1
|
Li X, Xia J, Hu J, Chen Q, Li Y, Yin M, Zou H, Zhou W, Zhang P. Functional Brain Network Alterations in Patients With Systemic Lupus Erythematosus With Different Cognitive Function States: A Graph Theory Analysis Study. J Comput Assist Tomogr 2024; 48:283-291. [PMID: 37757812 DOI: 10.1097/rct.0000000000001546] [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: 09/29/2023]
Abstract
OBJECTIVE The study aimed to investigate the characteristics of brain functional network disruption in patients with systemic lupus erythematosus (SLE) with different cognitive function states by using graph theory analysis and to explore their relationship with clinical data and neuropsychiatric scales. METHODS Resting-state functional magnetic resonance imaging data were collected from 38 female SLE patients and 44 healthy controls. Based on Montreal Cognitive Assessment (MoCA) scores, SLE patients were divided into a high MoCA group (MoCA-H; MoCA score, ≥26) and a low MoCA group (MoCA-L; MoCA score, <26). The matrix of resting-state functional brain networks of subjects in the 3 groups was constructed by using the graph theory approach. The topological properties of the functional brain networks, including global and local metrics, in the 3 groups were calculated. The differences in the topological properties of networks between the 3 groups were compared. In addition, Spearman correlation analysis was used to explore the correlation between altered topological properties of brain networks and clinical indicators, as well as neuropsychiatric scales in SLE patients in the MoCA-L group. RESULTS At the global level, in the sparsity threshold range of 0.10 to 0.34, the values of small-world properties were greater than 1 in all 3 groups, indicating that functional brain networks of both 3 groups had small-world properties. There were statistically significant differences in the characteristic path length, global, and local efficiency between 3 groups ( F = 3.825, P = 0.0260; F = 3.722, P = 0.0285; and F = 3.457, P = 0.0364, respectively). Systemic lupus erythematosus patients in the MoCA-L group showed increased characteristic path length ( t = 2.816, P = 0.00651), decreased global ( t = -2.729, P = 0.00826), and local efficiency ( t = -2.623, P = 0.0109) compared with healthy controls. No statistically significant differences in local metrics were found between the MoCA-H group and the healthy control, MoCA-L groups. At the local level, there was statistically significant difference in the node efficiency among the 3 groups ( P < 0.05 after Bonferroni correction). Compared with healthy controls, SLE patients in the MoCA-L group showed decreased node efficiency in left anterior cingulate paracingulate gyrus, bilateral putamen, bilateral pallidum, and left Heschl gyrus. No statistically significant differences in the local metrics were found between the MoCA-H, MoCA-L, and healthy control groups. Correlation analysis in SLE patients in the MoCA-L group showed that the characteristic path length was positively correlated with C4 levels ( r = 0.587, P = 0.007), the global and local efficiencies were negatively correlated with C4 levels ( r = -0.599, P = 0.005; r = -0.599, P = 0.005, respectively), and the node efficiency in the bilateral putamen was negatively correlated with C4 levels ( r = -0.611, P = 0.004; r = -0.570, P = 0.009). The node efficiency in the left pallidum was negatively correlated with disease duration ( r = -0.480, P = 0.032). The node efficiency in the left Heschl gyrus was negatively correlated with IgM levels ( r = -0.478, P = 0.033). No correlation was noted between other network metrics, clinical indicators, and neuropsychological scales. CONCLUSIONS The topological properties of functional brain networks were disrupted in SLE patients with low MoCA scores, suggesting that altered topological properties of the brain networks were associated with cognitive function in SLE patients. Correlation between altered topological properties of the brain networks and clinical indicators was noted in SLE patients with low MoCA scores, suggesting that altered topological properties of brain networks in SLE patients may have clinical significance as imaging markers for monitoring disease changes in patients with SLE.
Collapse
Affiliation(s)
- Xiaolu Li
- From the Department of Radiology, Graduate School of Dalian Medical University, Dalian
| | | | | | | | | | | | - Hongmei Zou
- Department of Rheumatology and Immunology, Taizhou People's Hospital, Jiangsu, China
| | - Wensu Zhou
- From the Department of Radiology, Graduate School of Dalian Medical University, Dalian
| | - Peng Zhang
- From the Department of Radiology, Graduate School of Dalian Medical University, Dalian
| |
Collapse
|
2
|
Gelbenegger G, Berentsen S, Jilma B. Monoclonal antibodies for treatment of cold agglutinin disease. Expert Opin Biol Ther 2023; 23:395-406. [PMID: 37128907 DOI: 10.1080/14712598.2023.2209265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
INTRODUCTION Cold agglutinin disease (CAD) is a difficult-to-treat autoimmune hemolytic anemia and B cell lymphoproliferative disorder associated with fatigue, acrocyanosis and a risk of thromboembolic events. Cold-induced binding of autoantibodies agglutinates red blood cells and triggers the classical complement pathway, leading to predominantly extravascular hemolysis. AREAS COVERED This review summarizes clinical and experimental antibody-based treatments for CAD and analyzes the risks and benefits of B cell and complement directed therapies, and discusses potential future treatments for CAD. EXPERT OPINION Conventional treatment of CAD includes a B cell targeted treatment approach with rituximab, yielding only limited treatment success. Addition of a cytotoxic agent (e.g. bendamustine) increases efficacy but this is accompanied by an increased risk of neutropenia and infection. Novel complement-directed therapies have emerged and were shown to have a good efficacy against hemolysis and safety profile but are expensive and unable to address circulatory symptoms. Complement inhibition with sutimlimab may be used as a bridging strategy until B cell directed therapy with rituximab takes effect or continued indefinitely if needed. Future antibody-based treatment approaches for CAD involve the further development of complement-directed antibodies, combination of rituximab and bortezomib, and daratumumab. Non-antibody based prospective treatments may include the use of Bruton tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, Haugesund, Norway
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Berentsen S. Sutimlimab for the Treatment of Cold Agglutinin Disease. Hemasphere 2023; 7:e879. [PMID: 37153870 PMCID: PMC10155901 DOI: 10.1097/hs9.0000000000000879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Cold agglutinin disease (CAD) is a rare autoimmune hemolytic anemia and a bone marrow clonal lymphoproliferative disorder. Hemolysis in CAD is complement-dependent and mediated by the classical activation pathway. Patients also frequently suffer from fatigue and cold-induced circulatory symptoms. Although not all patients need treatment, the symptom burden has previously been underestimated. Effective therapies target the clonal lymphoproliferation or the complement activation. Sutimlimab, a humanized monoclonal IgG4 antibody that binds and inactivates complement protein C1s, is the most extensively investigated complement inhibitor for the treatment of CAD. This review addresses the preclinical studies of sutimlimab and the studies of pharmacokinetics and pharmacodynamics. We then describe and discuss the prospective clinical trials that established sutimlimab as a rapidly acting, highly efficacious, and low-toxic therapeutic agent. This complement inhibitor does not improve the cold-induced circulatory symptoms, which are not complement-mediated. Sutimlimab is approved for the treatment of CAD in the US, Japan, and the European Union. A tentative therapeutic algorithm is presented. The choice of therapy for CAD should be based on an individual assessment, and patients requiring therapy should be considered for inclusion in clinical trials.
Collapse
Affiliation(s)
- Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Hospital Trust, Haugesund, Norway
| |
Collapse
|
4
|
Berentsen S, Barcellini W, D'Sa S, Jilma B. Sutimlimab for treatment of cold agglutinin disease: why, how and for whom? Immunotherapy 2022; 14:1191-1204. [PMID: 35946351 DOI: 10.2217/imt-2022-0085] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Therapies for cold agglutinin disease have been directed at the pathogenic B-cell clone. Sutimlimab, a monoclonal antibody that targets C1s, is the first complement inhibitor to be extensively studied in cold agglutinin disease. Sutimlimab selectively blocks the classical activation pathway and leaves the alternative and lectin pathways intact. Trials have documented high response rates with rapid improvement in hemolysis, hemoglobin levels and fatigue scores and low toxicity. Sutimlimab was recently approved in the USA. This drug appears to be particularly useful in severely anemic patients who require a rapid response, in acute exacerbations that do not resolve spontaneously and in patients in whom chemoimmunotherapy is contraindicated or has failed. The choice of therapy in cold agglutinin disease should be individualized.
Collapse
Affiliation(s)
- Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Hospital Trust, Haugesund, Norway
| | - Wilma Barcellini
- Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Shirley D'Sa
- University College London Hospitals Centre for Waldenström and Associated Conditions, University College London Hospitals National Health Service Foundation Trust, London, UK
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Berentsen S, Tjønnfjord GE. Current treatment options in cold agglutinin disease: B-cell directed or complement directed therapy? Transfus Med Rev 2022; 36:181-187. [DOI: 10.1016/j.tmrv.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 10/15/2022]
|
6
|
Padron GT, Hernandez-Trujillo VP. Autoimmunity in Primary Immunodeficiencies (PID). Clin Rev Allergy Immunol 2022:10.1007/s12016-022-08942-0. [PMID: 35648371 DOI: 10.1007/s12016-022-08942-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 11/25/2022]
Abstract
Primary immunodeficiency (PID) may impact any component of the immune system. The number of PID and immune dysregulation disorders is growing steadily with advancing genetic detection methods. These expansive recognition methods have changed the way we characterize PID. While PID were once characterized by their susceptibility to infection, the increase in genetic analysis has elucidated the intertwined relationship between PID and non-infectious manifestations including autoimmunity. The defects permitting opportunistic infections to take hold may also lead the way to the development of autoimmune disease. In some cases, it is the non-infectious complications that may be the presenting sign of PID autoimmune diseases, such as autoimmune cytopenia, enteropathy, endocrinopathies, and arthritis among others, have been reported in PID. While autoimmunity may occur with any PID, this review will look at certain immunodeficiencies most often associated with autoimmunity, as well as their diagnosis and management strategies.
Collapse
Affiliation(s)
- Grace T Padron
- Nicklaus Children's Hospital, Miami, FL, USA.
- Allergy and Immunology Care Center of South Florida, Miami Lakes, FL, USA.
| | - Vivian P Hernandez-Trujillo
- Nicklaus Children's Hospital, Miami, FL, USA
- Allergy and Immunology Care Center of South Florida, Miami Lakes, FL, USA
| |
Collapse
|
7
|
Ezeh N, Ardalan K, Buhr KA, Nguyen C, Al Ahmed O, Ardoin SP, Barton V, Bell S, Brandling-Bennett H, Castelo-Soccio L, Chiu YE, Chong BF, Lara-Corrales I, Cintosun A, Curran ML, Diaz LZ, Elman SA, Faith EF, Garcia-Romero MT, Grossman-Kranseler J, Hogeling M, Hudson AD, Hunt RD, Ibler EM, Marques MC, Monir RL, Oza V, Paller AS, Putterman E, Rodriguez-Salgado P, Schoch JJ, Truong A, Wang J, Lee LW, Vleugels RA, Klein-Gitelman MS, von Scheven E, Werth VP, Arkin LM. Cross-Sectional Characteristics of Pediatric-Onset Discoid Lupus Erythematosus: Results of a Multicenter, Retrospective Cohort Study. J Am Acad Dermatol 2022; 87:559-566. [PMID: 35487332 PMCID: PMC10082546 DOI: 10.1016/j.jaad.2022.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 11/15/2022]
Abstract
• Discoid lupus may be associated with SLE. In this study, most children with discoid lupus did not have systemic disease. Concurrent SLE was highest in female adolescents (>10 years of age) with generalized discoid lupus, who had clinically aggressive disease. • Discoid lupus in adolescence should prompt thorough evaluation for SLE.
Collapse
Affiliation(s)
- N Ezeh
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - K Ardalan
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, Departments of Pediatrics and Medical Social Sciences, Northwestern University Feinberg School of Medicine/Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - K A Buhr
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - C Nguyen
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - O Al Ahmed
- Department of Pediatrics, Nationwide Children's Hospital/Ohio State University, Columbus OH
| | - S P Ardoin
- Department of Pediatrics, Nationwide Children's Hospital/Ohio State University, Columbus OH
| | - V Barton
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC
| | - S Bell
- Department of Pediatrics, Division of Rheumatology, University of California at San Francisco, San Francisco, CA
| | - H Brandling-Bennett
- Division of Dermatology; Department of Pediatrics, Seattle Children's Hospital/University of Washington School of Medicine, Seattle, WA
| | - L Castelo-Soccio
- Children's Hospital of Philadelphia, Department of Pediatrics, Section of Dermatology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Y E Chiu
- Departments of Dermatology (Section of Pediatric Dermatology) and Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - B F Chong
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX; Department of Pediatrics, Division of Allergy/Immunology/Rheumatology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - I Lara-Corrales
- Department of Pediatrics, Section of Dermatology, The Hospital for Sick Children/University of Toronto, Toronto, ON
| | - A Cintosun
- Department of Pediatrics, Section of Dermatology, The Hospital for Sick Children/University of Toronto, Toronto, ON
| | - M L Curran
- Department of Pediatrics, Section of Rheumatology, Children's Hospital Colorado/University of Colorado School of Medicine, Aurora, CO
| | - L Z Diaz
- Department of Pediatrics, Division of Dermatology, Dell Medical School/Dell Children's Hospital, Austin, TX
| | - S A Elman
- Department of Dermatology, Boston's Children Hospital/Harvard Medical School, Boston, MA
| | - E Fernandez Faith
- Department of Pediatrics, Nationwide Children's Hospital/Ohio State University, Columbus OH
| | - M T Garcia-Romero
- Department of Dermatology, National Institute for Pediatrics, Mexico City, MX
| | - J Grossman-Kranseler
- Division of Dermatology; Department of Pediatrics, Seattle Children's Hospital/University of Washington School of Medicine, Seattle, WA
| | - M Hogeling
- Department of Dermatology, University of California at Los Angeles, Los Angeles, CA
| | - A D Hudson
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX
| | - R D Hunt
- Department of Dermatology and Pediatrics, Texas Children's Hospital, Houston, TX
| | - E M Ibler
- Departments of Dermatology (Section of Pediatric Dermatology) and Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - M C Marques
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - R L Monir
- Department of Dermatology, University of Florida School of Medicine, Gainesville, FL
| | - V Oza
- Departments of Pediatrics and Dermatology, New York University Grossman School of Medicine, New York, NY
| | - A S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine/Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - E Putterman
- Children's Hospital of Philadelphia, Department of Pediatrics, Section of Dermatology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - P Rodriguez-Salgado
- Department of Dermatology, National Institute for Pediatrics, Mexico City, MX
| | - J J Schoch
- Department of Dermatology, University of Florida School of Medicine, Gainesville, FL
| | - A Truong
- Department of Dermatology, University of California at Los Angeles, Los Angeles, CA
| | - J Wang
- Departments of Pediatrics and Dermatology, New York University Grossman School of Medicine, New York, NY
| | - L Wine Lee
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC
| | - R A Vleugels
- Department of Dermatology, Boston's Children Hospital/Harvard Medical School, Boston, MA
| | - M S Klein-Gitelman
- Department of Pediatrics, Northwestern University Feinberg School of Medicine/Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - E von Scheven
- Department of Pediatrics, Division of Rheumatology, University of California at San Francisco, San Francisco, CA
| | - V P Werth
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania and Corporal Michael J. Crescenz VAMC, Philadelphia, PA
| | - L M Arkin
- Departments of Dermatology and Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
8
|
Parisis D, Bernier C, Chasset F, Arnaud L. Impact of tobacco smoking upon disease risk, activity and therapeutic response in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2019; 18:102393. [PMID: 31520802 DOI: 10.1016/j.autrev.2019.102393] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/19/2019] [Indexed: 01/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a complex disease with variable presentations, course and prognosis. Published studies present conflicting data regarding the impact of cigarette smoking on SLE risk, disease activity, clinical manifestations and treatment response. We performed a comprehensive literature search using Medline, EMBASE and the Cochrane Collaboration database, and hand searches of relevant bibliographies. All original studies investigating the relationship between smoking and SLE were included in TABALUP. Two investigators systematically extracted data from the relevant studies. When possible, meta-analyses were performed. The meta-analysis of 9 case-controls studies show an increased risk of SLE in current-smokers compared to never-smokers (OR: 1.49 [95%CI: 1.06-2.08]), while former-smokers were not at increased risk of SLE. Data on passive smoking remains scarce and controversial. Pooled analysis studies did not find an over-risk of anti-dsDNA, anti-Sm or anti-SSA positivity according to smoking status. Tobacco smoking significantly reduced the therapeutic effectiveness of hydroxychloroquine in cutaneous lesions (pooled OR 0.53; 95%CI: 0.305-0.927) and belimumab in systemic manifestations (HR 0.10; 95% CI 0.02-0.43). In addition to its usual adverse effects, cigarette smoking is a risk factor of SLE and negatively influences the course of the disease and its treatment. We believe that smoking cessation should be one of the main target of physicians treating SLE patients.
Collapse
Affiliation(s)
- Dorian Parisis
- Service de rhumatologie et de médecine physique, CUB Erasme (ULB), Brussels, Belgium
| | - Charlie Bernier
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Centre National de Référence des Maladies Systémiques Rares Est Sud-Ouest (RESO), Université de Strasbourg, F-67000 Strasbourg, France
| | - François Chasset
- Sorbonne Université, Faculté de médecine sorbonne université, ap-hp, Service de dermatologie et allergologie, Hôpital tenon, F-75020 Paris, france
| | - Laurent Arnaud
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Centre National de Référence des Maladies Systémiques Rares Est Sud-Ouest (RESO), Université de Strasbourg, F-67000 Strasbourg, France.
| |
Collapse
|
9
|
Berentsen S, Hill A, Hill QA, Tvedt THA, Michel M. Novel insights into the treatment of complement-mediated hemolytic anemias. Ther Adv Hematol 2019; 10:2040620719873321. [PMID: 31523413 PMCID: PMC6734604 DOI: 10.1177/2040620719873321] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Complement-mediated hemolytic anemias can either be caused by deficiencies in regulatory complement components or by autoimmune pathogenesis that triggers inappropriate complement activation. In paroxysmal nocturnal hemoglobinuria (PNH) hemolysis is entirely complement-driven. Hemolysis is also thought to be complement-dependent in cold agglutinin disease (CAD) and in paroxysmal cold hemoglobinuria (PCH), whereas warm antibody autoimmune hemolytic anemia (wAIHA) is a partially complement-mediated disorder, depending on the subtype of wAIHA and the extent of complement activation. The pathophysiology, clinical presentation, and current therapies for these diseases are reviewed in this article. Novel, complement-directed therapies are being rapidly developed. Therapeutic terminal complement inhibition using eculizumab has revolutionized the therapy and prognosis in PNH but has proved less efficacious in CAD. Upstream complement modulation is currently being investigated and appears to be a highly promising therapy, and two such agents have entered phase II and III trials. Of these, the anti-C1s monoclonal antibody sutimlimab has shown favorable activity in CAD, while the anti-C3 cyclic peptide pegcetacoplan appears to be promising in PNH as well as CAD, and may also have a therapeutic potential in wAIHA.
Collapse
Affiliation(s)
- Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, P.O. Box 2170, Haugesund, 5504, Norway
| | - Anita Hill
- Department of Haematology, Leeds Teaching Hospitals, Leeds, UK
| | - Quentin A Hill
- Department of Haematology, Leeds Teaching Hospitals, Leeds, UK
| | | | - Marc Michel
- Department of Medicine, Henri Mondor Hospital, Université Paris-Est, Assistance Publique Hôpitaux de Paris Creteil, Creteil, France
| |
Collapse
|
10
|
Durcan L, O'Dwyer T, Petri M. Management strategies and future directions for systemic lupus erythematosus in adults. Lancet 2019; 393:2332-2343. [PMID: 31180030 DOI: 10.1016/s0140-6736(19)30237-5] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2019] [Accepted: 01/23/2019] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterised by the loss of self-tolerance and formation of nuclear autoantigens and immune complexes resulting in inflammation of multiple organs. The clinical presentation of SLE is heterogeneous, can involve one or more organs, including the skin, kidneys, joints, and nervous system, and take a chronic or relapsing and remitting disease course. SLE is most common in women and in those of non-white ethnicity. Because of the multitude of presentations, manifestations, and serological abnormalities in patients with SLE, diagnosis can be challenging. Therapeutic approaches predominantly involve immunomodulation and immunosuppression and are targeted to the specific organ manifestation, with the aim of achieving low disease activity. Despite many treatment advances and improved diagnostics, SLE continues to cause substantial morbidity and premature mortality. Current management strategies, although helpful, are limited by high failure rates and toxicity. An overreliance on corticosteroid therapy contributes to much of the long-term organ damage. In this Seminar, we outline the classification criteria for SLE, current treatment strategies and medications, the evidence supporting their use, and explore potential future therapies.
Collapse
Affiliation(s)
- Laura Durcan
- Department of Rheumatology, Beaumont Hospital, Dublin, Ireland; Department of Medicine, The Royal College of Surgeons of Ireland, Dublin, Ireland.
| | - Tom O'Dwyer
- School of Physiotherapy, Trinity College, Dublin, Ireland
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| |
Collapse
|
11
|
Berentsen S, Röth A, Randen U, Jilma B, Tjønnfjord GE. Cold agglutinin disease: current challenges and future prospects. J Blood Med 2019; 10:93-103. [PMID: 31114413 PMCID: PMC6497508 DOI: 10.2147/jbm.s177621] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
Cold agglutinin disease (CAD) is a complement-dependent, classical pathway-mediated immune hemolytic disease, accounting for 15–25% of autoimmune hemolytic anemia, and at the same time, a distinct clonal B-cell lymphoproliferative disorder of the bone marrow. The disease burden is often high, but not all patients require pharmacological treatment. Several therapies directed at the pathogenic B-cells are now available. Rituximab plus bendamustine or rituximab monotherapy should be considered first-line treatment, depending on individual patient characteristics. Novel treatment options that target the classical complement pathway are under development and appear very promising, and the C1s inhibitor sutimlimab is currently being investigated in two clinical Phase II and III trials. These achievements have raised new challenges and further prospects, which are discussed. Patients with CAD requiring therapy should be considered for clinical trials.
Collapse
Affiliation(s)
- Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, Haugesund, Norway
| | - Alexander Röth
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulla Randen
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Geir E Tjønnfjord
- Department of Haematology, Oslo University Hospital, Oslo, Norway.,KG Jebsen's Center for B-cell Malignancies, University of Oslo, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Amaya-Uribe L, Rojas M, Azizi G, Anaya JM, Gershwin ME. Primary immunodeficiency and autoimmunity: A comprehensive review. J Autoimmun 2019; 99:52-72. [PMID: 30795880 DOI: 10.1016/j.jaut.2019.01.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023]
Abstract
The primary immunodeficiency diseases (PIDs) include many genetic disorders that affect different components of the innate and adaptive responses. The number of distinct genetic PIDs has increased exponentially with improved methods of detection and advanced laboratory methodology. Patients with PIDs have an increased susceptibility to infectious diseases and non-infectious complications including allergies, malignancies and autoimmune diseases (ADs), the latter being the first manifestation of PIDs in several cases. There are two types of PIDS. Monogenic immunodeficiencies due to mutations in genes involved in immunological tolerance that increase the predisposition to develop autoimmunity including polyautoimmunity, and polygenic immunodeficiencies characterized by a heterogeneous clinical presentation that can be explained by a complex pathophysiology and which may have a multifactorial etiology. The high prevalence of ADs in PIDs demonstrates the intricate relationships between the mechanisms of these two conditions. Defects in central and peripheral tolerance, including mutations in AIRE and T regulatory cells respectively, are thought to be crucial in the development of ADs in these patients. In fact, pathology that leads to PID often also impacts the Treg/Th17 balance that may ease the appearance of a proinflammatory environment, increasing the odds for the development of autoimmunity. Furthermore, the influence of chronic and recurrent infections through molecular mimicry, bystander activation and super antigens activation are supposed to be pivotal for the development of autoimmunity. These multiple mechanisms are associated with diverse clinical subphenotypes that hinders an accurate diagnosis in clinical settings, and in some cases, may delay the selection of suitable pharmacological therapies. Herein, a comprehensively appraisal of the common mechanisms among these conditions, together with clinical pearls for treatment and diagnosis is presented.
Collapse
Affiliation(s)
- Laura Amaya-Uribe
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Doctoral Program in Biomedical Sciences, Universidad Del Rosario, Bogota, Colombia
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA.
| |
Collapse
|
13
|
Berentsen S. Complement Activation and Inhibition in Autoimmune Hemolytic Anemia: Focus on Cold Agglutinin Disease. Semin Hematol 2018; 55:141-149. [PMID: 30032751 DOI: 10.1053/j.seminhematol.2018.04.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/03/2018] [Indexed: 12/25/2022]
Abstract
The classical complement pathway and, to some extent, the terminal pathway, are involved in the immune pathogenesis of autoimmune hemolytic anemia (AIHA). In primary cold agglutinin disease (CAD), secondary cold agglutinin syndrome and paroxysmal cold hemoglobinuria, the hemolytic process is entirely complement dependent. Complement activation also plays an important pathogenetic role in some warm-antibody AIHAs, especially when immunoglobulin M is involved. This review describes the complement-mediated hemolysis in AIHA with a major focus on CAD, in which activation of the classical pathway is essential and particularly relevant for complement-directed therapy. Several complement inhibitors are candidate therapeutic agents in CAD and other AIHAs, and some of these drugs seem very promising. The relevant in vitro findings, early clinical data and future perspectives are reviewed.
Collapse
Affiliation(s)
- Sigbjørn Berentsen
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna HF, Haugesund, Norway.
| |
Collapse
|
14
|
C1r/C1s deficiency is insufficient to induce murine systemic lupus erythematosus. Genes Immun 2018; 20:121-130. [PMID: 29550838 DOI: 10.1038/s41435-018-0020-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/14/2018] [Accepted: 01/18/2018] [Indexed: 11/08/2022]
Abstract
C1s deficiency is strongly associated with the development of human systemic lupus erythematosus (SLE); however, the mechanisms by which C1s deficiency contributes to the development of SLE have not yet been elucidated in detail. Using ICR-derived-glomerulonephritis (ICGN) mouse strain that develops SLE and very weakly expresses C1s in the liver, we investigated the protective roles of C1s against SLE. A genetic sequence analysis revealed complete deletion of the C1s1 gene, a mouse homolog of the human C1s gene, with partial deletion of the C1ra and C1rb genes in the ICGN strain. This deletion led to the absence of C1r/C1s and a low level of C1q in the circulation. In order to investigate whether the C1r/C1s deficiency induces SLE, we produced a congenic mouse strain by introducing the deletion region of ICGN into the C57BL/6 strain. Congenic mice exhibited no C1r/C1s and a low level of C1q in the circulation, but did not have any autoimmune defects. These results suggest that C1r/C1s deficiency is not sufficient to drive murine SLE and also that other predisposing genes exist in ICGN mice.
Collapse
|
15
|
Hu W, Niu G, Li H, Gao H, Kang R, Chen X, Lin L. The association between expression of IFIT1 in podocytes of MRL/lpr mice and the renal pathological changes it causes: An animal study. Oncotarget 2018; 7:76464-76470. [PMID: 27823966 PMCID: PMC5363523 DOI: 10.18632/oncotarget.13045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/28/2016] [Indexed: 11/29/2022] Open
Abstract
Renal damage is the major cause of SLE associated mortality, and IFIT1expression was elevated in SLE cases in accordance of previous studies. Therefore, we conducted an animal study to identify the role of IFIT1 expression in renal pathological changes.18 female MRL/lpr mice and same number of female BALB/c mice were enrolled in present study. Quantitative analysis of urine protein, Complement C3 and C4, and anti-ds DNA antibody were conducted. HE and PAS staining and TEM analysis were employed to observe the pathological changes in renal tissue. Significant elevation on urine protein and anti-dsDNA and reduction on Complement C3 and C4 were observed in MRL/lpr mice when comparing the controls in same age. Staining and TEM analysis observed several pathological changes in glomerulus among MRL/lpr mice, including cellular enlargement, basement membrane thickening, and increased cellularcasts. The linear regression analysis found the optical density of IFIT1 was inversely associated with F-actin, Nephrin, and Podocin, but not Synatopodin. In summary, IFIT1 expression is associated with podocytes damage, and capable of suppressing some proteins essential to glomerular filtration.
Collapse
Affiliation(s)
- Weiping Hu
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Guodong Niu
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Hongbo Li
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Hanyuan Gao
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Rudian Kang
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xiaoqing Chen
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Ling Lin
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
16
|
Al-Mayouf SM, AlSaleem A, AlMutairi N, AlSonbul A, Alzaid T, Alazami AM, Al-Mousa H. Monogenic interferonopathies: Phenotypic and genotypic findings of CANDLE syndrome and its overlap with C1q deficient SLE. Int J Rheum Dis 2017; 21:208-213. [PMID: 29115062 DOI: 10.1111/1756-185x.13228] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To report the clinical and genetic features of the first cases of chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome in an Arab population and to compare them with patients of C1q deficient systemic lupus erythematosus (SLE). MATERIALS AND METHODS This is a retrospective case series of patients with CANDLE syndrome and C1q deficient SLE seen at a single tertiary hospital. Medical records were reviewed for demographic data, clinical and laboratory features, histopathology and imaging findings, and response to therapeutic intervention. Descriptive data were summarized. RESULTS Three patients from unrelated families fulfilled the clinical manifestations of CANDLE syndrome. The disease onset was within the first 4 months of age. Two patients had uncommon features including uveitis, pulmonary involvement, aseptic meningitis and global delay. Skin biopsy showed heterogeneous findings. Genomic DNA screening was homozygous for mutation in PSMB8, (NM_004159.4:c.212C>T, p.T71M) in one patient and inconclusive for the other two patients. The comparison group was three patients with familial C1q deficient SLE from three unrelated families, who were born to consanguineous parents with at least one affected sibling. They presented with extensive mucocutaneous lesions, discoid rash and scarring alopecia. They required frequent admissions due to infections. CONCLUSION This is the first report of CANDLE syndrome in an Arab population; our patients had heterogeneous phenotypic and genetic features with overlap manifestations with C1q deficient SLE. Both are monogenic interferonopathies. However, C1q deficient SLE had more systemic inflammatory disease.
Collapse
Affiliation(s)
- Sulaiman M Al-Mayouf
- Department of Pediatric Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alhanouf AlSaleem
- Department of Pediatric Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nora AlMutairi
- Department of Pediatric Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdullah AlSonbul
- Department of Pediatric Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Tariq Alzaid
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hamoud Al-Mousa
- Department of Allergy and Immunology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Dunn AB, Dunlop AL, Hogue CJ, Miller A, Corwin EJ. The Microbiome and Complement Activation: A Mechanistic Model for Preterm Birth. Biol Res Nurs 2017; 19:295-307. [PMID: 28073296 DOI: 10.1177/1099800416687648] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Preterm birth (PTB, <37 completed weeks' gestation) is one of the leading obstetrical problems in the United States, affecting approximately one of every nine births. Even more concerning are the persistent racial disparities in PTB, with particularly high rates among African Americans. There are several recognized pathophysiologic pathways to PTB, including infection and/or exaggerated systemic or local inflammation. Intrauterine infection is a causal factor linked to PTB thought to result most commonly from inflammatory processes triggered by microbial invasion of bacteria ascending from the vaginal microbiome. Trials to treat various infections have shown limited efficacy in reducing PTB risk, suggesting that other complex mechanisms, including those associated with inflammation, may be involved in the relationship between microbes, infection, and PTB. The complement system, a key mediator of the inflammatory response, is an innate defense mechanism involved in both normal physiologic processes that occur during pregnancy implantation and processes that promote the elimination of pathogenic microbes. Recent research has demonstrated an association between this system and PTB. The purpose of this article is to present a mechanistic model of inflammation-associated PTB, which hypothesizes a relationship between the microbiome and dysregulation of the complement system. Exploring the relationships between the microbial environment and complement biomarkers may elucidate a potentially modifiable biological pathway to PTB.
Collapse
Affiliation(s)
- Alexis B Dunn
- 1 Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Anne L Dunlop
- 1 Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Carol J Hogue
- 2 Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Andrew Miller
- 3 School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Elizabeth J Corwin
- 1 Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| |
Collapse
|
18
|
A pathophysiology-based approach to the diagnosis and treatment of lupus nephritis. Kidney Int 2016; 90:493-501. [PMID: 27378475 DOI: 10.1016/j.kint.2016.05.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/09/2016] [Accepted: 05/10/2016] [Indexed: 12/31/2022]
Abstract
Lupus is no longer an unknown chameleon of medicine. Significant progress has been made on unraveling the pathogenesis of lupus and lupus nephritis, and how to treat the disease. Here we provide an update on the pathophysiology of lupus and its related kidney disease, consider areas of controversy in disease management, and discuss the unmet needs of lupus nephritis and how to address these needs. We focus on rethinking how innovative therapies for lupus nephritis should be evaluated and evolving strategies to more efficiently mitigate irreversible nephron loss in patients with lupus nephritis.
Collapse
|
19
|
The crossroads of autoimmunity and immunodeficiency: Lessons from polygenic traits and monogenic defects. J Allergy Clin Immunol 2016; 137:3-17. [DOI: 10.1016/j.jaci.2015.11.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/16/2023]
|
20
|
Early Complement Component Deficiency in a Single-Centre Cohort of Pediatric Onset Lupus. J Clin Immunol 2015; 35:777-85. [PMID: 26563161 DOI: 10.1007/s10875-015-0212-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/05/2015] [Indexed: 11/26/2022]
|
21
|
Regal JF, Gilbert JS, Burwick RM. The complement system and adverse pregnancy outcomes. Mol Immunol 2015; 67:56-70. [PMID: 25802092 DOI: 10.1016/j.molimm.2015.02.030] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/20/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023]
Abstract
Adverse pregnancy outcomes significantly contribute to morbidity and mortality for mother and child, with lifelong health consequences for both. The innate and adaptive immune system must be regulated to insure survival of the fetal allograft, and the complement system is no exception. An intact complement system optimizes placental development and function and is essential to maintain host defense and fetal survival. Complement regulation is apparent at the placental interface from early pregnancy with some degree of complement activation occurring normally throughout gestation. However, a number of pregnancy complications including early pregnancy loss, fetal growth restriction, hypertensive disorders of pregnancy and preterm birth are associated with excessive or misdirected complement activation, and are more frequent in women with inherited or acquired complement system disorders or complement gene mutations. Clinical studies employing complement biomarkers in plasma and urine implicate dysregulated complement activation in components of each of the adverse pregnancy outcomes. In addition, mechanistic studies in rat and mouse models of adverse pregnancy outcomes address the complement pathways or activation products of importance and allow critical analysis of the pathophysiology. Targeted complement therapeutics are already in use to control adverse pregnancy outcomes in select situations. A clearer understanding of the role of the complement system in both normal pregnancy and complicated or failed pregnancy will allow a rational approach to future therapeutic strategies for manipulating complement with the goal of mitigating adverse pregnancy outcomes, preserving host defense, and improving long term outcomes for both mother and child.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Jeffrey S Gilbert
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Richard M Burwick
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Oregon Health & Science University, Mail Code: L-458, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
22
|
Patzelt J, Verschoor A, Langer HF. Platelets and the complement cascade in atherosclerosis. Front Physiol 2015; 6:49. [PMID: 25784879 PMCID: PMC4345806 DOI: 10.3389/fphys.2015.00049] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/04/2015] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis and its late sequels are still the number one cause of death in western societies. Platelets are a driving force not only during the genesis of atherosclerosis, but especially in its late stages, as evidenced by complications such as arterial thrombosis, myocardial infarction, and ischemic stroke. Atherosclerosis is increasingly recognized as an inflammatory disease, influenced by various immune mechanisms. The complement system is part of our innate immune system, and its diverse roles in atherosclerosis have become evident over the past years. In this review we identify points of intersection between platelets and the complement system and discuss their relevance for atherosclerosis. Specifically, we will focus on roles for platelets in the onset as well as progression of the disease, a possible dual role for complement in the genesis and development of atherosclerosis, and review emerging literature revealing previously unrecognized cross-talk between platelets and the complement system and discuss its possible impact for atherosclerosis. Finally, we identify limitations of current research approaches and discuss perspectives of complement modulation in the control of the disease.
Collapse
Affiliation(s)
- Johannes Patzelt
- University Clinic for Cardiovascular Medicine, University of Tuebingen Tuebingen, Germany
| | - Admar Verschoor
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München Munich, Germany
| | - Harald F Langer
- University Clinic for Cardiovascular Medicine, University of Tuebingen Tuebingen, Germany ; Section for Cardioimmunology, Department of Cardiovascular Medicine, University of Tuebingen Tuebingen, Germany
| |
Collapse
|
23
|
Liphaus BL, Umetsu N, Jesus AA, Bando SY, Silva CA, Carneiro-Sampaio M. Molecular characterization of the complement C1q, C2 and C4 genes in Brazilian patients with juvenile systemic lupus erythematosus. Clinics (Sao Paulo) 2015; 70:220-7. [PMID: 26017655 PMCID: PMC4449463 DOI: 10.6061/clinics/2015(03)12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/05/2015] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To perform a molecular characterization of the C1q, C2 and C4 genes in patients with juvenile systemic lupus erythematosus. METHODS Patient 1 (P1) had undetectable C1q, patient 2 (P2) and patient 3 (P3) had decreased C2 and patient 4 (P4) had decreased C4 levels. All exons and non-coding regions of the C1q and C2 genes were sequenced. Mononuclear cells were cultured and stimulated with interferon gamma to evaluate C1q, C2 and C4 mRNA expression by quantitative real-time polymerase chain reaction. RESULTS C1q sequencing revealed heterozygous silent mutations in the A (c.276 A>G Gly) and C (c.126 C>T Pro) chains, as well as a homozygous single-base change in the 3' non-coding region of the B chain (c*78 A>G). C1qA mRNA expression without interferon was decreased compared with that of healthy controls (p<0.05) and was decreased after stimulation compared with that of non-treated cells. C1qB mRNA expression was decreased compared with that of controls and did not change with stimulation. C1qC mRNA expression was increased compared with that of controls and was even higher after stimulation. P2 and P3 had Type I C2 deficiency (heterozygous 28 bp deletion at exon 6). The C2 mRNA expression in P3 was 23 times lower compared with that of controls and did not change after stimulation. The C4B mRNA expression of P4 was decreased compared with that of controls and increased after stimulation. CONCLUSIONS Silent mutations and single-base changes in the 3' non-coding regions may modify mRNA transcription and C1q production. Type I C2 deficiency should be evaluated in JSLE patients with decreased C2 serum levels. Further studies are needed to clarify the role of decreased C4B mRNA expression in JSLE pathogenesis.
Collapse
Affiliation(s)
- Bernadete L Liphaus
- Laboratório de Investigação Médica 36, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Natalia Umetsu
- Laboratório de Investigação Médica 36, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Adriana A Jesus
- Unidade de Reumatologia, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvia Y Bando
- Laboratório de Investigação Médica 36, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Clovis A Silva
- Unidade de Reumatologia, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Magda Carneiro-Sampaio
- Laboratório de Investigação Médica 36, Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
24
|
Hereditary angioedema and lupus: A French retrospective study and literature review. Autoimmun Rev 2015; 14:564-8. [PMID: 25660269 DOI: 10.1016/j.autrev.2015.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 01/31/2015] [Indexed: 11/23/2022]
Abstract
Hereditary angioedema (HAE) is a rare genetic disorder that is primarily caused by a defect in the C1 inhibitor (C1-INH). The recurrent symptoms are subcutaneous edema and abdominal pain. Laryngeal edema, which can also occur, is life threatening if it goes untreated. HAE can be associated with some inflammatory and autoimmune disorders, particularly lupus. The aim of this study was to describe cases of lupus among HAE patients in France and to perform a literature review of lupus and HAE studies. Case detection and data collection (a standardized form) were performed, thanks to the French Reference Center for Kinin-related angioedema. Data were collected from 6 patients with type 1 HAE and lupus in France; no cases of systemic lupus erythematosus were reported. In the literature review, 32 cases of lupus combined with HAE were identified, including 26 female patients. The median patient age at the time of first reported HAE symptoms and at diagnosis were 17.5 years (range, 9-41 years) and 19 years (range, 9-64 years), respectively for our 6 patients and 14 years (range, 3-30 years) and 17 years (range, 7-48 years), respectively, for the literature review. The clinical manifestations of HAE were mainly abdominal pain (83% in our patients vs 47% in the literature) and edema of the limbs (83% vs 38%). The C4 levels were low (for 100% of our cases vs 93% in the literature). Eighteen patients in the literature demonstrated HAE symptoms prior to the lupus onset vs 5 for our patients. The mean patient age at lupus onset was 20 years (range, 13-76 years) for our patients and 19.5 years (range, 1-78 years) in the literature, respectively. In the literature, 81% of the patients had skin manifestations, 25% had renal involvement and 28% received systemic steroids to treat lupus. Treatment with danazol did not modify the clinical expression of lupus. The association between lupus and HAE is a rare but not unanticipated event. Patients are often symptomatic for HAE before developing lupus. Lupus cases associated with HAE share some characteristics of lupus cases related to other complement deficiencies, such as the absence of severity and the predominance of cutaneous symptoms.
Collapse
|
25
|
Kottyan L, Kelly JA, Harley JB. Genetics of lupus. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
26
|
Kurien BT, D'Sousa A, Bruner BF, Gross T, James JA, Targoff IN, Maier-Moore JS, Harley ITW, Wang H, Scofield RH. Prolidase deficiency breaks tolerance to lupus-associated antigens. Int J Rheum Dis 2013; 16:674-80. [PMID: 24330273 DOI: 10.1111/1756-185x.12254] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM Prolidase deficiency is a rare autosomal recessive disease in which one of the last steps of collagen metabolism, cleavage of proline-containing dipeptides, is impaired. Only about 93 patients have been reported with about 10% also having systemic lupus erythematosus (SLE). METHODS We studied a large extended Amish pedigree with four prolidase deficiency patients and three heterozygous individuals for lupus-associated autoimmunity. Eight unaffected Amish children served as normal controls. Prolidase genetics and enzyme activity were confirmed. Antinuclear antibodies (ANA) were determined using indirect immunofluorescence and antibodies against extractable nuclear antigens were determined by various methods, including double immunodiffusion, immunoprecipitation and multiplex bead assay. Serum C1q levels were determined by enzyme-linked immunosorbent assay. RESULTS Two of the four homozygous prolidase deficiency subjects had a positive ANA. One had anti-double-stranded DNA, while another had precipitating anti-Ro. By the simultaneous microbead assay, three of the four had anti-Sm and anti-chromatin. One of the three heterozygous subjects had a positive ANA and immunoprecipitation of a 75 000 molecular weight protein. The unaffected controls had normal prolidase activity and were negative for autoantibodies. CONCLUSIONS Prolidase deficiency may be associated with the loss of immune tolerance to lupus-associated autoantigens even without clinical SLE.
Collapse
Affiliation(s)
- Biji T Kurien
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Aringer M, Günther C, Lee-Kirsch MA. Innate immune processes in lupus erythematosus. Clin Immunol 2013; 147:216-22. [DOI: 10.1016/j.clim.2012.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/23/2012] [Accepted: 11/24/2012] [Indexed: 11/30/2022]
|
28
|
Kronbichler A, Mayer G. Renal involvement in autoimmune connective tissue diseases. BMC Med 2013; 11:95. [PMID: 23557013 PMCID: PMC3616816 DOI: 10.1186/1741-7015-11-95] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/11/2013] [Indexed: 01/04/2023] Open
Abstract
Connective tissue diseases (CTDs) are a heterogeneous group of disorders that share certain clinical presentations and a disturbed immunoregulation, leading to autoantibody production. Subclinical or overt renal manifestations are frequently observed and complicate the clinical course of CTDs. Alterations of kidney function in Sjögren syndrome, systemic scleroderma (SSc), auto-immune myopathies (dermatomyositis and polymyositis), systemic lupus erythematosus (SLE), antiphospholipid syndrome nephropathy (APSN) as well as rheumatoid arthritis (RA) are frequently present and physicians should be aware of that.In SLE, renal prognosis significantly improved based on specific classification and treatment strategies adjusted to kidney biopsy findings. Patients with scleroderma renal crisis (SRC), which is usually characterized by severe hypertension, progressive decline of renal function and thrombotic microangiopathy, show a significant benefit of early angiotensin-converting-enzyme (ACE) inhibitor use in particular and strict blood pressure control in general. Treatment of the underlying autoimmune disorder or discontinuation of specific therapeutic agents improves kidney function in most patients with Sjögren syndrome, auto-immune myopathies, APSN and RA.In this review we focus on impairment of renal function in relation to underlying disease or adverse drug effects and implications on treatment decisions.
Collapse
Affiliation(s)
- Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | | |
Collapse
|
29
|
Abstract
Genome-wide association studies have identified many dozen genetic intervals that harbor single-nucleotide polymorphisms (SNPs) showing statistical association with systemic lupus erythematosus. Despite the wealth of data produced, there are limitations of these studies. The causal alleles at a given locus are not identified; only SNP is strong linkage disequilibrium with the putative causative alleles. In order to address identification of the causative SNPs for lupus susceptibility genes, we have initiated a candidate gene study for which more than 40 investigators have contributed patient and control samples. In addition, these investigators have designated SNPs to be placed on a custom array. In this way fine mapping of genetic association findings can occur in order to identify causal alleles. These efforts have thus far benefitted greatly from comparisons of different ethnicities. Work on about ten previously identified associations has been published using this resource. Genome-wide association studies cannot identify rare SNPs or mutations, which may impart greater relative risks than common variants. Much of the genetics of lupus may be from rare variants or mutations. In order to approach this aspect of lupus genetics, next-generation sequencing has begun in which all exons will be sequenced in controls and patients. This effort can also be used to identify causal alleles from association intervals not yet otherwise identified.
Collapse
|