10.3390/jcm13164732] [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: 07/08/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 09/02/2024] Open
Abstract
Catastrophic Antiphospholipid Syndrome (CAPS) is a rare complication that can occur in patients with Antiphospholipid Syndrome (APS). CAPS occurs even more rarely during pregnancy/puerperium and pregnant patients, even less likely to show cardiac involvement without signs of damage on ultrasound and angiography with non-obstructive coronary arteries. We present a case of a 26-year-old breastfeeding woman, the youngest described with CAPS and acute myocardial infarction, whose diagnosis was made with cardiac magnetic resonance imaging (CMRI). A literature review of pregnant patients with similar problems was performed. There are diagnostic and therapeutic difficulties in treating these patients. CMRI demonstrated a transmural late enhancement area. A combination of therapies led to rapid clinical improvement. CMRI is an underused tool that reaffirms the pathophysiology of CAPS and leads clinicians to the possibility of a diffuse thrombotic process. CAPS involves more organs with high mortality rates. CMRI could be optimized in order to reach an early diagnosis and the most effective treatment. This study provides real-world evidence of the feasibility of MRI in a primary care setting during pregnancy/puerperium. Evidence from this study may influence future APS screening and inform policymakers regarding the use of leading MRI technology in the detection of the thrombotic process in a primary care setting.
Collapse
Affiliation(s)
- Leonardo Varotto
- Department of Cardiology, San Bortolo Hospital, viale Rodolfi 37, 36100 Vicenza, Italy; (G.B.); (L.F.C.)
| | - Luca Spigolon
- Department of Radiology, San Bortolo Hospital, Viale Rodolfi 37, 36100 Vicenza, Italy;
| | - Alberto Dotto
- Department of Medicine—Division of Cardiology, University of Verona, Piazzale A. Stefani 1, 37126 Verona, Italy; (A.D.); (D.L.)
| | - Denis Leonardi
- Department of Medicine—Division of Cardiology, University of Verona, Piazzale A. Stefani 1, 37126 Verona, Italy; (A.D.); (D.L.)
| | - Giulia Bragantini
- Department of Cardiology, San Bortolo Hospital, viale Rodolfi 37, 36100 Vicenza, Italy; (G.B.); (L.F.C.)
| | - Luca Felice Cerrito
- Department of Cardiology, San Bortolo Hospital, viale Rodolfi 37, 36100 Vicenza, Italy; (G.B.); (L.F.C.)
| | - Cristina Deluca
- Department of Neurology, San Bortolo Hospital, Viale Rodolfi 37, 36100 Vicenza, Italy;
| | - Ariela Hoxha
- Internal Medicine Unit, Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padova, Italy;
| |
Collapse
|
12
|
Manning JE, Arachchillage DJ. Dilemmas in the diagnosis and management of antiphospholipid syndrome. J Thromb Haemost 2024; 22:2156-2170. [PMID: 38705387 DOI: 10.1016/j.jtha.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 05/07/2024]
Abstract
Antiphospholipid syndrome (APS) is characterized by thrombosis (which may be venous, arterial, or microvascular) and/or pregnancy morbidity in association with persistently positive antiphospholipid antibodies. Although thrombosis and pregnancy morbidity are the main clinical criteria for a diagnosis of APS in the revised Sapporo (Sydney) criteria, recently published American College of Rheumatology/European Alliance of Associations for Rheumatology classification criteria for APS have significantly refined the diagnostic algorithm to include a scoring system clustered into 6 clinical domains (macrovascular venous thromboembolism, macrovascular arterial thrombosis, microvascular thrombosis, obstetric, cardiac valve, and hematologic). Diagnosis of APS is complicated by the fact that significant heterogeneity exists in patients' clinical presentation, underlying vascular risk factors, and methods of detecting antiphospholipid antibodies. Despite the autoimmune nature of APS, anticoagulation remains the main strategy for secondary prevention of thrombosis. Furthermore, optimal antithrombotic treatment in APS patients with arterial thrombosis remains controversial due to a paucity of data from randomized controlled studies. In this paper, we present 2 cases and highlight the diagnostic and therapeutic challenges they pose and how we approach them in the light of current evidence.
Collapse
Affiliation(s)
- James E Manning
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom; Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Deepa J Arachchillage
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom; Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
13
|
Feng W, Qiao J, Tan Y, Liu Q, Wang Q, Yang B, Yang S, Cui L. Interaction of antiphospholipid antibodies with endothelial cells in antiphospholipid syndrome. Front Immunol 2024; 15:1361519. [PMID: 39044818 PMCID: PMC11263079 DOI: 10.3389/fimmu.2024.1361519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease with arteriovenous thrombosis and recurrent miscarriages as the main clinical manifestations. Due to the complexity of its mechanisms and the diversity of its manifestations, its diagnosis and treatment remain challenging issues. Antiphospholipid antibodies (aPL) not only serve as crucial "biomarkers" in diagnosing APS but also act as the "culprits" of the disease. Endothelial cells (ECs), as one of the core target cells of aPL, bridge the gap between the molecular level of these antibodies and the tissue and organ level of pathological changes. A more in-depth exploration of the relationship between ECs and the pathogenesis of APS holds the potential for significant advancements in the precise diagnosis, classification, and therapy of APS. Many researchers have highlighted the vital involvement of ECs in APS and the underlying mechanisms governing their functionality. Through extensive in vitro and in vivo experiments, they have identified multiple aPL receptors on the EC membrane and various intracellular pathways. This article furnishes a comprehensive overview and summary of these receptors and signaling pathways, offering prospective targets for APS therapy.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| |
Collapse
|
14
|
Bobrova LA, Kozlovskaya NL. [Lupus nephritis and thrombotic microangiopathy: A review]. TERAPEVT ARKH 2024; 96:628-634. [PMID: 39106505 DOI: 10.26442/00403660.2024.06.202731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 07/07/2024] [Indexed: 08/09/2024]
Abstract
Lupus nephritis (LN) is one of the most common organ-specific manifestations of systemic lupus erythematosus (SLE). Various clinical signs of LN develop in at least 50% of patients with SLE. In addition to LN, the spectrum of renal lesions associated with SLE also includes vascular pathology. One of the variants of renal microvascular injury is thrombotic microangiopathy (TMA), the mechanisms of which are diverse. The review focuses on the main forms of TMA, including antiphospholipid syndrome and nephropathy associated with antiphospholipid syndrome, TMA caused by complement system regulation disorders and deficiency of ADAMTS13. In most cases, these forms of TMA are combined with LN. However, they may also exist as a single form of kidney damage. This article discusses the TMA pathogenesis, the impact on kidney prognosis, and treatment options.
Collapse
Affiliation(s)
- L A Bobrova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - N L Kozlovskaya
- Patrice Lumumba People's Friendship University of Russia
- Yeramishantsev City Clinical Hospital
| |
Collapse
|
15
|
Zerbib L, Ladraa S, Fraissenon A, Bayard C, Firpion M, Venot Q, Protic S, Hoguin C, Thomas A, Fraitag S, Duong JP, Kaltenbach S, Balducci E, Lefevre C, Villarese P, Asnafi V, Broissand C, Goudin N, Nemazanyy I, Autret G, Tavitian B, Legendre C, Arzouk N, Minard-Colin V, Chopinet C, Dussiot M, Adams DM, Mirault T, Guibaud L, Isenring P, Canaud G. Targeted therapy for capillary-venous malformations. Signal Transduct Target Ther 2024; 9:146. [PMID: 38880808 PMCID: PMC11180659 DOI: 10.1038/s41392-024-01862-9] [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: 10/02/2023] [Revised: 04/19/2024] [Accepted: 05/12/2024] [Indexed: 06/18/2024] Open
Abstract
Sporadic venous malformations are genetic conditions primarily caused by somatic gain-of-function mutation of PIK3CA or TEK, an endothelial transmembrane receptor signaling through PIK3CA. Venous malformations are associated with pain, bleedings, thrombosis, pulmonary embolism, esthetic deformities and, in severe cases, life-threatening situations. No authorized medical treatment exists for patients with venous malformations. Here, we created a genetic mouse model of PIK3CA-related capillary venous malformations that replicates patient phenotypes. We showed that these malformations only partially signal through AKT proteins. We compared the efficacy of different drugs, including rapamycin, a mTORC1 inhibitor, miransertib, an AKT inhibitor and alpelisib, a PI3Kα inhibitor at improving the lesions seen in the mouse model. We demonstrated the effectiveness of alpelisib in preventing vascular malformations' occurrence, improving the already established ones, and prolonging survival. Considering these findings, we were authorized to treat 25 patients with alpelisib, including 7 children displaying PIK3CA (n = 16) or TEK (n = 9)-related capillary venous malformations resistant to usual therapies including sirolimus, debulking surgical procedures or percutaneous sclerotherapies. We assessed the volume of vascular malformations using magnetic resonance imaging (MRI) for each patient. Alpelisib demonstrated improvement in all 25 patients. Vascular malformations previously considered intractable were reduced and clinical symptoms were attenuated. MRI showed a decrease of 33.4% and 27.8% in the median volume of PIK3CA and TEK malformations respectively, over 6 months on alpelisib. In conclusion, this study supports PI3Kα inhibition as a promising therapeutic strategy in patients with PIK3CA or TEK-related capillary venous malformations.
Collapse
Affiliation(s)
- Lola Zerbib
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sophia Ladraa
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Antoine Fraissenon
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
- CREATIS UMR 5220, Villeurbanne, 69100, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
| | - Charles Bayard
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Marina Firpion
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Quitterie Venot
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sanela Protic
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Clément Hoguin
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Amandine Thomas
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sylvie Fraitag
- Service d'Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Jean-Paul Duong
- Université Paris Cité, Paris, France
- Service d'Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Sophie Kaltenbach
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Estelle Balducci
- Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Coline Lefevre
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Patrick Villarese
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Vahid Asnafi
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | - Nicolas Goudin
- Necker Bio-Image Analysis, INSERM US24/CNRS UMS 3633, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Gwennhael Autret
- Plateforme Imageries du Vivant, Université de Paris, PARCC, INSERM, Paris, France
| | - Bertrand Tavitian
- Plateforme Imageries du Vivant, Université de Paris, PARCC, INSERM, Paris, France
| | - Christophe Legendre
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Nadia Arzouk
- Service de Transplantation, Hôpital La Pitié Salpétrière, AP-HP, Paris, France
| | - Veronique Minard-Colin
- Department of Pediatric and Adolescent Oncology, INSERM 1015, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Caroline Chopinet
- Service de Physiologie & Explorations Fonctionnelles Cardiovasculaires, CHU de Lille, Lille, 59000, France
| | - Michael Dussiot
- INSERM U1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Denise M Adams
- Division of Oncology, Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tristan Mirault
- Université Paris Cité, Paris, France
- Service de Médecine Vasculaire, hôpital Européen Georges-Pompidou, Paris, France
| | - Laurent Guibaud
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Quebec, QC, G1R2J6, Canada
| | - Guillaume Canaud
- Université Paris Cité, Paris, France.
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.
- Unité de médecine translationnelle et thérapies ciblées, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
- CNRS UMR8253, Institut Necker-Enfants Malades, Paris, France.
| |
Collapse
|
16
|
Yamaguchi J, Isnard P, Robil N, de la Grange P, Hoguin C, Schmitt A, Hummel A, Megret J, Goudin N, Luka M, Ménager MM, Masson C, Zarhrate M, Bôle-Feysot C, Janiszewska M, Polyak K, Dairou J, Baldassari S, Baulac S, Broissand C, Legendre C, Terzi F, Canaud G. PIK3CA inhibition in models of proliferative glomerulonephritis and lupus nephritis. J Clin Invest 2024; 134:e176402. [PMID: 38842935 PMCID: PMC11290976 DOI: 10.1172/jci176402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
Proliferative glomerulonephritis is a severe condition that often leads to kidney failure. There is a significant lack of effective treatment for these disorders. Here, following the identification of a somatic PIK3CA gain-of-function mutation in podocytes of a patient, we demonstrate using multiple genetically engineered mouse models, single-cell RNA sequencing, and spatial transcriptomics the crucial role played by this pathway for proliferative glomerulonephritis development by promoting podocyte proliferation, dedifferentiation, and inflammation. Additionally, we show that alpelisib, a PI3Kα inhibitor, improves glomerular lesions and kidney function in different mouse models of proliferative glomerulonephritis and lupus nephritis by targeting podocytes. Surprisingly, we determined that pharmacological inhibition of PI3Kα affects B and T lymphocyte populations in lupus nephritis mouse models, with a decrease in the production of proinflammatory cytokines, autoantibodies, and glomerular complement deposition, which are all characteristic features of PI3Kδ inhibition, the primary PI3K isoform expressed in lymphocytes. Importantly, PI3Kα inhibition does not impact lymphocyte function under normal conditions. These findings were then confirmed in human lymphocytes isolated from patients with active lupus nephritis. In conclusion, we demonstrate the major role played by PI3Kα in proliferative glomerulonephritis and show that in this condition, alpelisib acts on both podocytes and the immune system.
Collapse
Affiliation(s)
- Junna Yamaguchi
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Pierre Isnard
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d’Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Noémie Robil
- Genosplice Technology, Paris Biotech Santé, Paris, France
| | | | - Clément Hoguin
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | | | - Aurélie Hummel
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Jérôme Megret
- Structure Fédérative de Recherche Necker, INSERM US24, CNRS UAR 3633, Institut Necker-Enfants Malades, Paris, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, INSERM US24, CNRS UAR 3633, Institut Necker-Enfants Malades, Paris, France
| | - Marine Luka
- Inflammatory Responses and Transcriptomic Networks in Diseases
- INSERM U1163
| | - Mickaël M. Ménager
- Inflammatory Responses and Transcriptomic Networks in Diseases
- INSERM U1163
| | - Cécile Masson
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, and
| | | | | | - Michalina Janiszewska
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, Florida, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Julien Dairou
- Université Paris Cité, Paris, France
- Laboratoire de Chimie et Biologie Pharmacologiques et Toxicologiques, Paris, France
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | | | - Christophe Legendre
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Fabiola Terzi
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Guillaume Canaud
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| |
Collapse
|
17
|
Laniak OT, Winans T, Patel A, Park J, Perl A. Redox Pathogenesis in Rheumatic Diseases. ACR Open Rheumatol 2024; 6:334-346. [PMID: 38664977 PMCID: PMC11168917 DOI: 10.1002/acr2.11668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 06/14/2024] Open
Abstract
Despite being some of the most anecdotally well-known roads to pathogenesis, the mechanisms governing autoimmune rheumatic diseases are not yet fully understood. The overactivation of the cellular immune system and the characteristic development of autoantibodies have been linked to oxidative stress. Typical clinical manifestations, such as joint swelling and deformities and inflammation of the skin and internal organs, have also been connected directly or indirectly to redox mechanisms. The differences in generation and restraint of oxidative stress provide compelling evidence for the broad variety in pathology among rheumatic diseases and explain some of the common triggers and discordant manifestations in these diseases. Growing evidence of redox mechanisms in pathogenesis has provided a broad array of new potential therapeutic targets. Here, we explore the mechanisms by which oxidative stress is generated, explore its roles in autoimmunity and end-organ damage, and discuss how individual rheumatic diseases exhibit unique features that offer targets for therapeutic interventions.
Collapse
Affiliation(s)
- Olivia T. Laniak
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Thomas Winans
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Akshay Patel
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Joy Park
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Andras Perl
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| |
Collapse
|
18
|
Knight JS, Erkan D. Rethinking antiphospholipid syndrome to guide future management and research. Nat Rev Rheumatol 2024; 20:377-388. [PMID: 38702511 DOI: 10.1038/s41584-024-01110-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 05/06/2024]
Abstract
Antiphospholipid syndrome (APS) consists of thrombotic, non-thrombotic and obstetric clinical manifestations developing in individuals with persistent antiphospholipid antibodies (aPL). Although researchers have made progress in characterizing different clinical phenotypes of aPL-positive people, the current approach to clinical management is still mostly based on a 'one size fits all' strategy, which is derived from the results of a limited number of prospective, controlled studies. With the 2023 publication of the ACR-EULAR APS classification criteria, it is now possible to rethink APS, to lay the groundwork for subphenotyping through novel pathophysiology-informed approaches, and to set a future APS research agenda guided by unmet needs in clinical management.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Tektonidou MG, Vlachogiannis NI, Sfikakis PP. T cell involvement in antiphospholipid syndrome. Clin Immunol 2024; 263:110218. [PMID: 38640985 DOI: 10.1016/j.clim.2024.110218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
Antiphospholipid syndrome (APS) is a systemic autoimmune disorder characterized by arterial and venous thrombosis, and obstetric complications in the presence of antiphospholipid antibodies (aPL), including lupus anticoagulant, anticardiolipin and anti-β2-glycoprotein I antibodies. APS manifests as single, often as recurrent events, and rarely as a catastrophic condition. Most studies of APS pathogenesis to date have focused on the prothrombotic role of aPL, while innate immune responses such as monocyte, complement and neutrophil activation have been also recognized as part of the thrombo-inflammatory cascade in APS. While the presence of autoreactive T cells against β2-glycoprotein I has been long known, less data are available on their pathogenetic role in APS. In this review, we summarize current knowledge on the involvement of T cells in APS pathophysiology, alterations of T cell subsets in peripheral blood, and clinical associations. We also highlight potential therapeutic opportunities by targeting T helper-B cell interactions in these patients.
Collapse
Affiliation(s)
- Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine and Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Nikolaos I Vlachogiannis
- First Department of Propaedeutic Internal Medicine and Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine and Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
20
|
Sircana MC, Erre GL, Castagna F, Manetti R. Crosstalk between Inflammation and Atherosclerosis in Rheumatoid Arthritis and Systemic Lupus Erythematosus: Is There a Common Basis? Life (Basel) 2024; 14:716. [PMID: 38929699 PMCID: PMC11204900 DOI: 10.3390/life14060716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the context of these systemic connective tissue diseases. Over the past few years, our understanding of the pathophysiology of atherosclerosis has changed from it being a lipid-centric to an inflammation-centric process. In this review, we examine the pathogenesis of atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis, the two most common systemic connective tissue diseases, and consider them as emblematic models of the effect of chronic inflammation on the human body. We explore the roles of the inflammasome, cells of the innate and acquired immune system, neutrophils, macrophages, lymphocytes, chemokines and soluble pro-inflammatory cytokines in rheumatoid arthritis and systemic lupus erythematosus, and the roles of certain autoantigens and autoantibodies, such as oxidized low-density lipoprotein and beta2-glycoprotein, which may play a pathogenetic role in atherosclerosis progression.
Collapse
Affiliation(s)
| | | | | | - Roberto Manetti
- Department of Medical, Surgical and Pharmacology, University of Sassari, 07100 Sassari, Italy; (G.L.E.); (F.C.)
| |
Collapse
|
21
|
Tsoi A, Nikolopoulos D, Parodis I. Advances in the pharmacological management of systemic lupus erythematosus. Expert Opin Pharmacother 2024; 25:705-716. [PMID: 38756102 DOI: 10.1080/14656566.2024.2354457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Despite setbacks in clinical trials for systemic lupus erythematosus (SLE), three drugs have been approved for SLE and lupus nephritis (LN) treatment in the past decade. Several ongoing clinical trials, some viewed optimistically by the scientific community, underscore the evolving landscape. Emerging clinical data have established specific therapeutic targets in routine clinical practice for treating SLE, aiming to improve long-term outcomes. AREAS COVERED Research related to treatment of SLE and LN is discussed, focusing on randomized clinical trials during the last 5 years and recommendations for the management of SLE published by the European Alliance of Associations for Rheumatology (EULAR), American College of Rheumatology (ACR), Asia Pacific League of Associations for Rheumatology (APLAR), and Pan-American League of Associations of Rheumatology (PANLAR). EXPERT OPINION The landscape of SLE and LN treatments is evolving, as new drugs and combination treatment approaches redefine the traditional concepts of induction and maintenance treatment phases. As the therapeutic armamentarium in SLE continues to expand, the research focus is shifting from the imperative for new therapies to advancing our understanding of optimal treatment selection for individual patients, steering toward precision medicine strategies.
Collapse
Affiliation(s)
- Alexander Tsoi
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Dionysis Nikolopoulos
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
22
|
Jacobs L, Wauters N, Lablad Y, Morelle J, Taghavi M. Diagnosis and Management of Catastrophic Antiphospholipid Syndrome and the Potential Impact of the 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. Antibodies (Basel) 2024; 13:21. [PMID: 38534211 DOI: 10.3390/antib13010021] [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: 12/31/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Catastrophic antiphospholipid syndrome (CAPS) is a rare and life-threatening condition characterized by the persistence of antiphospholipid antibodies and occurrence of multiple vascular occlusive events. CAPS currently remains a diagnostic challenge and requires urgent treatment. The diagnosis of CAPS is made difficult by classification criteria used as diagnostic criteria in clinical practice, knowledge derived from retrospective data and case reports, confounding clinical and biological features, and its rapid onset and mortality. The absence of prospective studies of CAPS limits the strength of evidence for guideline treatment protocols. This comprehensive review summarizes the current understanding of the disease, and discusses how the 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria impact the definition and therapeutic management of CAPS, which is considered the most severe form of APS. The correct integration of 2023 ACR/EULAR APS classification criteria is poised to facilitate CAPS diagnosis, particularly in critical situations, offering a promising avenue for improved outcomes.
Collapse
Affiliation(s)
- Lucas Jacobs
- Internal Medicine Department, Brugmann University Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
- Internal Medicine Department, Tivoli University Hospital, Université Libre de Bruxelles, 7100 La Louvière, Belgium
| | - Nader Wauters
- Internal Medicine Department, Tivoli University Hospital, Université Libre de Bruxelles, 7100 La Louvière, Belgium
| | - Yahya Lablad
- Internal Medicine Department, Brugmann University Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
| | - Johann Morelle
- Division of Nephrology, Namur University Hospitals (CHU UCL Namur), 5000 Namur, Belgium
| | - Maxime Taghavi
- Internal Medicine Department, Brugmann University Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
| |
Collapse
|
23
|
Frederick R, Zolio L, Romas E, Ierino F. Eculizumab therapy and complement regulation in a case of resistant catastrophic antiphospholipid syndrome. BMJ Case Rep 2024; 17:e254449. [PMID: 38442982 PMCID: PMC10916160 DOI: 10.1136/bcr-2022-254449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Catastrophic antiphospholipid syndrome (CAPS) is a life-threatening form of antiphospholipid syndrome characterised by diffuse arterial and venous thrombosis, in the presence of positive antiphospholipid antibodies. The multiple sites of thrombosis in small, medium and large vessels progress to multiorgan failure, accounting for the high mortality rate associated with CAPS. Unregulated complement activation is increasingly recognised as critical to the pathogenesis of CAPS. Early diagnosis is essential to initiate prompt life-saving treatment with the triple therapy of anticoagulation, immunosuppression and either plasmapheresis or intravenous immunoglobulin. Among other immunosuppressive agents, eculizumab, a complement inhibitor has demonstrated efficacy in treatment-resistant cases.We report an instructive case of a woman presenting with both clinical and laboratory findings consistent with primary CAPS, resistant to initial treatment and responsive to eculizumab, with emphasis on genetic testing and implications for future therapy.
Collapse
Affiliation(s)
- Rachel Frederick
- Department of Nephrology, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
- Department of General Medicine, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
| | - Luigi Zolio
- Department of Rheumatology, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
| | - Evange Romas
- Department of Rheumatology, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| | - Frank Ierino
- Department of Nephrology, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Ruiz-Irastorza G, Tektonidou MG, Khamashta M. Anticoagulant and non-anticoagulant therapy in thrombotic antiphospholipid syndrome: old drugs and new treatment targets. Rheumatology (Oxford) 2024; 63:SI96-SI106. [PMID: 38320592 DOI: 10.1093/rheumatology/kead538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 02/08/2024] Open
Abstract
In this review, we discuss the current evidence on classic and newer oral anticoagulant therapy, older drugs such as HCQ and statins, and new potential treatment targets in APS. Vitamin K antagonists (VKAs) remain the cornerstone treatment for thrombotic events in APS. In patients fulfilling criteria for definite APS presenting with a first venous thrombosis, treatment with VKAs with a target international normalized ratio (INR) 2.0-3.0 is recommended. In patients with arterial thrombosis, treatment with VKA with target INR 2.0-3.0 or 3.0-4.0 is recommended by recent guidelines, considering the individual's bleeding and thrombosis recurrence risk. A combination of VKAs and low-dose aspirin (75-100 mg/daily) may also be considered. According to available evidence direct oral anticoagulants should be avoided in patients with arterial thrombosis and/or those with triple aPL positivity. Adjunctive treatment with HCQ and/or statins can be considered, especially in anticoagulation treatment-refractory APS. Potential targeted treatments in APS include B-cell targeting, complement inhibition, mammalian target of rapamycin inhibition, IFN targeting, adenosine receptors agonists, CD38 targeting or chimeric antigen receptor T-cell therapy. The safety and efficacy of these treatment targets needs to be examined in well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Guillermo Ruiz-Irastorza
- Autoimmune Diseases Research Unit, Biocruces Bizkaia Health Research Institute, The Basque Country, Bizkaia, Spain
- University of The Basque Country, The Basque Country, Bizkaia, Spain
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Munther Khamashta
- Department of Women & Children's Health, King's College London, London, UK
| |
Collapse
|
25
|
Barbhaiya M, Taghavi M, Zuily S, Domingues V, Chock EY, Tektonidou MG, Erkan D, Seshan SV. Efforts to Better Characterize "Antiphospholipid Antibody Nephropathy" for the 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria: Renal Pathology Subcommittee Report. J Rheumatol 2024; 51:150-159. [PMID: 37399462 DOI: 10.3899/jrheum.2022-1200] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE Antiphospholipid antibody (aPL) nephropathy (-N) can be challenging to recognize due to a lack of established classification or diagnostic criteria. As part of efforts to develop new antiphospholipid syndrome (APS) classification criteria (CC), the APS CC Renal Pathology Subcommittee aimed to better characterize the entity of aPL-N. METHODS We used a 4-pronged approach that included (1) administering Delphi surveys to worldwide APS physicians to generate aPL-N terminology; (2) conducting a literature review to demonstrate the association of nephropathy with aPL and identify published aPL-N histopathological terminology and descriptions; (3) evaluating aPL-N terminology used in renal biopsy reports from an international patient registry; and (4) evaluating proposed kidney pathologic features for aPL-N by assessment of international Renal Pathology Society (RPS) members. RESULTS After completing our metaanalysis demonstrating an association between nephropathy and aPL, we used Delphi surveys, a literature review, and international renal biopsy reports to develop a preliminary definition of aPL-N. The preliminary definition included include specific terms associated with acute (ie, thrombotic microangiopathy in glomeruli or arterioles/arteries) and chronic (ie, organized arterial or arteriolar microthrombi with or without recanalization, organized glomerular thrombi, fibrous and fibrocellular [arterial or arteriolar] occlusions, focal cortical atrophy with or without thyroidization, and fibrous intimal hyperplasia) lesions. Most RPS survey respondents agreed with this terminology and the importance of knowing aPL results for histopathological diagnosis. CONCLUSION Our results support the inclusion of aPL-N in the 2023 American College of Rheumatology/European Alliance of Associations for Rheumatology APS CC, and provide the most widely accepted terminology to date for both acute and chronic pathologic lesions of aPL-N.
Collapse
Affiliation(s)
- Medha Barbhaiya
- M. Barbhaiya, MD, MPH, D. Erkan, MD, MPH, Hospital for Special Surgery, and Weill Cornell Medicine, New York, New York, USA;
| | - Maxime Taghavi
- M. Taghavi, MD, Department of Nephrology, Centre Hospitalier Universitaire Brugmann, Université Libre de Bruxelles, Belgium
| | - Stephane Zuily
- S. Zuily, MD, PhD, Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aiguë et Chronique, and Centre Hospitalier Régional Universitaire de Nancy, Vascular Medicine Division, and French National Referral Center for Rare Autoimmune Diseases, Nancy, France
| | | | - Eugenia Y Chock
- E.Y. Chock, MD, MPH, Section of Rheumatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Maria G Tektonidou
- M.G. Tektonidou, First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Doruk Erkan
- M. Barbhaiya, MD, MPH, D. Erkan, MD, MPH, Hospital for Special Surgery, and Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
26
|
Taghavi M, Jabrane A, Jacobs L, Mesquita MDCF, Demulder A, Nortier J. Antiphospholipid Antibodies Associated with Native Arteriovenous Fistula Complications in Hemodialysis Patients: A Comprehensive Review of the Literature. Antibodies (Basel) 2024; 13:1. [PMID: 38247565 PMCID: PMC10801604 DOI: 10.3390/antib13010001] [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: 10/30/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Antiphospholipid antibody (aPL)-persistent positivity is frequent in hemodialysis (HD) patients. Native arteriovenous fistula (AVF) complications such as stenosis and thrombosis are among the most important causes of morbidity and mortality in hemodialysis patients. The association between aPL positivity and AVF thrombosis seems to now be well established. However, whether aPL positivity is associated with other AVF complications, such as maturation failure or stenosis, is not well known. Given the significant impact of AVF failure on patient's prognosis, it is of interest to further investigate this particular point in order to improve prevention, surveillance and treatment, and, ultimately, the patient's outcome. This literature review aims to report the recent literature on aPL-associated native AVF complications.
Collapse
Affiliation(s)
- Maxime Taghavi
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium; (A.J.); (L.J.); (M.D.C.F.M.); (J.N.)
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Abla Jabrane
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium; (A.J.); (L.J.); (M.D.C.F.M.); (J.N.)
| | - Lucas Jacobs
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium; (A.J.); (L.J.); (M.D.C.F.M.); (J.N.)
| | - Maria Do Carmo Filomena Mesquita
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium; (A.J.); (L.J.); (M.D.C.F.M.); (J.N.)
| | - Anne Demulder
- Laboratory of Hematology and Haemostasis LHUB-ULB, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium;
| | - Joëlle Nortier
- Nephrology and Dialysis Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), 1020 Brussels, Belgium; (A.J.); (L.J.); (M.D.C.F.M.); (J.N.)
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
27
|
De SK. Novel Pyrazino[2,3-b] Pyrazines as mTOR Kinase Inhibitors for Treating Cancer and other Diseases. Curr Med Chem 2024; 31:5657-5659. [PMID: 37493157 DOI: 10.2174/0929867331666230726112328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/18/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023]
Abstract
This paper describes the synthesis of some heteroaryl compounds and compositions comprising an effective amount of one or more such compounds and methods for treating or preventing cancer, inflammatory conditions, immunological conditions, metabolic conditions and conditions treatable or preventable by inhibition of a kinase pathway, comprising administering an adequate amount of a heteroaryl compound to a patient in need thereof. These compounds are mTOR/PI3K/Akt pathway inhibitors.
Collapse
Affiliation(s)
- Surya K De
- Department of Chemistry, Conju-Probe, San Diego, California, USA
- Bharath University, Chennai, Tamil Nadu, 600126, India
| |
Collapse
|
28
|
Kello N, Cho YM. Natural supplements in antiphospholipid syndrome: A case for further study. Clin Immunol 2024; 258:109848. [PMID: 38036277 DOI: 10.1016/j.clim.2023.109848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by thrombotic events and/or pregnancy complications in the presence of persistently positive antiphospholipid antibodies (aPL). Although long-term anticoagulation with vitamin K antagonists is considered standard of care, there is an unmet need for safe therapeutics as primary thromboprophylaxis or adjuncts to standard of care in APS. APS is driven by oxidative stress, procoagulant, proinflammatory and angiogenic pathways. For these reasons there has been an increased interest into the investigation of antithrombotic, anti-inflammatory and anti-oxidant properties of natural supplements in APS. The objective of this review is to summarize the mechanistic, epidemiologic and clinical evidence behind the use of natural supplements in APS, with a specific focus on vitamin D, omega-3 fatty acids, coenzyme Q10, gingerol, and isoquercetin. This review should serve as a compelling argument for the future study of natural supplements in APS.
Collapse
Affiliation(s)
- Nina Kello
- Northwell Health, Division of Rheumatology, Donald and Barbara Zucker School of Medicine, Great Neck, NY, United States of America.
| | - Young Min Cho
- Northwell Health, Division of Rheumatology, Donald and Barbara Zucker School of Medicine, Great Neck, NY, United States of America
| |
Collapse
|
29
|
Cole MA, Gerber GF, Chaturvedi S. Complement biomarkers in the antiphospholipid syndrome - Approaches to quantification and implications for clinical management. Clin Immunol 2023; 257:109828. [PMID: 37913840 PMCID: PMC10759159 DOI: 10.1016/j.clim.2023.109828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Complement is a major driver of antiphospholipid syndrome (APS) and a promising therapeutic target in refractory and catastrophic APS. Complement testing in APS is largely limited to research settings, and reliable, rapid-turnaround biomarkers are needed to predict those at risk for adverse clinical outcomes and most likely to benefit from complement inhibition. We review complement biomarkers and their association with thrombosis and obstetric outcomes, including: (i) complement proteins and activation fragments in the fluid phase; (ii) assays that evaluate complement on cell membranes (e.g. in vivo cell-bound complement fragments, hemolytic assays, and ex vivo 'functional' cell-based assays, and (iii) sequencing of complement genes. Current studies highlight the inconsistencies in testing both between studies and various aPL/APS subgroups, suggesting that either cell-based testing or multiplex panels employing a combination of biomarkers simultaneously may be most clinically relevant. Standardization of complement assays is needed to ensure reproducibility and establish clinically relevant applications.
Collapse
Affiliation(s)
- Michael A Cole
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gloria F Gerber
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shruti Chaturvedi
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
30
|
Ji L, Zhang Z, Perl A. The mTOR pathway in the antiphospholipid syndrome. MEDICAL REVIEW (2021) 2023; 3:511-513. [PMID: 38282799 PMCID: PMC10808841 DOI: 10.1515/mr-2023-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/26/2023] [Indexed: 01/30/2024]
Abstract
This perspective discussed the available evidence on the involvement of mTOR pathway in antiphospholipid syndrome (APS), from the aspects of endothelial cells, platelets, monocytes and anti-phospholipid antibodies (PLs), which may lead to future therapeutic applications of mTOR inhibition in APS.
Collapse
Affiliation(s)
- Lanlan Ji
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Zhuoli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, Division of Rheumatology and Clinical Immunology, State University of New York, UMU, College of Medicine, Syracuse, NY, USA
| |
Collapse
|
31
|
Zhao Y, Qi W, Huang C, Zhou Y, Wang Q, Tian X, Li M, Zhao Y, Zeng X, Zhao J. Serum Calprotectin as a Potential Predictor of Microvascular Manifestations in Patients with Antiphospholipid Syndrome. Rheumatol Ther 2023; 10:1769-1783. [PMID: 37906398 PMCID: PMC10654303 DOI: 10.1007/s40744-023-00610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Microvascular manifestations constitute a subtype of antiphospholipid syndrome, and those patients have relatively poor prognoses, so it is important to find markers for microvascular manifestations. This study was conducted to explore whether serum calprotectin could be a predictor of microvascular manifestations in antiphospholipid antibody (aPL)-positive patients. METHODS Consecutive patients with persistent aPL positivity referred to Peking Union Medical College Hospital and age- and sex-matched health controls (HCs) were included. Microvascular manifestations included antiphospholipid syndrome (APS) nephropathy, livedo reticularis, valvular lesions, non-stroke central nervous system manifestations, myocarditis, catastrophic APS, and other microvascular manifestations confirmed by pathology, imaging, or clinical diagnosis. Calprotectin was measured by an enzyme-linked immunosorbent assay (ELISA). The cutoff value was defined as mean + 2 standard deviations of HCs. Multivariable logistic regression analysis was used to analyze risk factors. Pearson correlation analysis was used to detect the correlation between calprotectin and other laboratory data. RESULTS Of the 466 patients included in the study, 281 (60.3%) patients met the 2006 Sydney Revised Classification Criteria; among the latter, 77.2% were patients with primary APS. The mean age was 39.10 ± 13.05 years old, and 77.0% were female. Thirty-eight age- and sex-matched HCs were included in the study. Serum calprotectin levels were increased in aPL-positive patients compared with HCs (649.66 ± 240.79 vs 484.62 ± 149.37 ng/ml, p < 0.001), and were increased in patients with microvascular manifestations compared with patients without (693.03 ± 271.90 vs 639.43 ± 232.06 ng/ml, p = 0.044). The cutoff value was 783.36 ng/ml. Ninety-three patients (20.0%) were positive for calprotectin. Calprotectin positivity was independently associated with microvascular manifestations (odds ratio [OR] 1.90, 95% confidence interval [CI] 1.07-3.36) and platelet count (PLT) < 100 (OR 2.04, 95% CI 1.08-3.88). Age (OR 0.98, 95% CI 0.96-1.00), systemic lupus erythematosus (OR 2.08, 95% CI 1.15-3.75), calprotectin positivity (OR 1.83, 95% CI 1.02-3.26), hypertension (OR 2.73, 95% CI 1.36-5.45), hemolytic anemia (OR 2.66, 95% CI 1.13-6.23), and anti-β2GPI antibodies (OR 2.06, 95% CI 1.11-3.83) could independently predict microvascular manifestations in aPL-positive patients. Serum calprotectin negatively correlated with PLT (R = - 0.101, p = 0.031). CONCLUSION Serum calprotectin levels are increased in aPL-positive patients and could be a potential predictor of microvascular manifestations.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wanting Qi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yangzhong Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| |
Collapse
|
32
|
Jiang H, Li ML, Zhao JL, Zhao Y. Antiphospholipid antibodies induced cerebrovascular occlusion without either arteriosclerosis or thrombosis. Int J Rheum Dis 2023; 26:2323-2325. [PMID: 37439583 DOI: 10.1111/1756-185x.14820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023]
Affiliation(s)
- Hui Jiang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ming L Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiu L Zhao
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
33
|
Mostafa M, Mahmoud A, Egiza HA, Niu C, Elbahnasawy M, Yusuf Y, Kouides P. Severe cutaneous necrosis in antiphospholipid syndrome. Am J Hematol 2023; 98:E328-E333. [PMID: 37713504 DOI: 10.1002/ajh.27067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 09/17/2023]
Affiliation(s)
| | - Amir Mahmoud
- Rochester General Hospital, Rochester, New York, USA
| | - Hebat-Allah Egiza
- Egypt-Japan University for Science and Technology, Alexandria, Egypt
| | - Chengu Niu
- Rochester General Hospital, Rochester, New York, USA
| | | | | | - Peter Kouides
- Rochester General Hospital, Rochester, New York, USA
| |
Collapse
|
34
|
Tang Z, Shi H, Chen C, Teng J, Dai J, Ouyang X, Liu H, Hu Q, Cheng X, Ye J, Su Y, Sun Y, Pan H, Wang X, Liu J, Su B, Yang C, Xu Y, Liu T. Activation of Platelet mTORC2/Akt Pathway by Anti-β2GP1 Antibody Promotes Thrombosis in Antiphospholipid Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:1818-1832. [PMID: 37381985 DOI: 10.1161/atvbaha.123.318978] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Anti-β2GP1 (β2-glycoprotein 1) antibodies are the primary pathogenic antibody to promote thrombosis in antiphospholipid syndrome (APS), yet the underlying mechanism remains obscure. We aimed to explore the intracellular pathway that mediated platelet activation. METHODS Platelets were isolated from patients with APS and subjected to RNA sequencing. Platelet aggregation, the release of platelet granules, platelet spreading, and clot retraction were detected to evaluate platelet activation. We purified anti-β2GP1 antibodies from patients with APS and the total IgG from healthy donors to stimulate platelets with/without FcγRIIA (Fcγ receptor IIA) blocking antibody or Akt (protein kinase B) inhibitor. Platelet-specific Sin1 (stress-activated protein kinase-interacting protein) deficiency mice were established. The thrombus model of inferior vena cava flow restriction, ferric chloride-induced carotid injury model, and laser-induced vessel wall injury in cremaster arterioles model were constructed after administration of anti-β2GP1 antibodies. RESULTS Combined RNA sequencing and bioinformatics analysis suggested that APS platelets exhibited increased levels of mRNA associated with platelet activation, which was in line with the hyperactivation of APS platelets in response to stimuli. Platelet activation in APS platelets was accompanied by upregulation of the mTORC2 (mammalian target of the rapamycin complex 2)/Akt pathway and increased levels of SIN1 phosphorylation at threonine 86. Anti-β2GP1 antibody derived from patients with APS enhanced platelet activation and upregulated the mTORC2/Akt pathway. Moreover, the Akt inhibitor weakened the potentiating effect of the anti-β2GP1 antibody on platelet activation. Notably, Sin1 deficiency suppresses anti-β2GP1 antibody-enhanced platelet activation in vitro and thrombosis in all 3 models. CONCLUSIONS This study elucidated the novel mechanism involving the mTORC2/Akt pathway, which mediates the promotion of platelet activation and induction of thrombosis by the anti-β2GP1 antibody. The findings suggest that SIN1 may be a promising therapeutic target for the treatment of APS.
Collapse
Affiliation(s)
- Zihan Tang
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Changming Chen
- Department of Laboratory Medicine, Ruijin Hospital (C.C., J.D., X.W.), Shanghai Jiao Tong University School of Medicine, China
| | - Jialin Teng
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Jing Dai
- Department of Laboratory Medicine, Ruijin Hospital (C.C., J.D., X.W.), Shanghai Jiao Tong University School of Medicine, China
| | - Xinxing Ouyang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Ministry of Education Key Laboratory of Cell Death and Differentiation (X.O., B.S.), Shanghai Jiao Tong University School of Medicine, China
- Department of Tumor Biology, Shanghai Chest Hospital (X.O.), Shanghai Jiao Tong University School of Medicine, China
| | - Honglei Liu
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Qiongyi Hu
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaobing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Yutong Su
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Yue Sun
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Haoyu Pan
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital (C.C., J.D., X.W.), Shanghai Jiao Tong University School of Medicine, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology (J.L., Y.X.), Shanghai Jiao Tong University School of Medicine, China
| | - Bing Su
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Ministry of Education Key Laboratory of Cell Death and Differentiation (X.O., B.S.), Shanghai Jiao Tong University School of Medicine, China
- Center for Human Translational Immunology at Shanghai Institute of Immunology, Ruijin Hospital (B.S.), Shanghai Jiao Tong University School of Medicine, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism (B.S.), Shanghai Jiao Tong University School of Medicine, China
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China (B.S.)
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology (J.L., Y.X.), Shanghai Jiao Tong University School of Medicine, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital (Z.T., H.S., J.T., H.L., Q.H., X.C., J.Y., Y. Su, Y. Sun, H.P., C.Y., T.L.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
35
|
Ruzdija D, Bertolo A, Pandya SK, Amaraneni A. A Case Report of Refractory Diffuse Alveolar Hemorrhage as a Complication of Antiphospholipid Syndrome and Review of Its Management. Cureus 2023; 15:e47267. [PMID: 38022217 PMCID: PMC10655834 DOI: 10.7759/cureus.47267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Diffuse alveolar hemorrhage is a rare complication of antiphospholipid syndrome with high mortality rates. Early diagnosis and treatment with steroids and immunosuppressive agents, along with the achievement of hemostasis and hemodynamic stability, is critical to improving outcomes. This case demonstrates the complexity of managing refractory diffuse alveolar hemorrhage in a 48-year-old woman with antiphospholipid syndrome requiring treatment with high-dose corticosteroids, rituximab, and cyclophosphamide.
Collapse
Affiliation(s)
- Denis Ruzdija
- Internal Medicine, University of Arizona, Tucson, USA
| | - Aaron Bertolo
- Internal Medicine, University of Arizona, Tucson, USA
| | | | - Akshay Amaraneni
- Hematology and Oncology, University of Arizona Cancer Center, Tucson, USA
| |
Collapse
|
36
|
Xourgia E, Tektonidou MG. Antiphospholipid syndrome nephropathy: Current knowledge and unanswered questions. Clin Immunol 2023; 255:109735. [PMID: 37572950 DOI: 10.1016/j.clim.2023.109735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
The definition of acute and chronic antiphospholipid syndrome (APS) nephropathy was recently updated using a multiphase methodology in the context of the development of the new APS classification criteria. Currently, there is no consensus for the treatment of APS nephropathy, which mainly relies on the general recommendations for the management of APS. Based on evidence from experimental studies and a few clinical studies and case series, targeted treatments such as B-cell depletion, anti-B-cell activating factor antibody, complement inhibition, mammalian target of rapamycin inhibition, and neutrophil extracellular traps or interferon targeting may show promise for the treatment of microvascular manifestations in APS, including APS nephropathy. Validation of the new APS nephropathy definition and/or efforts for improvement in proposed terminology, along with the assessment of the safety and efficacy of potential targeted treatments in randomized controlled trials, are major future research directions. In this review, we summarize the current knowledge of APS nephropathy and discuss unanswered questions.
Collapse
Affiliation(s)
- Eleni Xourgia
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece.
| |
Collapse
|
37
|
Dar S, Koirala S, Khan A, Bellary MD, Patel AV, Mathew B, Singh R, Baigam N, Razzaq W, Abdin ZU, Khawaja UA. A Comprehensive Literature Review on Managing Systemic Lupus Erythematosus: Addressing Cardiovascular Disease Risk in Females and Its Autoimmune Disease Associations. Cureus 2023; 15:e43725. [PMID: 37727166 PMCID: PMC10505685 DOI: 10.7759/cureus.43725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
This review aimed to evaluate the mechanism of premature cardiovascular disease (CVD) in systemic lupus erythematosus (SLE) patients, particularly in the female population, and emphasize the need for early management interventions; explore the association between SLE and two autoimmune diseases, myasthenia gravis (MG) and antiphospholipid antibody syndrome (APS), and their management strategies; and evaluate the effectiveness of pharmacological and non-pharmacological interventions in managing SLE, focusing on premenopausal females, females of childbearing age, and pregnant patients. We conducted a comprehensive literature review to achieve these objectives using various databases, including PubMed, Google Scholar, and Cochrane. The collected data were analyzed and synthesized to provide an evidence-based overview of SLE, its management strategies as an independent disease, and some disease associations. The treatment should be focused on remission, preventing organ damage, and improving the overall quality of life (QOL). Extensive emphasis should also be focused on diagnosing SLE and concurrent underlying secondary diseases timely and managing them appropriately.
Collapse
Affiliation(s)
- Saleha Dar
- Department of Adult Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sabina Koirala
- Department of Medicine, Gandaki Medical College, Pokhara, NPL
| | - Arooba Khan
- Department of Internal Medicine, Khyber Medical College, Peshawar, PAK
| | | | - Arya V Patel
- Department of Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand (NHL) Municipal Medical College, Ahmedabad, IND
| | - Bejoi Mathew
- Department of Internal Medicine, Sri Devaraj Urs Medical College, Kolar, IND
| | - Rahul Singh
- Department of Medicine, Armed Forces Medical College, Pune, IND
| | - Nahida Baigam
- Department of Medicine, Association of Physicians of Pakistani Descent of North America (APPNA), Westmont, USA
| | - Waleed Razzaq
- Department of Internal Medicine, Services Hospital Lahore, Lahore, PAK
| | - Zain U Abdin
- Department of Medicine, District Head Quarter Hospital, Faisalabad, PAK
| | - Uzzam Ahmed Khawaja
- Department of Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Department of Clinical and Translational Research, Dr. Ferrer BioPharma, South Miami, USA
| |
Collapse
|
38
|
Solé C, Royo M, Sandoval S, Moliné T, Cortés-Hernández J. Small-Extracellular-Vesicle-Derived miRNA Profile Identifies miR-483-3p and miR-326 as Regulators in the Pathogenesis of Antiphospholipid Syndrome (APS). Int J Mol Sci 2023; 24:11607. [PMID: 37511365 PMCID: PMC10380201 DOI: 10.3390/ijms241411607] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/06/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Primary antiphospholipid syndrome (PAPS) is a systemic autoimmune disease associated with recurrent thrombosis and/or obstetric morbidity with persistent antiphospholipid antibodies (aPL). Although these antibodies drive endothelial injury and thrombophilia, the underlying molecular mechanism is still unclear. Small extracellular vesicles (sEVs) contain miRNAs, key players in intercellular communication. To date, the effects of miRNA-derived sEVs in PAPS are not well understood. We characterised the quantity, cellular origin and miRNA profile of sEVs isolated from thrombotic APS patients (PAPS, n = 50), aPL-carrier patients (aPL, n = 30) and healthy donors (HD, n = 30). We found higher circulating sEVs mainly of activated platelet origin in PAPS and aPL patients compared to HD, that were highly engulfed by HUVECs and monocyte. Through miRNA-sequencing analysis, we identified miR-483-3p to be differentially upregulated in sEVs from patients with PAPS and aPL, and miR-326 to be downregulated only in PAPS sEVs. In vitro studies showed that miR-483-3p overexpression in endothelial cells induced an upregulation of the PI3K-AKT pathway that led to endothelial proliferation/dysfunction. MiR-326 downregulation induced NOTCH pathway activation in monocytes with the upregulation of NFKB1, tissue factor and cytokine production. These results provide evidence that miRNA-derived sEVs contribute to APS pathogenesis by producing endothelial cell proliferation, monocyte activation and adhesion/procoagulant factors.
Collapse
Affiliation(s)
- Cristina Solé
- Rheumatology Research Group—Lupus Unit, Vall d’Hebrón University Hospital, Vall d’Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (M.R.); (S.S.); (J.C.-H.)
| | - Maria Royo
- Rheumatology Research Group—Lupus Unit, Vall d’Hebrón University Hospital, Vall d’Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (M.R.); (S.S.); (J.C.-H.)
| | - Sebastian Sandoval
- Rheumatology Research Group—Lupus Unit, Vall d’Hebrón University Hospital, Vall d’Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (M.R.); (S.S.); (J.C.-H.)
| | - Teresa Moliné
- Department of Pathology, Vall d’Hebrón University Hospital, 08035 Barcelona, Spain;
| | - Josefina Cortés-Hernández
- Rheumatology Research Group—Lupus Unit, Vall d’Hebrón University Hospital, Vall d’Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (M.R.); (S.S.); (J.C.-H.)
| |
Collapse
|
39
|
Raccis M, Dossi F, Bernardelli A, Aste M, Berisso G, Parodi G. Catastrophic antiphospholipid syndrome immediately after coronary angiography: the unexpected complication. Blood Coagul Fibrinolysis 2023; 34:239-243. [PMID: 37115989 DOI: 10.1097/mbc.0000000000001219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
A 44-year-old woman with obstetric antiphospholipid syndrome (APS) presented to our institution with suspected non ST-elevation myocardial infarction (NSTEMI). Shortly after coronary angiography, she developed diplopia, hypotension and chest pain with inferior ST elevation in 12-lead ECG. According to multidisciplinary evaluation, she promptly underwent systemic thrombolysis, with clinical and haemodynamic improvement. Eventually, a diagnosis of catastrophic APS was made, with multiorgan ischemic involvement confirmed by blood examinations and multimodal imaging techniques. A pluridisciplinary approach was central to define optimal medical therapy and in-hospital management that lead to clinical condition improvement at discharge. In this case, catastrophic APS was triggered or worsened by catheters insertion and invasive manoeuvres within the arterial lumen during coronary angiography. Recognizing catastrophic APS and its potential triggers, can be helpful to deliver prompt and accurate medical assistance. Moreover, in APS patients undergoing coronary angiography, preventive strategies are important to prevent possible unfavourable evolution in catastrophic APS.
Collapse
Affiliation(s)
- Mario Raccis
- Arrhythmologic Centre, Department of Cardiology, ASL4 Liguria, Lavagna
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari
| | - Filippo Dossi
- Arrhythmologic Centre, Department of Cardiology, ASL4 Liguria, Lavagna
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari
| | - Alice Bernardelli
- Arrhythmologic Centre, Department of Cardiology, ASL4 Liguria, Lavagna
- Department of Internal Medicine and Medical Specialities (DIMI), University of Genoa, Genova
| | - Milena Aste
- Arrhythmologic Centre, Department of Cardiology, ASL4 Liguria, Lavagna
| | | | - Guido Parodi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari
| |
Collapse
|
40
|
Bahar Keleşoğlu Dinçer A, Erkan D. The ABCs of antiphospholipid syndrome. Arch Rheumatol 2023; 38:163-173. [PMID: 37680521 PMCID: PMC10481699 DOI: 10.46497/archrheumatol.2023.41875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 05/28/2023] [Indexed: 09/09/2023] Open
Abstract
Antiphospholipid syndrome (APS) is a thromboinflammatory syndrome characterized by thrombotic, microvascular, obstetric, or non-thrombotic events in the setting of persistent antiphospholipid antibodies (aPL), namely anticardiolipin antibody (aCL), anti-β2 glycoprotein-I antibody (aβ2GPI), and lupus anticoagulant (LA). The diagnosis of APS requires careful assessment of the aPL profile, the clinical phenotype, and additional risk factors. The standard management of aPL-related thrombosis is anticoagulation, which is not effective for microvascular and non-thrombotic events. In parallel to our improved understanding of aPL-related mechanisms, the role of immunosuppression has been increasingly investigated. In this review, we summarize the basic concepts and future perspectives in APS.
Collapse
Affiliation(s)
- Ayşe Bahar Keleşoğlu Dinçer
- Division of Rheumatology, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Türkiye
- Hospital for Special Surgery, New York, NY, USA
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery, and Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
41
|
Misra DP, Singh K, Sharma A, Agarwal V. Arterial wall fibrosis in Takayasu arteritis and its potential for therapeutic modulation. Front Immunol 2023; 14:1174249. [PMID: 37256147 PMCID: PMC10225504 DOI: 10.3389/fimmu.2023.1174249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/20/2023] [Indexed: 06/01/2023] Open
Abstract
Arterial wall damage in Takayasu arteritis (TAK) can progress despite immunosuppressive therapy. Vascular fibrosis is more prominent in TAK than in giant cell arteritis (GCA). The inflamed arterial wall in TAK is infiltrated by M1 macrophages [which secrete interleukin-6 (IL-6)], which transition to M2 macrophages once the inflammation settles. M2 macrophages secrete transforming growth factor beta (TGF-β) and glycoprotein non-metastatic melanoma protein B (GPNMB), both of which can activate fibroblasts in the arterial wall adventitia. Mast cells in the arterial wall of TAK also activate resting adventitial fibroblasts. Th17 lymphocytes play a role in both TAK and GCA. Sub-populations of Th17 lymphocytes, Th17.1 lymphocytes [which secrete interferon gamma (IFN-γ) in addition to interleukin-17 (IL-17)] and programmed cell death 1 (PD1)-expressing Th17 (which secrete TGF-β), have been described in TAK but not in GCA. IL-6 and IL-17 also drive fibroblast activation in the arterial wall. The Th17 and Th1 lymphocytes in TAK demonstrate an activation of mammalian target organ of rapamycin 1 (mTORC1) driven by Notch-1 upregulation. A recent study reported that the enhanced liver fibrosis score (derived from serum hyaluronic acid, tissue inhibitor of metalloproteinase 1, and pro-collagen III amino-terminal pro-peptide) had a moderate-to-strong correlation with clinically assessed and angiographically assessed vascular damage. In vitro experiments suggest the potential to target arterial wall fibrosis in TAK with leflunomide, tofacitinib, baricitinib, or mTORC1 inhibitors. Since arterial wall inflammation is followed by fibrosis, a strategy of combining immunosuppressive agents with drugs that have an antifibrotic effect merits exploration in future clinical trials of TAK.
Collapse
Affiliation(s)
- Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Kritika Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Aman Sharma
- Clinical Immunology and Rheumatology Services, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| |
Collapse
|
42
|
Saadh MJ, Kazemi K, Khorramdelazad H, Mousavi MJ, Noroozi N, Masoumi M, Karami J. Role of T cells in the pathogenesis of systemic lupus erythematous: Focus on immunometabolism dysfunctions. Int Immunopharmacol 2023; 119:110246. [PMID: 37148769 DOI: 10.1016/j.intimp.2023.110246] [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: 03/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
Evidence demonstrates that T cells are implicated in developing SLE, and each of them dominantly uses distinct metabolic pathways. Indeed, intracellular enzymes and availability of specific nutrients orchestrate fate of T cells and lead to differentiation of regulatory T cells (Treg), memory T cells, helper T cells, and effector T cells. The function of T cells in inflammatory and autoimmune responses is determined by metabolic processes and activity of their enzymes. Several studies were conducted to determine metabolic abnormalities in SLE patients and clarify how these modifications could control the functions of the involved T cells. Metabolic pathways such as glycolysis, mitochondrial pathways, oxidative stress, mTOR pathway, fatty acid and amino acid metabolisms are dysregulated in SLE T cells. Moreover, immunosuppressive drugs used in treating autoimmune diseases, including SLE, could affect immunometabolism. Developing drugs to regulate autoreactive T cell metabolism could be a promising therapeutic approach for SLE treatment. Accordingly, increased knowledge about metabolic processes paves the way to understanding SLE pathogenesis better and introduces novel therapeutic options for SLE treatment. Although monotherapy with metabolic pathways modulators might not be sufficient to prevent autoimmune disease, they may be an ideal adjuvant to reduce administration doses of immunosuppressive drugs, thus reducing drug-associated adverse effects. This review summarized emerging data about T cells that are involved in SLE pathogenesis, focusing on immunometabolism dysregulation and how these modifications could affect the disease development.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Department of Basic Sciences, Faculty of Pharmacy, Middle East University, Amman, Jordan; Applied Science Private University, Amman, Jordan
| | | | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, School of Para-Medicine, Bushehr University of Medical Sciences, Bushehr, Iran; Student Research and Technology Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Negar Noroozi
- Student Research and Technology Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Masoumi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran.
| | - Jafar Karami
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
43
|
Patsouras M, Alexopoulou E, Foutadakis S, Tsiki E, Karagianni P, Agelopoulos M, Vlachoyiannopoulos PG. Antiphospholipid antibodies induce proinflammatory and procoagulant pathways in endothelial cells. J Transl Autoimmun 2023; 6:100202. [PMID: 37216142 PMCID: PMC10197110 DOI: 10.1016/j.jtauto.2023.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/01/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia characterized by recurrent thrombotic events and/or pregnancy morbidity in the presence of antiphospholipid antibodies detected either as anti-cardiolipin, anti-β2 Glycoprotein I (anti-β2GPI) or Lupus anticoagulant (LA). Endothelial deregulation characterizes the syndrome. To address gene expression changes accompanying the development of autoimmune phenotype in endothelial cells in the context of APS, we performed transcriptomics analysis in Human Umbilical Vein Endothelial Cells (HUVECs) stimulated with IgG from APS patients and β2GPI, followed by intersection of RNA-seq data with published microarray and ChIP-seq results (Chromatin Immunoprecipitation). Our strategy revealed that during HUVEC activation diverse signaling pathways such as TNF-α, TGF-β, MAPK38, and Hippo are triggered as indicated by Gene Ontology (GO) classification and pathway analysis. Finally, cell biology approaches performed side-by-side in naïve and stimulated cultured HUVECs, as well as, in placenta specimens derived from Healthy donors (HDs) and APS-patients verified the evolution of an APS-characteristic gene expression program in endothelial cells during the initial stages of the disease's development.
Collapse
Affiliation(s)
- Markos Patsouras
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eirini Alexopoulou
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Spyros Foutadakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | - Eirini Tsiki
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Panagiota Karagianni
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Marios Agelopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | | |
Collapse
|
44
|
Liu X, Mao Z, Yuan M, Li L, Tan Y, Qu Z, Chen M, Yu F. Glomerular mTORC1 activation was associated with podocytes to endothelial cells communication in lupus nephritis. Lupus Sci Med 2023; 10:10/1/e000896. [PMID: 37147021 PMCID: PMC10163597 DOI: 10.1136/lupus-2023-000896] [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: 01/04/2023] [Accepted: 04/15/2023] [Indexed: 05/07/2023]
Abstract
OBJECTIVE This study was initiated to evaluate the mammalian target of the rapamycin (mTOR) signalling pathway involved in renal endothelial-podocyte crosstalk in patients with lupus nephritis (LN). METHODS We compared the kidney protein expression patterns of 10 patients with LN with severe endothelial-podocyte injury and 3 patients with non-severe endothelial-podocyte injury on formalin-fixed paraffin-embedded kidney tissues using label-free liquid chromatography-mass spectrometry for quantitative proteomics analysis. Podocyte injury was graded by foot process width (FPW). The severe group was referred to patients with both glomerular endocapillary hypercellularity and FPW >1240 nm. The non-severe group included patients with normal endothelial capillaries and FPW in the range of 619~1240 nm. Gene Ontology (GO) enrichment analyses were performed based on the protein intensity levels of differentially expressed proteins in each patient. An enriched mTOR pathway was selected, and the activation of mTOR complexes in renal biopsied specimens was further verified in 176 patients with LN. RESULTS Compared with those of the non-severe group, 230 proteins were upregulated and 54 proteins were downregulated in the severe group. Furthermore, GO enrichment analysis showed enrichment in the 'positive regulation of mTOR signalling' pathway. The glomerular activation of mTOR complex 1 (mTORC1) was significantly increased in the severe group compared with the non-severe group (p=0.034), and mTORC1 was located in podocytes and glomerular endothelial cells. Glomerular activation of mTORC1 was positively correlated with endocapillary hypercellularity (r=0.289, p<0.001) and significantly increased in patients with both endocapillary hypercellularity and FPW >1240 nm (p<0.001). CONCLUSIONS Glomerular mTORC1 was highly activated in patients with both glomerular endocapillary hypercellularity and podocyte injury, which might be involved in podocytes to endothelial cells communication in lupus nephritis.
Collapse
Affiliation(s)
- Xiaotian Liu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhaomin Mao
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Mo Yuan
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Linlin Li
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Tan
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhen Qu
- Department of Nephrology, Peking University International Hospital, Beijing, China
| | - Min Chen
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Feng Yu
- Department of Nephrology, Peking University International Hospital, Beijing, China
| |
Collapse
|
45
|
Sciascia S, Yazdany J, Moroni G, Becker JU, Seshan SV, Andrade D, Emmi G, Cuadrado MJ, Radin M, Cecchi I, De Simone E, Barreca A, Caroti L, Innocenti S, Fenoglio R, Roccatello D. Clinical-Pathological Characteristics of Renal Injuries Identify Different Clusters in Patients With Antiphospholipid Antibodies. Kidney Int Rep 2023; 8:754-763. [PMID: 37069974 PMCID: PMC10105059 DOI: 10.1016/j.ekir.2023.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Introduction Significant heterogeneity still exists in the nomenclature of renal involvement in antiphospholipid syndrome (APS). Methods We applied a hierarchical cluster analysis to determine subgroups of patients according to clinical, laboratory, and renal histology characteristics in a cohort of subjects with confirmed antiphospholipid antibodies (aPL) positivity and biopsy proven aPL-related renal injuries. Kidney outcomes were then assessed at 12 months. Results A total of 123 aPL-positive patients were included in the study (101 [82%] female, 109 [88.6%] with systemic lupus erythematosus [SLE], 14 (11.4%) with primary APS [PAPS]). Three clusters were identified. Twenty-three patients (18.7%) were included in the first cluster (cluster 1), characterized by a higher prevalence of glomerular capillary and arteriolar thrombi and fragmented red blood cells in the subendothelial space. Cluster 2 included 33 patients (26.8%) and showed a higher prevalence of fibromyointimal proliferative lesions as seen in hyperplastic vasculopathy. Cluster 3 was the largest (67 patients, mainly with SLE) and was characterized by higher prevalence of subendothelial edema, of both glomerular capillaries and arterioles. Conclusion Three different clusters of patients with aPL and renal injuries emerged from our study as follows: the first, with the worst renal prognosis, was associated with features of thrombotic microangiopathy (TMA), thrombosis, triple aPL positivity and higher adjusted Global APS Score (aGAPSS) values; the second, characterized by hyperplastic vasculopathy with an intermediate prognosis, was seen more frequently in patients with cerebrovascular manifestations; and the third, more benign in terms of outcomes and with no overt association with thrombotic features, was characterized by endothelial swelling in concomitant lupus nephritis (LN).
Collapse
Affiliation(s)
- Savino Sciascia
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| | - Jinoos Yazdany
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, California, USA
| | - Gabriella Moroni
- Department of Biomedical Sciences, IRCCS Humanitas Research Hospital, Rozzano, Lombardia, Italy
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Surya V. Seshan
- Department of Pathology and Laboratory Medicine, Weil Cornell Medicine, New York, New York, USA
| | | | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria J. Cuadrado
- Division of Pathology, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Massimo Radin
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| | - Irene Cecchi
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| | - Emanuele De Simone
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| | - Antonella Barreca
- Division of Pathology, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Leonardo Caroti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Samantha Innocenti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Roberta Fenoglio
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| | - Dario Roccatello
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases, Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital, Turin, Italy
| |
Collapse
|
46
|
Abstract
Antiphospholipid syndrome (APS) is a thrombo-inflammatory disease propelled by circulating autoantibodies that recognize cell surface phospholipids and phospholipid binding proteins. The result is an increased risk of thrombotic events, pregnancy morbidity, and various other autoimmune and inflammatory complications. Although antiphospholipid syndrome was first recognized in patients with lupus, the stand alone presentation of antiphospholipid syndrome is at least equally common. Overall, the diagnosis appears to affect at least one in 2000 people. Studies of antiphospholipid syndrome pathogenesis have long focused on logical candidates such as coagulation factors, endothelial cells, and platelets. Recent work has shed light on additional potential therapeutic targets within the innate immune system, including the complement system and neutrophil extracellular traps. Vitamin K antagonists remain the mainstay of treatment for most patients with thrombotic antiphospholipid syndrome and, based on current data, appear superior to the more targeted direct oral anticoagulants. The potential role of immunomodulatory treatments in antiphospholipid syndrome management is receiving increased attention. As for many systemic autoimmune diseases, the most important future direction is to more precisely identify mechanistic drivers of disease heterogeneity in pursuit of unlocking personalized and proactive treatments for patients.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - D Ware Branch
- James R. and Jo Scott Research Chair, Department of Obstetrics and Gynecology, University of Utah Health and Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Thomas L Ortel
- Division of Hematology, Departments of Medicine and Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
47
|
Capozzi A, Manganelli V, Riitano G, Caissutti D, Longo A, Garofalo T, Sorice M, Misasi R. Advances in the Pathophysiology of Thrombosis in Antiphospholipid Syndrome: Molecular Mechanisms and Signaling through Lipid Rafts. J Clin Med 2023; 12:jcm12030891. [PMID: 36769539 PMCID: PMC9917860 DOI: 10.3390/jcm12030891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
The pathological features of antiphospholipid syndrome (APS) are related to the activity of circulating antiphospholipid antibodies (aPLs) associated with vascular thrombosis and obstetric complications. Indeed, aPLs are not only disease markers, but also play a determining pathogenetic role in APS and exert their effects through the activation of cells and coagulation factors and inflammatory mediators for the materialization of the thromboinflammatory pathogenetic mechanism. Cellular activation in APS necessarily involves the interaction of aPLs with target receptors on the cell membrane, capable of triggering the signal transduction pathway(s). This interaction occurs at specific microdomains of the cell plasma membrane called lipid rafts. In this review, we focus on the key role of lipid rafts as signaling platforms in the pathogenesis of APS, and propose this pathogenetic step as a strategic target of new therapies in order to improve classical anti-thrombotic approaches with "new" immunomodulatory drugs.
Collapse
|
48
|
Furmańczyk-Zawiska A, Bułło-Piontecka B, Komorniczak M, Dębska-Ślizień A, Augustyniak-Bartosik H, Durlik M. Antiphospholipid Syndrome in Renal Allograft Recipients-A Long-Term Multicenter Analysis. J Clin Med 2023; 12:jcm12020667. [PMID: 36675596 PMCID: PMC9864673 DOI: 10.3390/jcm12020667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/03/2023] [Accepted: 01/07/2023] [Indexed: 01/18/2023] Open
Abstract
Antiphospholipid syndrome (APS) is a devastating autoimmune disease and in renal transplant recipients may result in allograft thrombosis or in extra-renal manifestation, mostly venous thromboembolism. There are many non- and immune risk factors affecting renal allograft in recipients with APS. However, renal allograft outcome in recipients with APS without APS nephropathy remains unknown. Aim: The aim of the study was to assess renal allograft function and survival in recipients with APS. Methods: Retrospective, multicenter study included 19 adult renal recipients with definite APS (primary or lupus-related) from three Polish transplant centers. Renal allograft function was assessed using serum creatinine concentration (SCr1) at 3rd month post-transplant and at the end of the observation (SCr2) and glomerular filtration rate (GFR) was estimated based on modification of diet in renal disease (MDRD) formula. General linear model was used to assess 12 month GFR change over time. Kaplan-Meier curves and restricted mean survival time were used for allograft survival. Matched control group consisted of 21 stable renal recipients without history of thrombosis and without anticoagulation/antiplatelet treatment. Results: The study group differs in induction therapy (p = 0.019), high-urgency procedure (p = 0.04), proteinuria (p = 0.0058), primary disease (lupus) (p < 0.0001), re-transplantation in primary APS (p = 0.0046) and shorter time since engraftment to SCr2 (p = 0.016). Primary APS was more often diagnosed post-transplant (p = 0.0005). Allograft biopsy revealed thrombotic microangiopathy (TMA) with acute rejection (AR) or isolated AR vs AR or chronic rejection in controls but did not reach significance (p = 0.054). Renal allograft function was inferior in the study group but did not reach significance: mean SCr2 (mg/dL) was 2.18 ± 1.41 and 1.5 ± 0.68 in controls, respectively, p = 0.27; mean GFR2 (ml/min/1.73m2) was 39.9 ± 20.83 and 51.23 ± 19.03, respectively, p = 0.102. Renal allograft duration was inferior in patients with APS and was (in years) 11.22 ± 1.44 vs. 14.36 ± 0.42, respectively, p = 0.037, in patients with primary APS (p = 0.021), in patients with APS diagnosed post-transplant (p = 0.012) but not in lupus-related APS (p = ns). Fifteen year renal allograft survival was inferior in APS vs. controls (73,86% vs. 90.48%, respectively, p = 0.049). Conclusions: Recipients with APS are at higher risk for allograft loss due to immune and non-immune causes. Renal allograft survival was inferior in recipients with APS and renal function remains impaired but stable.
Collapse
Affiliation(s)
- Agnieszka Furmańczyk-Zawiska
- Department of Transplant Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
- Correspondence:
| | - Barbara Bułło-Piontecka
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Dębinki 7, 80-952 Gdańsk, Poland
| | - Michał Komorniczak
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Dębinki 7, 80-952 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Dębinki 7, 80-952 Gdańsk, Poland
| | - Hanna Augustyniak-Bartosik
- Department of Nephrology and Transplant Medicine, Medical University of Wrocław, Borowska 213, 50-556 Wrocław, Poland
| | - Magdalena Durlik
- Department of Transplant Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| |
Collapse
|
49
|
Targeting thromboinflammation in antiphospholipid syndrome. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2022; 21:744-757. [PMID: 36696191 DOI: 10.1016/j.jtha.2022.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/26/2023]
Abstract
Antiphospholipid syndrome (APS) is a systemic autoimmune disease, where persistent presence of antiphospholipid antibodies (aPL) leads to thrombotic and obstetric complications. APS is a paradigmatic thromboinflammatory disease. Thromboinflammation is a pathophysiological mechanism coupling inflammation and thrombosis, which contributes to the pathophysiology of cardiovascular disease. APS can serve as a model to unravel mechanisms of thromboinflammation and the relationship between innate immune cells and thrombosis. Monocytes are activated by aPL into a proinflammatory and procoagulant phenotype, producing proinflammatory cytokines such as tumor necrosis factor α, interleukin 6, as well as tissue factor. Important cellular signaling pathways involved are the NF-κB-pathway, mammalian target of rapamycin (mTOR) signaling, and the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome. All of these may serve as future therapeutic targets. Neutrophils produce neutrophil extracellular traps in response to aPL, and this leads to thrombosis. Thrombosis in APS also stems from increased interaction of neutrophils with endothelial cells through P-selectin glycoprotein ligand-1. NETosis can be targeted not only with several experimental therapeutics, such as DNase, but also through the redirection of current therapies such as defibrotide and the antiplatelet agent dipyridamole. Activation of platelets by aPL leads to a procoagulant phenotype. Platelet-leukocyte interactions are increased, possibly mediated by increased levels of soluble P-selectin and soluble CD40-ligand. Platelet-directed future treatment options involve the inhibition of several platelet receptors activated by aPL, as well as mTOR inhibition. This review discusses mechanisms underlying thromboinflammation in APS that present targetable therapeutic options, some of which may be generalizable to other thromboinflammatory diseases.
Collapse
|
50
|
Becker YLC, Duvvuri B, Fortin PR, Lood C, Boilard E. The role of mitochondria in rheumatic diseases. Nat Rev Rheumatol 2022; 18:621-640. [PMID: 36175664 DOI: 10.1038/s41584-022-00834-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
The mitochondrion is an intracellular organelle thought to originate from endosymbiosis between an ancestral eukaryotic cell and an α-proteobacterium. Mitochondria are the powerhouses of the cell, and can control several important processes within the cell, such as cell death. Conversely, dysregulation of mitochondria possibly contributes to the pathophysiology of several autoimmune diseases. Defects in mitochondria can be caused by mutations in the mitochondrial genome or by chronic exposure to pro-inflammatory cytokines, including type I interferons. Following the release of intact mitochondria or mitochondrial components into the cytosol or the extracellular space, the bacteria-like molecular motifs of mitochondria can elicit pro-inflammatory responses by the innate immune system. Moreover, antibodies can target mitochondria in autoimmune diseases, suggesting an interplay between the adaptive immune system and mitochondria. In this Review, we discuss the roles of mitochondria in rheumatic diseases such as systemic lupus erythematosus, antiphospholipid syndrome and rheumatoid arthritis. An understanding of the different contributions of mitochondria to distinct rheumatic diseases or manifestations could permit the development of novel therapeutic strategies and the use of mitochondria-derived biomarkers to inform pathogenesis.
Collapse
Affiliation(s)
- Yann L C Becker
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Paul R Fortin
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Eric Boilard
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada.
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada.
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|