1
|
Zhuang S, Guo D, Yu D. A mini review of the pathogenesis of acute rheumatic fever and rheumatic heart disease. Front Cell Infect Microbiol 2025; 15:1447149. [PMID: 40276383 PMCID: PMC12018407 DOI: 10.3389/fcimb.2025.1447149] [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: 06/11/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Acute rheumatic fever (ARF) is an autoimmune disease caused by group A streptococcal infection. Recurrent episodes of ARF can lead to rheumatic heart disease (RHD), which is the leading cause of cardiovascular mortality in children worldwide, especially in low- and middle-income countries. Investigations into the etiology of ARF and RHD constitute a crucial milestone in the advancement of both preventive measures and therapeutic interventions. The purpose of this mini review is to delineate the etiology and pathophysiological mechanisms underlying ARF and RHD. Selective searches were conducted in PubMed to retrieve literature published between 1968 and 2024, employing key terms such as "acute rheumatic fever", "rheumatic heart disease", "group A Streptococcus", "streptococcal pharyngitis", "pathogenesis", and "pathophysiology". The pathogenesis of infections caused by group A streptococci, and their effects on ARF and RHD, have been thoroughly examined. A central hypothesis is that autoimmune responses are triggered by molecular mimicry, but alternate pathogenic mechanisms are continuously being explored. There is an urgent need for high-quality research that can inform efforts aimed at decreasing the occurrence of ARF and halting the advancement of RHD, which requires researchers to understand its causes and to develop appropriate preventive and therapeutic programs.
Collapse
Affiliation(s)
| | | | - Dingle Yu
- Department of Respiration, Shenzhen Children’s Hospital, Shenzhen Univesity,
Shantou University Medical College, Shenzhen, China
| |
Collapse
|
2
|
Polis B, Cuda CM, Putterman C. Animal models of neuropsychiatric systemic lupus erythematosus: deciphering the complexity and guiding therapeutic development. Autoimmunity 2024; 57:2330387. [PMID: 38555866 PMCID: PMC12069686 DOI: 10.1080/08916934.2024.2330387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Systemic lupus erythematosus (SLE) poses formidable challenges due to its multifaceted etiology while impacting multiple tissues and organs and displaying diverse clinical manifestations. Genetic and environmental factors contribute to SLE complexity, with relatively limited approved therapeutic options. Murine models offer insights into SLE pathogenesis but do not always replicate the nuances of human disease. This review critically evaluates spontaneous and induced animal models, emphasizing their validity and relevance to neuropsychiatric SLE (NPSLE). While these models undoubtedly contribute to understanding disease pathophysiology, discrepancies persist in mimicking some NPSLE intricacies. The lack of literature addressing this issue impedes therapeutic progress. We underscore the urgent need for refining models that truly reflect NPSLE complexities to enhance translational fidelity. We encourage a comprehensive, creative translational approach for targeted SLE interventions, balancing scientific progress with ethical considerations to eventually improve the management of NPSLE patients. A thorough grasp of these issues informs researchers in designing experiments, interpreting results, and exploring alternatives to advance NPSLE research.
Collapse
Affiliation(s)
- Baruh Polis
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Carla M. Cuda
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Rekvig OP. SLE: a cognitive step forward-a synthesis of rethinking theories, causality, and ignored DNA structures. Front Immunol 2024; 15:1393814. [PMID: 38895113 PMCID: PMC11183320 DOI: 10.3389/fimmu.2024.1393814] [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: 02/29/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is classified by instinctual classification criteria. A valid proclamation is that these formally accepted SLE classification criteria legitimate the syndrome as being difficult to explain and therefore enigmatic. SLE involves scientific problems linked to etiological factors and criteria. Our insufficient understanding of the clinical condition uniformly denoted SLE depends on the still open question of whether SLE is, according to classification criteria, a well-defined one disease entity or represents a variety of overlapping indistinct syndromes. Without rational hypotheses, these problems harm clear definition(s) of the syndrome. Why SLE is not anchored in logic, consequent, downstream interdependent and interactive inflammatory networks may rely on ignored predictive causality principles. Authoritative classification criteria do not reflect consequent causality criteria and do not unify characterization principles such as diagnostic criteria. We need now to reconcile legendary scientific achievements to concretize the delimitation of what SLE really is. Not all classified SLE syndromes are "genuine SLE"; many are theoretically "SLE-like non-SLE" syndromes. In this study, progressive theories imply imperative challenges to reconsider the fundamental impact of "the causality principle". This may offer us logic classification and diagnostic criteria aimed at identifying concise SLE syndromes as research objects. Can a systems science approach solve this problem?
Collapse
Affiliation(s)
- Ole Petter Rekvig
- Fürst Medical Laboratory, Oslo, Norway
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
4
|
Carroll KR, Mizrachi M, Simmons S, Toz B, Kowal C, Wingard J, Tehrani N, Zarfeshani A, Kello N, El Khoury L, Weissman-Tsukamoto R, Levin JZ, Volpe BT, Diamond B. Lupus autoantibodies initiate neuroinflammation sustained by continuous HMGB1:RAGE signaling and reversed by increased LAIR-1 expression. Nat Immunol 2024; 25:671-681. [PMID: 38448779 PMCID: PMC11141703 DOI: 10.1038/s41590-024-01772-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Cognitive impairment is a frequent manifestation of neuropsychiatric systemic lupus erythematosus, present in up to 80% of patients and leading to a diminished quality of life. In the present study, we used a model of lupus-like cognitive impairment that is initiated when antibodies that crossreact with excitatory neuronal receptors penetrate the hippocampus, causing immediate, self-limited, excitotoxic death of hippocampal neurons, which is then followed by a significant loss of dendritic complexity in surviving neurons. This injury creates a maladaptive equilibrium that is sustained in mice for at least 1 year. We identified a feedforward loop of microglial activation and microglia-dependent synapse elimination dependent on neuronal secretion of high mobility group box 1 protein (HMGB1) which binds the receptor for advanced glycation end products (RAGE) and leads to microglial secretion of C1q, upregulation of interleukin-10 with consequent downregulation of leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1), an inhibitory receptor for C1q. Treatment with a centrally acting angiotensin-converting enzyme inhibitor or with an angiotensin-receptor blocker restored a healthy equilibrium, microglial quiescence and intact spatial memory.
Collapse
Affiliation(s)
- Kaitlin R Carroll
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Mark Mizrachi
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Sean Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bahtiyar Toz
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Czeslawa Kowal
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Jeffrey Wingard
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Nazila Tehrani
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Aida Zarfeshani
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | | | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Betty Diamond
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
5
|
Chessa E, Piga M, Perra A, Pintus E, Porcu M, Serafini C, Congia M, Angioni MM, Naitza MR, Floris A, Mathieu A, Saba L, Carta MG, Cauli A. Effect of anti-P ribosomal and anti-NR2 antibodies on depression and cognitive processes in SLE: an integrated clinical and functional MRI study. Lupus Sci Med 2023; 10:e001005. [PMID: 37918951 PMCID: PMC10626760 DOI: 10.1136/lupus-2023-001005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023]
Abstract
OBJECTIVES To explore the effects of anti-ribosomal P protein (anti-P) and anti-N-methyl-D-aspartic acid receptor subunit 2 (anti-NR2) autoantibodies on depression and cognitive dysfunction and their relationships with functional brain connectivity in SLE. METHODS This cross-sectional study included adult patients who fulfilled the American College of Rheumatology/European Alliance of Associations for Rheumatology 2019 SLE criteria. Anti-P and anti-NR2 were quantified using ELISA. A 1-hour battery of neuropsychological testing interpreted by a neuropsychologist explored depressive symptoms (Center for Epidemiologic Studies Depression Scale, CES-D), cognitive domains and quality of life (SF-12). Resting-state functional connectivity (rs-fc) MRI analysis was performed within 1 month, and region-of-interest to region-of-interest (ROI-to-ROI) analyses with the graph theory were performed. RESULTS Thirty-three patients with SLE (9% male) were enrolled, mean age (SD) of 43.5 (14) years and median disease duration of 10.4 years (2.9-25.4). Anti-P was positive in 6 (18.2%) and anti-NR2 in 14 (42.4%) patients. Depressive symptoms were found in 14 (42.4%) patients using the CES-D (range 0-51). After correction for age, disease duration, disease activity and white matter lesion load, the CES-D score was independently associated with anti-P serum level (β=0.32; p=0.049) and prednisone daily dose (β=0.38; p=0.023). Nineteen patients (57.6%) showed at least a cognitive test alteration, but no significant association with autoantibodies was found. The rs-fc MRI analysis revealed an independent association between the anti-P serum levels and many altered brain ROI properties but no anti-NR2 and prednisone effects on the cerebral network. CONCLUSIONS Anti-P was associated with brain network perturbation, which may be responsible for depressive symptoms in patients with SLE.
Collapse
Affiliation(s)
| | - Matteo Piga
- Rheumatology Unit, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Perra
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Elisa Pintus
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Michele Porcu
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Cristina Serafini
- Rheumatology Unit, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Maria Maddalena Angioni
- Rheumatology Unit, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Micaela Rita Naitza
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alberto Floris
- Rheumatology Unit, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandro Mathieu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Luca Saba
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Radiology Department, AOU Cagliari, Cagliari, Italy
| | - Mauro Giovanni Carta
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alberto Cauli
- Rheumatology Unit, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
6
|
Cocco C, Manca E, Corda G, Angioni MM, Noli B, Congia M, Loy F, Isola M, Chessa E, Floris A, Lorefice L, Saba L, Mathieu A, Ferri GL, Cauli A, Piga M. Brain-reactive autoantibodies in neuropsychiatric systemic lupus erythematosus. Front Immunol 2023; 14:1157149. [PMID: 37383228 PMCID: PMC10294074 DOI: 10.3389/fimmu.2023.1157149] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction The pathogenesis of neuropsychiatric systemic lupus erythematosus (NPSLE) is widely unknown, and the role of autoantibodies is still undetermined. Methods To identify brain-reactive autoantibodies possibly related to NPSLE, immunofluorescence (IF) and transmission electron microscopy (TEM) on rat and human brains were performed. ELISA was used to reveal the presence of known circulating autoantibodies, while western blot (WB) was applied to characterize potential unknown autoantigen(s). Results We enrolled 209 subjects, including patients affected by SLE (n=69), NPSLE (n=36), Multiple Sclerosis (MS, n=22), and 82 age- and gender-matched healthy donors (HD). Autoantibody reactivity by IF was observed in almost the entire rat brain (cortex, hippocampus, and cerebellum) using sera from NPSLE and SLE patients and was virtually negative in MS and HD. NPSLE showed higher prevalence (OR 2.4; p = 0.047), intensity, and titer of brain-reactive autoantibodies than SLE patients. Most of the patient sera with brain-reactive autoantibodies (75%) also stained human brains. Double staining experiments on rat brains mixing patients' sera with antibodies directed against neuronal (NeuN) or glial markers showed autoantibody reactivity restricted to NeuN-containing neurons. Using TEM, the targets of brain-reactive autoantibodies were located in the nuclei and, to a lesser extent, in the cytoplasm and mitochondria. Given the high degree of colocalization between NeuN and brain-reactive autoantibodies, we assumed NeuN was a possible autoantigen. However, WB analysis with HEK293T cell lysates expressing or not expressing the gene encoding for NeuN protein (RIBFOX3) showed that patients' sera carrying brain-reactive autoantibodies did not recognize the NeuN corresponding band size. Among the panel of NPSLE-associated autoantibodies (e.g., anti-NR2, anti-P-ribosomal protein, antiphospholipid) investigated by ELISA assay, only the anti-β2-glycoprotein-I (aβ2GPI) IgG was exclusively found in those sera containing brain-reactive autoantibodies. Conclusion In conclusion, SLE and NPSLE patients possess brain-reactive autoantibodies but with higher frequency and titers found in NPSLE patients. Although many target antigens of brain-reactive autoantibodies are still undetermined, they likely include β2GPI.
Collapse
Affiliation(s)
- Cristina Cocco
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Elias Manca
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Giulia Corda
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Maddalena Angioni
- Rheumatology Unit, University Clinic, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Mattia Congia
- Rheumatology Unit, University Clinic, AOU Cagliari, Cagliari, Italy
| | - Francesco Loy
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Michela Isola
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | | | - Alberto Floris
- Rheumatology Unit, University Clinic, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ATS Sardegna, ASSL Cagliari, Cagliari, Italy
| | - Luca Saba
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
- Radiology Department, University Clinic, AOU Cagliari, Cagliari, Italy
| | - Alessandro Mathieu
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
| | - Gian Luca Ferri
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Alberto Cauli
- Rheumatology Unit, University Clinic, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
| | - Matteo Piga
- Rheumatology Unit, University Clinic, AOU Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public health, University of Cagliari, Monserrato, Italy
| |
Collapse
|
7
|
Gomez-Bañuelos E, Yu Y, Li J, Cashman KS, Paz M, Trejo-Zambrano MI, Bugrovsky R, Wang Y, Chida AS, Sherman-Baust CA, Ferris DP, Goldman DW, Darrah E, Petri M, Sanz I, Andrade F. Affinity maturation generates pathogenic antibodies with dual reactivity to DNase1L3 and dsDNA in systemic lupus erythematosus. Nat Commun 2023; 14:1388. [PMID: 36941260 PMCID: PMC10027674 DOI: 10.1038/s41467-023-37083-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023] Open
Abstract
Anti-dsDNA antibodies are pathogenically heterogeneous, implying distinct origins and antigenic properties. Unexpectedly, during the clinical and molecular characterization of autoantibodies to the endonuclease DNase1L3 in patients with systemic lupus erythematosus (SLE), we identified a subset of neutralizing anti-DNase1L3 antibodies previously catalogued as anti-dsDNA. Based on their variable heavy-chain (VH) gene usage, these antibodies can be divided in two groups. One group is encoded by the inherently autoreactive VH4-34 gene segment, derives from anti-DNase1L3 germline-encoded precursors, and gains cross-reactivity to dsDNA - and some additionally to cardiolipin - following somatic hypermutation. The second group, originally defined as nephritogenic anti-dsDNA antibodies, is encoded by diverse VH gene segments. Although affinity maturation results in dual reactivity to DNase1L3 and dsDNA, their binding efficiencies favor DNase1L3 as the primary antigen. Clinical, transcriptional and monoclonal antibody data support that cross-reactive anti-DNase1L3/dsDNA antibodies are more pathogenic than single reactive anti-dsDNA antibodies. These findings point to DNase1L3 as the primary target of a subset of antibodies classified as anti-dsDNA, shedding light on the origin and pathogenic heterogeneity of antibodies reactive to dsDNA in SLE.
Collapse
Affiliation(s)
- Eduardo Gomez-Bañuelos
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Yikai Yu
- Department of Rheumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, P. R. China
| | - Jessica Li
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Kevin S Cashman
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Merlin Paz
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | | | - Regina Bugrovsky
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Youliang Wang
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Asiya Seema Chida
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Cheryl A Sherman-Baust
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Dylan P Ferris
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Daniel W Goldman
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Erika Darrah
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Michelle Petri
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Iñaki Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Felipe Andrade
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA.
| |
Collapse
|
8
|
Moore E, Huang MW, Reynolds CA, Macian F, Putterman C. Choroid Plexus-Infiltrating T Cells as Drivers of Murine Neuropsychiatric Lupus. Arthritis Rheumatol 2022; 74:1796-1807. [PMID: 35637551 PMCID: PMC9825865 DOI: 10.1002/art.42252] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE T cells are critical in the pathogenesis of systemic lupus erythematosus (SLE) in that they secrete inflammatory cytokines, help autoantibody production, and form autoreactive memory T cells. Although the contribution of T cells to several forms of organ-mediated damage in SLE has been previously demonstrated, the role of T cells in neuropsychiatric SLE (NPSLE), which involves diffuse central nervous system manifestations and is observed in 20-40% of SLE patients, is not known. Therefore, we conducted this study to evaluate how behavioral deficits are altered after depletion or transfer of T cells, to directly assess the role of T cells in NPSLE. METHODS MRL/lpr mice, an NPSLE mouse model, were either systemically depleted of CD4+ T cells or intracerebroventricularly injected with choroid plexus (CP)-infiltrating T cells and subsequently evaluated for alterations in neuropsychiatric manifestations. Our study end points included evaluation of systemic disease and assessment of central nervous system changes. RESULTS Systemic depletion of CD4+ T cells ameliorated systemic disease and cognitive deficits. Intracerebroventricular injection of CP-infiltrating T cells exacerbated depressive-like behavior and worsened cognition in recipient mice compared with mice who received injection of splenic lupus T cells or phosphate buffered saline. Moreover, we observed enhanced activation in CP-infiltrating T cells when cocultured with brain lysate-pulsed dendritic cells in comparison to the activation levels observed in cocultures with splenic T cells. CONCLUSION T cells, and more specifically CP-infiltrating antigen-specific T cells, contributed to the pathogenesis of NPSLE in mice, indicating that, in the development of more targeted treatments for NPSLE, modulation of T cells may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| | - Michelle W. Huang
- Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| | - Cara A. Reynolds
- Department of PathologyAlbert Einstein College of MedicineNew York
| | - Fernando Macian
- Department of PathologyAlbert Einstein College of MedicineNew York
| | - Chaim Putterman
- Azrieli Faculty of Medicine of Bar‐Ilan University, Safed, Israel, Galilee Research Institute, Nahariya, Israel, and Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| |
Collapse
|
9
|
Manca E. Autoantibodies in Neuropsychiatric Systemic Lupus Erythematosus (NPSLE): Can They Be Used as Biomarkers for the Differential Diagnosis of This Disease? Clin Rev Allergy Immunol 2022; 63:194-209. [PMID: 34115263 PMCID: PMC9464150 DOI: 10.1007/s12016-021-08865-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus is a complex immunological disease where both environmental factors and genetic predisposition lead to the dysregulation of important immune mechanisms. Eventually, the combination of these factors leads to the production of self-reactive antibodies that can target any organ or tissue of the human body. Autoantibodies can form immune complexes responsible for both the organ damage and the most severe complications. Involvement of the central nervous system defines a subcategory of the disease, generally known with the denomination of neuropsychiatric systemic lupus erythematosus. Neuropsychiatric symptoms can range from relatively mild manifestations, such as headache, to more severe complications, such as psychosis. The evaluation of the presence of the autoantibodies in the serum of these patients is the most helpful diagnostic tool for the assessment of the disease. The scientific progresses achieved in the last decades helped researchers and physicians to discover some of autoepitopes targeted by the autoantibodies, although the majority of them have not been identified yet. Additionally, the central nervous system is full of epitopes that cannot be found elsewhere in the human body, for this reason, autoantibodies that selectively target these epitopes might be used for the differential diagnosis between patients with and without the neuropsychiatric symptoms. In this review, the most relevant data is reported with regard to mechanisms implicated in the production of autoantibodies and the most important autoantibodies found among patients with systemic lupus erythematosus with and without the neuropsychiatric manifestations.
Collapse
Affiliation(s)
- Elias Manca
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Cagliari, Italy.
| |
Collapse
|
10
|
Bruschi M, Angeletti A, Kajana X, Moroni G, Sinico RA, Fredi M, Vaglio A, Cavagna L, Pratesi F, Migliorini P, Locatelli F, Pazzola G, Pesce G, Bagnasco M, Manfredi A, Ramirez GA, Esposito P, Negrini S, Bui F, Trezzi B, Emmi G, Cavazzana I, Binda V, Fenaroli P, Pisani I, Montecucco C, Santoro D, Scolari F, Volpi S, Mosca M, Tincani A, Candiano G, Verrina E, Franceschini F, Ravelli A, Prunotto M, Meroni PL, Ghiggeri GM. Evidence for charge-based mimicry in anti dsDNA antibody generation. J Autoimmun 2022; 132:102900. [PMID: 36087539 DOI: 10.1016/j.jaut.2022.102900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
Mechanisms for the generation of anti-dsDNA autoantibodies are still not completely elucidated. One theory states that dsDNA interacts for mimicry with antibodies raised versus other antigens but molecular features for mimicry are unknown. Here we show that, at physiological acid-base balance, anti-Annexin A1 binds IgG2 dsDNA in a competitive and dose-dependent way with Annexin A1 and that the competition between the two molecules is null at pH 9. On the other hand, these findings also show that dsDNA and Annexin A1 interact with their respective antibodies on a strictly pH-dependent basis: in both cases, the binding was minimal at pH 4 and maximal at pH9-10. The anionic charge of dsDNA is mainly conferred by the numerous phosphatidic residues. The epitope binding site of Annexin A1 for anti-Annexin A1 IgG2 was here characterized as a string of 34 amino acids at the NH2 terminus, 10 of which are anionic. Circulating levels of anti-dsDNA and anti-Annexin A1 IgG2 antibodies were strongly correlated in patients with systemic lupus erythematosus (n 496) and lupus nephritis (n 425) stratified for age, sex, etc. These results show that dsDNA competes with Annexin A1 for the binding with anti-Annexin A1 IgG2 on a dose and charged mediated base, being able to display an inhibition up to 75%. This study provides the first demonstration that dsDNA may interact with antibodies raised versus other anionic molecules (anti-Annexin A1 IgG2) because of charge mimicry and this interaction may contribute to anti-dsDNA antibodies generation.
Collapse
Affiliation(s)
- Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Angeletti
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Xhuliana Kajana
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gabriella Moroni
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | | | - Micaela Fredi
- Rheumatology and Clinical Immunology, ASST Spedali Civili and University of Brescia, Italy
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, And Nephrology and Dialysis Unit, Meyer Children's Hospital, Florence, Italy
| | - Lorenzo Cavagna
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Federico Pratesi
- Clinical Immunology Unit, Department of Internal Medicine, University of Pisa, Italy
| | - Paola Migliorini
- Clinical Immunology Unit, Department of Internal Medicine, University of Pisa, Italy
| | - Francesco Locatelli
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Giulia Pazzola
- Nephrology and Dialysis, Arciospedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Giampaola Pesce
- Medical and Radiometabolic Therapy Unit, Department of Internal Medicine, University of Genoa, Italy
| | - Marcello Bagnasco
- Medical and Radiometabolic Therapy Unit, Department of Internal Medicine, University of Genoa, Italy
| | - Angelo Manfredi
- Unit of Internal Medicine and Immunology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Pasquale Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Simone Negrini
- Department of Internal Medicine, University of Genoa, Italy
| | - Federica Bui
- Division of Nephrology, University of Genoa and Policlinico San Martino, Genoa, Italy
| | - Barbara Trezzi
- Department of Medicine and Surgery, University of Milan, Bicocca, Italy
| | - Giacomo Emmi
- Lupus Clinic Department of Biomedicine, University of Florence, University Hospital Careggi, Florence, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical Immunology, ASST Spedali Civili and University of Brescia, Italy
| | - Valentina Binda
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Paride Fenaroli
- Nephrology Unit, University Hospital, University of Parma, Parma, Italy
| | - Isabella Pisani
- Nephrology Unit, University Hospital, University of Parma, Parma, Italy
| | | | - Domenico Santoro
- Nephrology and Dialysis Unit, University of Messina and G Martino Hospital, Messina, Italy
| | - Francesco Scolari
- Division of Nephrology and Dialysis, University of Brescia and Ospedale di Montichiari, Brescia, Italy
| | - Stefano Volpi
- Division of Pediatric Rheumatology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marta Mosca
- Rheumatologu Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology, ASST Spedali Civili and University of Brescia, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Verrina
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunology, ASST Spedali Civili and University of Brescia, Italy
| | - Angelo Ravelli
- Division of Pediatric Rheumatology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Prunotto
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Pier Luigi Meroni
- Experimental Laboratory of Immunological and Rheumatologic Researches, IRCCS Istituto Auxologico Italiano, Milan, Italy.
| | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| |
Collapse
|
11
|
Rekvig OP. The Anti-DNA Antibodies: Their Specificities for Unique DNA Structures and Their Unresolved Clinical Impact-A System Criticism and a Hypothesis. Front Immunol 2022; 12:808008. [PMID: 35087528 PMCID: PMC8786728 DOI: 10.3389/fimmu.2021.808008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is diagnosed and classified by criteria, or by experience, intuition and traditions, and not by scientifically well-defined etiology(ies) or pathogenicity(ies). One central criterion and diagnostic factor is founded on theoretical and analytical approaches based on our imperfect definition of the term “The anti-dsDNA antibody”. “The anti-dsDNA antibody” holds an archaic position in SLE as a unique classification criterium and pathogenic factor. In a wider sense, antibodies to unique transcriptionally active or silent DNA structures and chromatin components may have individual and profound nephritogenic impact although not considered yet – not in theoretical nor in descriptive or experimental contexts. This hypothesis is contemplated here. In this analysis, our state-of-the-art conception of these antibodies is probed and found too deficient with respect to their origin, structural DNA specificities and clinical/pathogenic impact. Discoveries of DNA structures and functions started with Miescher’s Nuclein (1871), via Chargaff, Franklin, Watson and Crick, and continues today. The discoveries have left us with a DNA helix that presents distinct structures expressing unique operations of DNA. All structures are proven immunogenic! Unique autoimmune antibodies are described against e.g. ssDNA, elongated B DNA, bent B DNA, Z DNA, cruciform DNA, or individual components of chromatin. In light of the massive scientific interest in anti-DNA antibodies over decades, it is an unexpected observation that the spectrum of DNA structures has been known for decades without being implemented in clinical immunology. This leads consequently to a critical analysis of historical and contemporary evidence-based data and of ignored and one-dimensional contexts and hypotheses: i.e. “one antibody - one disease”. In this study radical viewpoints on the impact of DNA and chromatin immunity/autoimmunity are considered and discussed in context of the pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Ole Petter Rekvig
- Section of Autoimmunity, Fürst Medical Laboratory, Oslo, Norway.,Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
12
|
Sheard DE, Li W, O’Brien-Simpson NM, Separovic F, Wade JD. Peptide Multimerization as Leads for Therapeutic Development. BIOLOGICS 2021; 2:15-44. [DOI: 10.3390/biologics2010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Multimerization of peptide structures has been a logical evolution in their development as potential therapeutic molecules. The multivalent properties of these assemblies have attracted much attention from researchers in the past and the development of more complex branching dendrimeric structures, with a wide array of biocompatible building blocks is revealing previously unseen properties and activities. These branching multimer and dendrimer structures can induce greater effect on cellular targets than monomeric forms and act as potent antimicrobials, potential vaccine alternatives and promising candidates in biomedical imaging and drug delivery applications. This review aims to outline the chemical synthetic innovations for the development of these highly complex structures and highlight the extensive capabilities of these molecules to rival those of natural biomolecules.
Collapse
Affiliation(s)
- Dean E. Sheard
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Wenyi Li
- ACTV Research Group, Centre for Oral Health Research, The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Neil M. O’Brien-Simpson
- ACTV Research Group, Centre for Oral Health Research, The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Frances Separovic
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
| | - John D. Wade
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
13
|
Wollmuth LP, Chan K, Groc L. The diverse and complex modes of action of anti-NMDA receptor autoantibodies. Neuropharmacology 2021; 194:108624. [PMID: 34081993 PMCID: PMC8693782 DOI: 10.1016/j.neuropharm.2021.108624] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/21/2022]
Abstract
NMDA receptors are ligand-gated ion channels that are found throughout the brain and are required for both brain development and many higher order functions. A variety of human patients with diverse clinical phenotypes have been identified that carry autoantibodies directed against NMDA receptor subunits. Here we focus on two general classes of autoantibodies, anti-GluN1 antibodies associated with anti-NMDA receptor encephalitis and anti-GluN2 antibodies associated with systemic lupus erythematosus (SLE). These two general classes of anti-NMDA receptor autoantibodies display a wide range of pathophysiological mechanisms from altering synaptic composition to gating of NMDARs. While we have made progress in understanding how these autoantibodies work at the molecular and cellular level, many unanswered questions remain including their long-term actions on brain function, the significance of clonal variations, and their effects on different NMDA receptor-expressing cell types in local circuits. This information will be needed to define fully the transition from anti-NMDA receptor autoantibodies to a clinical phenotype.
Collapse
Affiliation(s)
- Lonnie P Wollmuth
- Department of Neurobiology & Behavior, USA; Department of Biochemistry & Cell Biology, USA; Center for Nervous System Disorders. Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| | - Kelvin Chan
- Graduate Program in Neuroscience, USA; Medical Scientist Training Program (MSTP), USA; Department of Neurobiology & Behavior, USA
| | - Laurent Groc
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, IINS UMR, 5297, Bordeaux, France
| |
Collapse
|
14
|
Wangriatisak K, Thanadetsuntorn C, Krittayapoositpot T, Leepiyasakulchai C, Suangtamai T, Ngamjanyaporn P, Khowawisetsut L, Khaenam P, Setthaudom C, Pisitkun P, Chootong P. The expansion of activated naive DNA autoreactive B cells and its association with disease activity in systemic lupus erythematosus patients. Arthritis Res Ther 2021; 23:179. [PMID: 34229724 PMCID: PMC8259008 DOI: 10.1186/s13075-021-02557-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/20/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Autoreactive B cells are well recognized as key participants in the pathogenesis of systemic lupus erythematosus (SLE). However, elucidating the particular subset of B cells in producing anti-dsDNA antibodies is limited due to their B cell heterogeneity. This study aimed to identify peripheral B cell subpopulations that display autoreactivity to DNA and contribute to lupus pathogenesis. METHODS Flow cytometry was used to detect total B cell subsets (n = 20) and DNA autoreactive B cells (n = 15) in SLE patients' peripheral blood. Clinical disease activities were assessed in SLE patients using modified SLEDAI-2 K and used for correlation analyses with expanded B cell subsets and DNA autoreactive B cells. RESULTS The increases of circulating double negative 2 (DN2) and activated naïve (aNAV) B cells were significantly observed in SLE patients. Expanded B cell subsets and DNA autoreactive B cells represented a high proportion of aNAV B cells with overexpression of CD69 and CD86. The frequencies of aNAV B cells in total B cell populations were significantly correlated with modified SLEDAI-2 K scores. Further analysis showed that expansion of aNAV DNA autoreactive B cells was more related to disease activity and serum anti-dsDNA antibody levels than to total aNAV B cells. CONCLUSION Our study demonstrated an expansion of aNAV B cells in SLE patients. The association between the frequency of aNAV B cells and disease activity patients suggested that these expanded B cells may play a role in SLE pathogenesis.
Collapse
Affiliation(s)
- Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Chokchai Thanadetsuntorn
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Thamonwan Krittayapoositpot
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Thanitta Suangtamai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pintip Ngamjanyaporn
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Prasong Khaenam
- Center of Standardization and Product Validation, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chavachol Setthaudom
- Immunology Laboratory, Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand.
- Translational Medicine Program, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand.
| |
Collapse
|
15
|
Browne K, Zhang E, Sullivan JK, Evonuk KS, DeSilva TM, Jorgensen TN. Lupus-prone B6.Nba2 male and female mice display anti-DWEYS reactivity and a neuropsychiatric phenotype. Brain Behav Immun 2021; 94:175-184. [PMID: 33607233 PMCID: PMC10874234 DOI: 10.1016/j.bbi.2021.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Neuropsychiatric lupus (NPSLE), a manifestation of the autoimmune disease systemic lupus erythematosus (SLE), is characterized by psychiatric symptoms including anxiety and depression and upregulated autoantibodies. The B6.Nba2 spontaneous mouse model develops SLE, but has not previously been tested for NPSLE. METHODS We investigated the NPSLE phenotype in male and female B6.Nba2 mice (n = 12 each) and age- and sex-matched B6 controls (n = 10 each) via behavioral assessments for anxiety, depression, and memory deficits. Serum anti-dsDNA, anti-nRNP, anti-DWEYS peptide reactive IgG autoantibody levels and soluble TWEAK levels were determined by ELISA. Hippocampal regions were stained for activated microglia and neurons. RESULTS Both male and female B6.Nba2 mice showed elevated anti-dsDNA IgG, anti-nRNP IgG and anti-DWEYS reactive antibodies, elevated serum soluble TWEAK levels, and a strong anxiety and depression phenotype (p < 0.05-0.0001). Male B6.Nba2 mice developed this phenotype at a slightly older age than females. Female B6.Nba2 mice displayed reduced numbers of neurons in the hippocampal region compared to female B6 controls (p < 0.05). CONCLUSION The B6.Nba2 mouse model recapitulates many known NPSLE phenotypes, making it a promising model to investigate the development of NPSLE in the context of SLE.
Collapse
Affiliation(s)
- Kim Browne
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Emily Zhang
- Cleveland Clinic at Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - James K Sullivan
- Cleveland Clinic at Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - Kirsten S Evonuk
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Tara M DeSilva
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Trine N Jorgensen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
16
|
Zarfeshani A, Carroll KR, Volpe BT, Diamond B. Cognitive Impairment in SLE: Mechanisms and Therapeutic Approaches. Curr Rheumatol Rep 2021; 23:25. [PMID: 33782842 PMCID: PMC11207197 DOI: 10.1007/s11926-021-00992-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
A wide range of patients with systemic lupus erythematosus (SLE) suffer from cognitive dysfunction (CD) which severely impacts their quality of life. However, CD remains underdiagnosed and poorly understood. Here, we discuss current findings in patients and in animal models. Strong evidence suggests that CD pathogenesis involves known mechanisms of tissue injury in SLE. These mechanisms recruit brain resident cells, in particular microglia, into the pathological process. While systemic immune activation is critical to central nervous system injury, the current focus of therapy is the microglial cell and not the systemic immune perturbation. Further studies are critical to examine additional potential therapeutic targets and more specific treatments based on the cause and progress of the disease.
Collapse
Affiliation(s)
- Aida Zarfeshani
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kaitlin R Carroll
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
17
|
Nowling TK, Kral M, Wolf B, Gilkeson G, Ruth NM. Formal neurocognitive function and anti-N-methyl-D-aspartate receptor antibodies in paediatric lupus. Lupus Sci Med 2021; 8:8/1/e000462. [PMID: 33758010 PMCID: PMC7993203 DOI: 10.1136/lupus-2020-000462] [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: 11/23/2020] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 11/22/2022]
Abstract
Objective SLE is a chronic multisystem autoimmune inflammatory disease impacting a number of organs, including the central nervous system (CNS). The pathophysiology of CNS lupus is multifactorial, making diagnosis problematic. Neurocognitive (NC) testing and specific biomarkers to identify the development of neuropsychiatric (NP) symptoms in lupus are needed. Paediatric patients with SLE have high incidence of NP disease. While serum anti-N-methyl-D-aspartate receptor (NMDAR) antibodies have shown promise as a biomarker of NP in adults with SLE, much less is known with regard to paediatric patients with SLE. Methods We performed a cross-sectional study in paediatric patients with SLE. Serum NMDAR antibodies were measured and compared with levels in patients with juvenile idiopathic arthritis (JIA). Formal NC testing was performed in accordance with the Childhood Arthritis & Rheumatology Research Alliance neuropsychological core test battery. NC functioning was compared in the two groups and with NMDAR antibody levels. Results Serum NMDAR antibody levels were significantly higher in paediatric patients with SLE compared with patients with JIA. There were no significant correlations between NMDAR antibody levels and any measure of NC functioning. In an exploratory examination of anti-ribosomal P (RibP) antibody and NC functioning in a subset of patients with SLE, RibP antibody-positive patients exhibited worse scores for Verbal Memory Index and Design Fluency Test Switching compared with RibP antibody-negative patients. A globally significant association between disease status and NC functioning was observed. Specifically, patients with SLE had lower scores compared with patients with JIA for full-scale IQ, letter–word recognition, reading fluency and calculation skills after adjusting for multiple comparisons. Conclusion These collective results suggest that although serum NMDAR may serve as a biomarker, formal NC testing is superior in identifying paediatric patients with SLE with NP manifestations. RibP also may potentially serve as a biomarker of NP manifestations in paediatric patients with SLE. Additional and longitudinal studies are needed.
Collapse
Affiliation(s)
- Tamara K Nowling
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mary Kral
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gary Gilkeson
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Natasha McKerran Ruth
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
18
|
Reséndiz-Mora A, Wong-Baeza C, Nevárez-Lechuga I, Landa-Saldívar C, Molina-Gómez E, Hernández-Pando R, Wong-Baeza I, Escobar-Gutiérrez A, Baeza I. Interleukin 4 deficiency limits the development of a lupus-like disease in mice triggered by phospholipids in a non-bilayer arrangement. Scand J Immunol 2020; 93:e13002. [PMID: 33247472 DOI: 10.1111/sji.13002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/30/2020] [Accepted: 11/22/2020] [Indexed: 11/29/2022]
Abstract
Non-bilayer phospholipids arrangements (NPAs) are transient molecular associations different from lipid bilayers. When they become stable, they can trigger a disease in mice resembling human lupus, which is mainly characterized by the production of anti-NPA IgG antibodies. NPAs are stabilized on liposomes or cell bilayers by the drugs procainamide or chlorpromazine, which produce drug-induced lupus in humans. Here, we evaluated the participation of the TH 2 response, through its hallmark cytokine IL-4, on the development of the lupus-like disease in mice. Wild-type or IL-4 knockout BALB/c mice received liposomes bearing drug-induced NPAs, the drugs alone, or an anti-NPA monoclonal antibody (H308) to induce the lupus-like disease (the last two procedures stabilize NPAs on mice cells). IL-4 KO mice showed minor disease manifestations, compared to wild-type mice, with decreased production of anti-NPA IgG antibodies, no anti-cardiolipin, anti-histones and anticoagulant antibodies, and no kidney or skin lesions. In these mice, H308 was the only inducer of anti-NPA IgG antibodies. These findings indicate that IL-4 has a central role in the development of the murine lupus-like disease induced by NPA stabilization.
Collapse
Affiliation(s)
- Albany Reséndiz-Mora
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Carlos Wong-Baeza
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Irene Nevárez-Lechuga
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Carla Landa-Saldívar
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Eréndira Molina-Gómez
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Ciudad de México, México
| | - Isabel Wong-Baeza
- Laboratorio de Inmunología Molecular II, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Isabel Baeza
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
19
|
Chan K, Nestor J, Huerta TS, Certain N, Moody G, Kowal C, Huerta PT, Volpe BT, Diamond B, Wollmuth LP. Lupus autoantibodies act as positive allosteric modulators at GluN2A-containing NMDA receptors and impair spatial memory. Nat Commun 2020; 11:1403. [PMID: 32179753 PMCID: PMC7075964 DOI: 10.1038/s41467-020-15224-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
Patients with Systemic lupus erythematosus (SLE) experience various peripheral and central nervous system manifestations including spatial memory impairment. A subset of autoantibodies (DNRAbs) cross-react with the GluN2A and GluN2B subunits of the NMDA receptor (NMDAR). We find that these DNRAbs act as positive allosteric modulators on NMDARs with GluN2A-containing NMDARs, even those containing a single GluN2A subunit, exhibiting a much greater sensitivity to DNRAbs than those with exclusively GluN2B. Accordingly, GluN2A-specific antagonists provide greater protection from DNRAb-mediated neuronal cell death than GluN2B antagonists. Using transgenic mice to perturb expression of either GluN2A or GluN2B in vivo, we find that DNRAb-mediated disruption of spatial memory characterized by early neuronal cell death and subsequent microglia-dependent pathologies requires GluN2A-containing NMDARs. Our results indicate that GluN2A-specific antagonists or negative allosteric modulators are strong candidates to treat SLE patients with nervous system dysfunction.
Collapse
Affiliation(s)
- Kelvin Chan
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Jacquelyn Nestor
- Donald & Barbara Zucker School of Medicine, Hofstra University, Hempstead, NY, 11549, USA
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Tomás S Huerta
- Donald & Barbara Zucker School of Medicine, Hofstra University, Hempstead, NY, 11549, USA
| | - Noele Certain
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Gabrielle Moody
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Czeslawa Kowal
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Patricio T Huerta
- Donald & Barbara Zucker School of Medicine, Hofstra University, Hempstead, NY, 11549, USA
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Bruce T Volpe
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
| | - Lonnie P Wollmuth
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| |
Collapse
|
20
|
Wang H, Lu M, Zhai S, Wu K, Peng L, Yang J, Xia Y. ALW peptide ameliorates lupus nephritis in MRL/lpr mice. Arthritis Res Ther 2019; 21:261. [PMID: 31791413 PMCID: PMC6889545 DOI: 10.1186/s13075-019-2038-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Background Lupus nephritis (LN) is a common and serious complication of systemic lupus erythematosus. Anti-double-stranded (ds) DNA immunoglobulin G (IgG) plays a pivotal role in the pathogenesis of LN. Currently, there are various therapies for patients with LN; however, most of them are associated with considerable side effects. We confirmed previously that ALW (ALWPPNLHAWVP), a 12-amino acid peptide, inhibited the binding of polyclonal anti-dsDNA antibodies to mesangial cells and isolated glomeruli in vitro. In this study, we further investigate whether the administration of ALW peptide decreases renal IgG deposition and relevant damage in MRL/lpr lupus-prone mice. Methods Forty female MRL/lpr mice were randomly divided into four groups. The mice were intravenously injected with D-form ALW peptide (ALW group), scrambled peptide (PLP group), and normal saline (NaCl group) or were not treated (blank group). The IgG deposition, the histopathologic changes, and the expressions of profibrotic factors were analyzed in the kidney of MRL/lpr mice. Results Compared with the other groups, glomerular deposition of IgG, IgG2a, IgG2b, and IgG3 was decreased in the ALW group. Moreover, ALW administration attenuated renal histopathologic changes in MRL/lpr mice, including mesangial proliferation and infiltration of inflammatory cells. Furthermore, the expressions of profibrotic cytokines, such as transforming growth factor-beta1 (TGF-β1) and platelet-derived growth factor B (PDGF-B), decreased in the serum and kidney tissue of ALW-treated mice. Conclusions Our study demonstrated that ALW peptide ameliorates the murine model of LN, possibly through inhibiting renal IgG deposition and relevant tissue inflammation and fibrosis.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Siyue Zhai
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Kunyi Wu
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lingling Peng
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jie Yang
- Department of Nephrology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
21
|
Cunningham MW. Molecular Mimicry, Autoimmunity, and Infection: The Cross-Reactive Antigens of Group A Streptococci and their Sequelae. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0045-2018. [PMID: 31373269 PMCID: PMC6684244 DOI: 10.1128/microbiolspec.gpp3-0045-2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
The group A streptococci are associated with a group of diseases affecting the heart, brain, and joints that are collectively referred to as acute rheumatic fever. The streptococcal immune-mediated sequelae, including acute rheumatic fever, are due to antibody and cellular immune responses that target antigens in the heart and brain as well as the group A streptococcal cross-reactive antigens as reviewed in this article. The pathogenesis of acute rheumatic fever, rheumatic heart disease, Sydenham chorea, and other autoimmune sequelae is related to autoantibodies that are characteristic of autoimmune diseases and result from the immune responses against group A streptococcal infection by the host. The sharing of host and streptococcal epitopes leads to molecular mimicry between the streptococcal and host antigens that are recognized by the autoantibodies during the host response. This article elaborates on the discoveries that led to a better understanding of the pathogenesis of disease and provides an overview of the history and the most current thought about the immune responses against the host and streptococcal cross-reactive antigens in group A streptococcal sequelae.
Collapse
Affiliation(s)
- Madeleine W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190
| |
Collapse
|
22
|
James JA, Chen H, Young KA, Bemis EA, Seifert J, Bourn RL, Deane KD, Demoruelle MK, Feser M, O'Dell JR, Weisman MH, Keating RM, Gaffney PM, Kelly JA, Langefeld CD, Harley JB, Robinson W, Hafler DA, O'Connor KC, Buckner J, Guthridge JM, Norris JM, Holers VM. Latent autoimmunity across disease-specific boundaries in at-risk first-degree relatives of SLE and RA patients. EBioMedicine 2019; 42:76-85. [PMID: 30952617 PMCID: PMC6491794 DOI: 10.1016/j.ebiom.2019.03.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/15/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Autoimmune disease prevention requires tools to assess an individual's risk of developing a specific disease. One tool is disease-associated autoantibodies, which accumulate in an asymptomatic preclinical period. However, patients sometimes exhibit autoantibodies associated with a different disease classification. When and how these alternative autoantibodies first appear remain unknown. This cross-sectional study characterizes alternative autoimmunity, and associated genetic and environmental factors, in unaffected first-degree relatives (FDRs) of patients, who exhibit increased future risk for the same disease. METHODS Samples (n = 1321) from disease-specific autoantibody-positive (aAb+) systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and type 1 diabetes (T1D) patients; and unaffected aAb+ and autoantibody-negative (aAb-) SLE and RA FDRs were tested for SLE, RA, and T1D aAbs, as well as anti-tissue transglutaminase, anti-cardiolipin and anti-thyroperoxidase. FDR SLE and RA genetic risk scores (GRS) were calculated. FINDINGS Alternative autoimmunity occurred in SLE patients (56%) and FDRs (57·4%), RA patients (32·6%) and FDRs (34·8%), and T1D patients (43%). Expanded autoimmunity, defined as autoantibodies spanning at least two other diseases, occurred in 18·5% of SLE patients, 16·4% of SLE FDRs, 7·8% of RA patients, 5·3% of RA FDRs, and 10·8% of T1D patients. SLE FDRs were more likely to have alternative (odds ratio [OR] 2·44) and expanded (OR 3·27) autoimmunity than RA FDRs. Alternative and expanded autoimmunity were associated with several environmental exposures. Alternative autoimmunity was associated with a higher RA GRS in RA FDRs (OR 1·41), and a higher SLE GRS in aAb+ RA FDRs (OR 1·87), but not in SLE FDRs. INTERPRETATION Autoimmunity commonly crosses disease-specific boundaries in systemic (RA, SLE) and organ-specific (T1D) autoimmune diseases. Alternative autoimmunity is more common in SLE FDRs than RA FDRs, and is influenced by genetic and environmental factors. These findings have substantial implications for preclinical disease pathogenesis and autoimmune disease prevention studies. FUND: NIH U01AI101981, R01AR051394, U19AI082714, P30AR053483, P30GM103510, U54GM104938, U01AI101934, R01AI024717, U01AI130830, I01BX001834, & U01HG008666.
Collapse
Affiliation(s)
- Judith A James
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Hua Chen
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | | | | | - Rebecka L Bourn
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | | | | | | | | | | | | | | | | | - John B Harley
- Cincinnati Children's Hospital Medical Center, The University of Cincinnati College of Medicine, and Cincinnati US Department of Veterans Affairs VA Medical Center, Cincinnati, OH, USA
| | | | | | | | - Jane Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | | | | | |
Collapse
|
23
|
Mazzoleni A, Mallet JM, Rovero P, Papini AM. Glycoreplica peptides to investigate molecular mechanisms of immune-mediated physiological versus pathological conditions. Arch Biochem Biophys 2019; 663:44-53. [PMID: 30594643 DOI: 10.1016/j.abb.2018.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 12/17/2022]
Abstract
Investigation of the role of saccharides and glycoconjugates in mechanisms of immune-mediated physiological and pathological conditions is a hot topic. In fact, in many autoimmune diseases cross-reactivity between sugar moieties exposed on exogenous pathogens and self-molecules has long been hinted. Several peptides have been reported as mimetics of glycans specifically interacting with sugar-binding antibodies. The seek for these glycoreplica peptides is instrumental in characterizing antigen mimicry pathways and their involvement in triggering autoimmunity. Therefore, peptides mimicking glycan-protein interactions are valuable molecular tools to overcome the difficulties of oligosaccharide preparations. The clinical impact of peptide-based probes for autoimmune diseases diagnosis and follow-up is emerging only recently as just the tip of the iceberg of an overlooked potential. Here we provide a brief overview of the relevance of the structural and functional aspects of peptide probes and their mimicry effect in autoimmunity mechanisms for promising applications in diagnostics and therapeutics.
Collapse
Affiliation(s)
- Antonio Mazzoleni
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy; Laboratoire des Biomolécules, UMR 7203, Département de chimie, École Normale Supérieure, PSL Research University, Sorbonne Universités, UPMC Université Paris 06, CNRS, 24 rue Lhomond, 75005, Paris, France
| | - Jean-Maurice Mallet
- Laboratoire des Biomolécules, UMR 7203, Département de chimie, École Normale Supérieure, PSL Research University, Sorbonne Universités, UPMC Université Paris 06, CNRS, 24 rue Lhomond, 75005, Paris, France
| | - Paolo Rovero
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Neurosciences, Psychology, Drug Research and Child Health - Section of Pharmaceutical Sciences and Nutraceuticals, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy; Platform of Peptide and Protein Chemistry and Biology - PeptLab@UCP and Laboratory of Chemical Biology EA4505, Université Paris-Seine, 5 Mail Gay-Lussac, 95031, Cergy-Pontoise CEDEX, France.
| |
Collapse
|
24
|
Porat A, Giat E, Kowal C, He M, Son M, Latz E, Ben-Zvi I, Al-Abed Y, Diamond B. DNA-Mediated Interferon Signature Induction by SLE Serum Occurs in Monocytes Through Two Pathways: A Mechanism to Inhibit Both Pathways. Front Immunol 2018; 9:2824. [PMID: 30619247 PMCID: PMC6297782 DOI: 10.3389/fimmu.2018.02824] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/15/2018] [Indexed: 02/05/2023] Open
Abstract
A primary mechanism for activation of innate immunity is recognition of damage or pathogen associated molecular patterns by pattern recognition receptors (PRRs). Nucleic acid is a damage associated molecular pattern molecule that when internalized into a monocyte and recognized by intracellular nucleic acid sensing toll like receptors will cause production of type 1 interferon. The process by which DNA or RNA is delivered into the cytosol of monocytes in systemic lupus erythematosus remains incompletely understood, and therapeutic approaches to prevent DNA-mediated monocyte activation are needed. We identified two mechanisms for internalization of DNA by monocytes. IgG-bound DNA was internalized by interacting with Fc gamma receptor IIa, while high-mobility group box-1 protein-bound DNA was internalized by interacting with the receptor for advanced glycation end products. Both pathways contribute to an inflammatory phenotype in monocytes exposed to serum from patients with SLE. Moreover, both of these pathways can be inhibited by a pentapeptide, DWEYS, which is a DNA mimetope. In one instance DWEYS directly competes with DNA for antibody binding and in the other DWEYS binds high-mobility group box-1 and blocks its interaction with RAGE. Our data highlight distinct pathways involved in nucleic acid enters monocytes in SLE, and identify a potential therapeutic to prevent nucleic acid internalization in SLE.
Collapse
Affiliation(s)
- Amit Porat
- Elmezzi Graduate School for Molecular Medicine, Manhasset, NY, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Eitan Giat
- Elmezzi Graduate School for Molecular Medicine, Manhasset, NY, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Czeslawa Kowal
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Mingzhu He
- Center for Molecular Innovation, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Myoungsun Son
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Eicke Latz
- Biomedical Centre (BMZ), Institute of Innate Immunity, 1G007 University Hospital, University of Bonn, Bonn, Germany
| | - Ilan Ben-Zvi
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Yousef Al-Abed
- Center for Molecular Innovation, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Betty Diamond
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
25
|
Duan Z, Fang Y, Sun Y, Luan N, Chen X, Chen M, Han Y, Yin Y, Mwangi J, Niu J, Wang K, Miao Y, Zhang Z, Lai R. Antimicrobial peptide LL-37 forms complex with bacterial DNA to facilitate blood translocation of bacterial DNA and aggravate ulcerative colitis. Sci Bull (Beijing) 2018; 63:1364-1375. [PMID: 36658908 DOI: 10.1016/j.scib.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/11/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023]
Abstract
Bacterial DNA (bacDNA) is frequently found in serum of patient with ulcerative colitis (UC) and Crohn's disease, even blood bacterial culture is negative. How bacDNA evades immune elimination and is translocated into blood remain unclear. Here, we showed that bacDNA avoids elimination and disables bacteria-killing function of antimicrobial peptide LL-37 (Cramp in mice) by forming complex with LL-37, which is inducible after culture with bacteria or bacterial products. Elevated LL-37-bacDNA complex was found in plasma and lesions of patients with UC. LL-37-bacDNA promoted inflammation by inducing Th1, Th2 and Th17 differentiation and activating toll-like receptor-9 (TLR9). The complex also increased paracellular permeability, which possibly combines its inflammatory effects to promote local damage and bacDNA translocation into blood. Cramp-bacDNA aggravated mouse colitis severity while interference with the complex ameliorated the disease. The study identifies that inflammatogenic bacDNA utilizes LL-37 as a vehicle for blood translocation and to evade immune elimination. Additionally, bacteria may make a milieu by releasing bacDNA to utilize and resist host antimicrobial peptides as a 'trojan horse'.
Collapse
Affiliation(s)
- Zilei Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Yaqun Fang
- Life Sciences College of Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ning Luan
- Life Sciences College of Nanjing Agricultural University, Nanjing 210095, China
| | - Xue Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Mengrou Chen
- Life Sciences College of Nanjing Agricultural University, Nanjing 210095, China
| | - Yajun Han
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Yizhu Yin
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - James Mwangi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Junkun Niu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kunhua Wang
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Zhiye Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China.
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
26
|
Nestor J, Arinuma Y, Huerta TS, Kowal C, Nasiri E, Kello N, Fujieda Y, Bialas A, Hammond T, Sriram U, Stevens B, Huerta PT, Volpe BT, Diamond B. Lupus antibodies induce behavioral changes mediated by microglia and blocked by ACE inhibitors. J Exp Med 2018; 215:2554-2566. [PMID: 30185634 PMCID: PMC6170183 DOI: 10.1084/jem.20180776] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/06/2018] [Accepted: 08/02/2018] [Indexed: 02/03/2023] Open
Abstract
Cognitive impairment occurs in 40-90% of patients with systemic lupus erythematosus (SLE), which is characterized by autoantibodies to nuclear antigens, especially DNA. We discovered that a subset of anti-DNA antibodies, termed DNRAbs, cross reacts with the N-methyl-d-aspartate receptor (NMDAR) and enhances NMDAR signaling. In patients, DNRAb presence associates with spatial memory impairment. In a mouse model, DNRAb-mediated brain pathology proceeds through an acute phase of excitotoxic neuron loss, followed by persistent alteration in neuronal integrity and spatial memory impairment. The latter pathology becomes evident only after DNRAbs are no longer detectable in the brain. Here we investigate the mechanism of long-term neuronal dysfunction mediated by transient exposure to antibody. We show that activated microglia and C1q are critical mediators of neuronal damage. We further show that centrally acting inhibitors of angiotensin-converting enzyme (ACE) can prevent microglial activation and preserve neuronal function and cognitive performance. Thus, ACE inhibition represents a strong candidate for clinical trials aimed at mitigating cognitive dysfunction.
Collapse
Affiliation(s)
- Jacquelyn Nestor
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Yoshiyuki Arinuma
- Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tomás S Huerta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Czeslawa Kowal
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Elham Nasiri
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Nina Kello
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Yuichiro Fujieda
- Department of Rheumatology, Endocrinology and Nephrology Faculty of Medicine and Graduate School of Medicine Hokkaido University, Sapporo, Japan
| | - Alison Bialas
- Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Tim Hammond
- Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Beth Stevens
- Kirby Neurobiology Center Boston Children's Hospital, Boston, MA
| | - Patricio T Huerta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Bruce T Volpe
- Center for Biomedical Sciences, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Betty Diamond
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| |
Collapse
|
27
|
Pozsgay J, Szekanecz Z, Sármay G. Antigen-specific immunotherapies in rheumatic diseases. Nat Rev Rheumatol 2017; 13:525-537. [DOI: 10.1038/nrrheum.2017.107] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Zong S, Hoffmann C, Mané-Damas M, Molenaar P, Losen M, Martinez-Martinez P. Neuronal Surface Autoantibodies in Neuropsychiatric Disorders: Are There Implications for Depression? Front Immunol 2017; 8:752. [PMID: 28725222 PMCID: PMC5497139 DOI: 10.3389/fimmu.2017.00752] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/13/2017] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases are affecting around 7.6-9.4% of the general population. A number of central nervous system disorders, including encephalitis and severe psychiatric disorders, have been demonstrated to associate with specific neuronal surface autoantibodies (NSAbs). It has become clear that specific autoantibodies targeting neuronal surface antigens and ion channels could cause severe mental disturbances. A number of studies have focused or are currently investigating the presence of autoantibodies in specific mental conditions such as schizophrenia and bipolar disorders. However, less is known about other conditions such as depression. Depression is a psychiatric disorder with complex etiology and pathogenesis. The diagnosis criteria of depression are largely based on symptoms but not on the origin of the disease. The question which arises is whether in a subgroup of patients with depression, the symptoms might be caused by autoantibodies targeting membrane-associated antigens. Here, we describe how autoantibodies targeting membrane proteins and ion channels cause pathological effects. We discuss the physiology of these antigens and their role in relation to depression. Finally, we summarize a number of studies detecting NSAbs with a special focus on cohorts that include depression diagnosis and/or show depressive symptoms.
Collapse
Affiliation(s)
- Shenghua Zong
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Carolin Hoffmann
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Marina Mané-Damas
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Peter Molenaar
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Mario Losen
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Pilar Martinez-Martinez
- Division Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
29
|
HoHsieh A, Wang CM, Wu YJJ, Chen A, Chang MI, Chen JY. B cell epitope of human cytomegalovirus phosphoprotein 65 (HCMV pp65) induced anti-dsDNA antibody in BALB/c mice. Arthritis Res Ther 2017; 19:65. [PMID: 28320458 PMCID: PMC5359867 DOI: 10.1186/s13075-017-1268-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/16/2017] [Indexed: 11/10/2022] Open
Abstract
Background HCMV phosphoprotein 65 (HCMVpp65) is a putative immunogen that acts as an accelerator, inducing autoantibody and exacerbating autoimmune response in susceptible animals. The immunity to pp65336-439 instigates autoimmunity, suggesting that pp65336-439 contains crucial B cell epitope(s) for the development of nephritis. This study narrowed down the target epitope to pp65422-439 for immunization of BALB/c mice and mapping of B cell epitope. Methods The target epitope pp65422-439 reactivity and B cell epitope mapping was examined in serum from pp65422-439-immunized mice and patients with systemic lupus erythematosus (SLE). Kidney tissue from immunized mice was examined for signs of immune complex nephritis. Results Anti-pp65422-439 antibody in serum either from patients with SLE or from pp65422-439-immunized mice exhibited cross-reactivity to several nuclear components such as double-stranded DNA (dsDNA). Moreover, the pp65422-439-immunized mice developed initial signs of glomerulonephritis such as deposition of immunoglobulin G/M (IgG/IgM) and third complement component (C3). With B cell epitope mapping by pp65422-439-derived decapeptides, one dominant epitope, pp65428-437, was identified in serum from pp65422-439-immunized mice and patients with SLE with anti-pp65422-439 antibody. Epitope spreading from pp65428-437 to pp65430-439 was found in pp65422-439-immunized mice in which we generated monoclonal antibodies to pp65425-434 and pp65430-439. However, dsDNA positive reactivity was exclusively observed in Crithidia luciliae stains with pp65430-439-reactive monoclonal antibody. Additionally, we observed the amelioration of autoimmunity following the elevation of IgM targeting pp65428-437. Conclusions Our data suggest that pp65428-437 may be an autoimmune or lupus-prone B cell epitope and may catalyze further epitope spreading for inducing autoantibodies in lupus-susceptible individuals. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1268-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ao HoHsieh
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China
| | - Chin Man Wang
- Department of Rehabilitation, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, Republic of China
| | - Yeong-Jian Jan Wu
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin St. Kwei-Shan, Taoyuan, 33375, Taiwan, Republic of China
| | - Albert Chen
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Ming-I Chang
- Biologics, Ruen Huei Biopharmaceuticals, 1F, No.16-1, Ln. 119, Sec. 1, Roosevelt Rd., Jhongjheng Dist., Taipei, Taiwan, Republic of China
| | - Ji-Yih Chen
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin St. Kwei-Shan, Taoyuan, 33375, Taiwan, Republic of China.
| |
Collapse
|
30
|
Rekvig OP, Thiyagarajan D, Pedersen HL, Horvei KD, Seredkina N. Future Perspectives on Pathogenesis of Lupus Nephritis: Facts, Problems, and Potential Causal Therapy Modalities. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2772-2782. [PMID: 27664472 DOI: 10.1016/j.ajpath.2016.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/30/2016] [Indexed: 12/22/2022]
Abstract
Divergent incommensurable models have been developed to explain the pathogenesis of lupus nephritis. Most contemporary models favor a central role for anti-chromatin antibodies. How they exert their pathogenic effect has, however, endorsed conflicts that at least for now preclude insight into definitive pathogenic pathways. The following paradigms are contemporarily in conflict with each other: i) the impact of anti-double-stranded DNA (dsDNA) antibodies that cross-react with inherent renal antigens, ii) the impact of anti-dsDNA antibodies targeting exposed chromatin in glomeruli, and iii) the impact of relative antibody avidity for dsDNA, chromatin fragments, or cross-reacting antigens. Aside from these three themes, the pathogenic role of T cells in lupus nephritis is not clear. These different models should be tested through a collaboration between scientists belonging to the different paradigms. If it turns out that there are different pathogenic pathways in lupus nephritis, the emerging pathogenic mechanism(s) may be encountered with new individual causal therapy modalities. Today, therapy is still unspecific and far from interfering with the cause(s) of the disorder. This review attempts to describe what we know about processes that may cause lupus nephritis and how such basic processes may be affected if we can specifically interrupt them. Secondary inflammatory mechanisms, cytokine signatures, activation of complement, and other contributors to inflammation will not be discussed herein; rather, the events that trigger these factors will be discussed.
Collapse
Affiliation(s)
- Ole P Rekvig
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway; Department of Radiology, University Hospital of North Norway, Tromsø, Norway; Norwegian Center for Molecular Medicine, University of Oslo, Oslo, Norway.
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Hege L Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Kjersti D Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Natalya Seredkina
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
31
|
Yadav P, Carr MT, Yu R, Mumbey-Wafula A, Spatz LA. Mapping an epitope in EBNA-1 that is recognized by monoclonal antibodies to EBNA-1 that cross-react with dsDNA. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:362-75. [PMID: 27621818 PMCID: PMC5004290 DOI: 10.1002/iid3.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 01/19/2023]
Abstract
Introduction The Epstein Barr Virus (EBV) has been associated with the autoimmune disease, Systemic Lupus Erythematosus (SLE). EBV nuclear antigen‐I (EBNA‐1) is the major nuclear protein of EBV. We previously generated an IgG monoclonal antibody (MAb) to EBNA‐1, 3D4, and demonstrated that it cross‐reacts with double stranded DNA (dsDNA) and binds the 148 amino acid viral binding site (VBS) in the carboxyl region of EBNA‐1. The aim of the present study was to characterize another antibody to EBNA‐1 that cross‐reacts with dsDNA, compare its immunoglobulin genes to 3D4, and finely map the epitope in EBNA‐1 that is recognized by these cross‐reactive antibodies. Methods We generated an IgM MAb to EBNA‐1, 16D2, from EBNA‐1 injected mice and demonstrated by ELISA that it cross‐reacts with dsDNA and binds the 148 amino acid VBS. We sequenced the variable heavy and light chain genes of 3D4 and 16D2 and compared V gene usage. To more finely map the epitope in EBNA‐1 recognized by these MAbs, we examined their binding by ELISA to 15 overlapping peptides spanning the 148 amino acid domain. Results Sequence analysis revealed that 3D4 and 16D2 utilize different VH and VL genes but identical JH and Jk regions with minimal junctional diversity. This accounts for similarities in their CDR3 regions and may explain their similar dual binding specificity. Epitope mapping revealed 3D4 and 16D2 bind the same peptide in the VBS. Based on the crystal structure of EBNA‐1, we observed that this peptide resides at the base of an exposed proline rich loop in EBNA‐1. Conclusion We have demonstrated that two MAbs that bind EBNA‐1 and cross‐react with dsDNA, recognize the same peptide in the VBS. This peptide may serve as a mimetope for dsDNA and may be of diagnostic and therapeutic value in SLE.
Collapse
Affiliation(s)
- Pragya Yadav
- Department of ChemistryCity College of New York160 Convent AvenueNew YorkNew York10031; Graduate Program in BiochemistryGraduate Center of the City University of New York160 Convent AvenueNew YorkNew York10031
| | - Matthew T Carr
- Department of ChemistryCity College of New York160 Convent AvenueNew YorkNew York10031; Graduate Program in BiochemistryGraduate Center of the City University of New York160 Convent AvenueNew YorkNew York10031
| | - Ruby Yu
- Department of Biology City College of New York 160 Convent Avenue New York New York 10031
| | - Alice Mumbey-Wafula
- Department of Pathobiology, Sophie Davis School of Biomedical Education City College of New York 160 Convent Avenue New York New York 10031
| | - Linda A Spatz
- Department of Pathobiology, Sophie Davis School of Biomedical Education City College of New York 160 Convent Avenue New York New York 10031
| |
Collapse
|
32
|
Ding M, Zhang J. Epitope spreading induced by immunization with synthetic SSB peptides. Exp Ther Med 2016; 12:147-150. [PMID: 27347030 PMCID: PMC4906616 DOI: 10.3892/etm.2016.3267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/18/2016] [Indexed: 11/25/2022] Open
Abstract
Sjogren's syndrome type B (SSB)/La antibody is an autoantibody generally observed in connective tissue diseases whereas double-stranded deoxyribonucleic acid (dsDNA) antibodies are the most characteristic autoantibodies found in systemic lupus erythematosus (SLE) patients. The relationship of these autoantibodies remains unclear. The aim of the study was to determine the profile of antibody production in rabbits immunized with synthetic SSB peptides alone or with dsDNA. For this purpose, 214–225aa peptide of SSB antigen was synthesized based on the organic chemistry solid-phase peptide synthesis. Rabbits were immunized with the following antigens: i) synthetic SSB peptides linked with keyhole limpet hemocyanin (KLH); ii) dsDNA; iii) SSB plus dsDNA; iv) KLH; and v) phosphate-buffered saline. SSB peptide antibody was measured using the enzyme-linked immunosorbent assay while extractable nuclear antigens (ENA) antibody and dsDNA antibody were measured by immunoblotting and immunofluorescence, respectively. The results showed that a specific anti-SSB peptide antibody was produced following immunization with SSB epitope alone or with dsDNA. The SSB peptide antibody titer in the coimmunization group was higher than that of the SSB alone group. In addition, antibodies against ribonucleoprotein (RNP), Smith and/or dsDNA were detected in rabbits of the coimmunization group. The presence of anti-dsDNA antibodies in the rabbits immunized with SSB peptide suggested the induction of epitope spreading. In conclusions, SSB antibodies were produced in rabbits immunized with SSB peptide or SSB+dsDNA, whereas SSB antibody titers were higher in the coimmunization group. Furthermore, coimmunization was associated with epitope spreading.
Collapse
Affiliation(s)
- Min Ding
- Department of Dermatology, Peking University People's Hospital, Beijing 100044, P.R. China; Department of Dermatology, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
33
|
Rubinstein TB, Putterman C, Goilav B. Biomarkers for CNS involvement in pediatric lupus. Biomark Med 2016; 9:545-58. [PMID: 26079959 DOI: 10.2217/bmm.15.26] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
CNS disease, or central neuropsychiatric lupus erythematosus (cNPSLE), occurs frequently in pediatric lupus, leading to significant morbidity and poor long-term outcomes. Diagnosing cNPSLE is especially difficult in pediatrics; many current diagnostic tools are invasive and/or costly, and there are no current accepted screening mechanisms. The most complicated aspect of diagnosis is differentiating primary disease from other etiologies; research to discover new biomarkers is attempting to address this dilemma. With many mechanisms involved in the pathogenesis of cNPSLE, biomarker profiles across several modalities (molecular, psychometric and neuroimaging) will need to be used. For the care of children with lupus, the challenge will be to develop biomarkers that are accessible by noninvasive measures and reliable in a pediatric population.
Collapse
Affiliation(s)
- Tamar B Rubinstein
- Department of Pediatrics, Division of Rheumatology, The Children's Hospital at Montefiore, Albert Einstein College of Medicine, 3415 Bainbridge Avenue, Bronx, NY 10467, USA
| | - Chaim Putterman
- Department of Medicine, Division of Rheumatology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Beatrice Goilav
- Department of Pediatrics, Division of Nephrology, The Children's Hospital at Montefiore, Albert Einstein College of Medicine, 3415 Bainbridge Avenue, Bronx, NY 10467, USA
| |
Collapse
|
34
|
Dema B, Charles N. Autoantibodies in SLE: Specificities, Isotypes and Receptors. Antibodies (Basel) 2016; 5:antib5010002. [PMID: 31557984 PMCID: PMC6698872 DOI: 10.3390/antib5010002] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/09/2015] [Accepted: 12/11/2015] [Indexed: 12/23/2022] Open
Abstract
Systemic Lupus Erythematosus (SLE) is characterized by a wide spectrum of auto-antibodies which recognize several cellular components. The production of these self-reactive antibodies fluctuates during the course of the disease and the involvement of different antibody-secreting cell populations are considered highly relevant for the disease pathogenesis. These cells are developed and stimulated through different ways leading to the secretion of a variety of isotypes, affinities and idiotypes. Each of them has a particular mechanism of action binding to a specific antigen and recognized by distinct receptors. The effector responses triggered lead to a chronic tissue inflammation. DsDNA autoantibodies are the most studied as well as the first in being characterized for its pathogenic role in Lupus nephritis. However, others are of growing interest since they have been associated with other organ-specific damage, such as anti-NMDAR antibodies in neuropsychiatric clinical manifestations or anti-β2GP1 antibodies in vascular symptomatology. In this review, we describe the different auto-antibodies reported to be involved in SLE. How autoantibody isotypes and affinity-binding to their antigen might result in different pathogenic responses is also discussed.
Collapse
Affiliation(s)
- Barbara Dema
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Paris 75018, France.
| | - Nicolas Charles
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Paris 75018, France.
| |
Collapse
|
35
|
Xia Y, Eryilmaz E, Der E, Pawar RD, Guo X, Cowburn D, Putterman C. A peptide mimic blocks the cross-reaction of anti-DNA antibodies with glomerular antigens. Clin Exp Immunol 2015; 183:369-79. [PMID: 26482679 DOI: 10.1111/cei.12734] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2015] [Indexed: 12/29/2022] Open
Abstract
Anti-DNA antibodies play a pivotal role in the pathogenesis of lupus nephritis by cross-reacting with renal antigens. Previously, we demonstrated that the binding affinity of anti-DNA antibodies to self-antigens is isotype-dependent. Furthermore, significant variability in renal pathogenicity was seen among a panel of anti-DNA isotypes [derived from a single murine immunoglobulin (Ig)G3 monoclonal antibody, PL9-11] that share identical variable regions. In this study, we sought to select peptide mimics that effectively inhibit the binding of all murine and human anti-DNA IgG isotypes to glomerular antigens. The PL9-11 panel of IgG anti-DNA antibodies (IgG1, IgG2a, IgG2b and IgG3) was used for screening a 12-mer phage display library. Binding affinity was determined by surface plasmon resonance. Enzyme-linked immunosorbent assay (ELISA), flow cytometry and glomerular binding assays were used for the assessment of peptide inhibition of antibody binding to nuclear and kidney antigens. We identified a 12 amino acid peptide (ALWPPNLHAWVP, or 'ALW') which binds to all PL9-11 IgG isotypes. Preincubation with the ALW peptide reduced the binding of the PL9-11 anti-DNA antibodies to DNA, laminin, mesangial cells and isolated glomeruli significantly. Furthermore, we confirmed the specificity of the amino acid sequence in the binding of ALW to anti-DNA antibodies by alanine scanning. Finally, ALW inhibited the binding of murine and human lupus sera to dsDNA and glomeruli significantly. In conclusion, by inhibiting the binding of polyclonal anti-DNA antibodies to autoantigens in vivo, the ALW peptide (or its derivatives) may potentially be a useful approach to block anti-DNA antibody binding to renal tissue.
Collapse
Affiliation(s)
- Y Xia
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E Eryilmaz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E Der
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R D Pawar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - X Guo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - D Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - C Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
36
|
Gregersen PK, Klein G, Keogh M, Kern M, DeFranco M, Simpfendorfer KR, Kim SJ, Diamond B. The Genotype and Phenotype (GaP) registry: a living biobank for the analysis of quantitative traits. Immunol Res 2015; 63:107-12. [PMID: 26467974 DOI: 10.1007/s12026-015-8711-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We describe the development of the Genotype and Phenotype (GaP) Registry, a living biobank of normal volunteers who are genotyped for genetic markers related to human disease. Participants in the GaP can be recalled for hypothesis driven study of disease associated genetic variants. The GaP has facilitated functional studies of several autoimmune disease associated loci including Csk, Blk, PDRM1 (Blimp-1) and PTPN22. It is likely that expansion of such living biobank registries will play an important role in studying and understanding the function of disease associated alleles in complex disease.
Collapse
Affiliation(s)
- Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Gila Klein
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Mary Keogh
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Marlena Kern
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Margaret DeFranco
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kim R Simpfendorfer
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| |
Collapse
|
37
|
Abstract
The inclusion of 'the anti-DNA antibody' by the ACR and the Systemic Lupus International Collaborating Clinics (SLICC) as a criterion for systemic lupus erythematosus does not convey the diverse origins of these antibodies, whether their production is transient or persistent (which is heavily influenced by the nature of the inducing antigens), the specificities exerted by these antibodies or their clinical impact-or lack thereof. A substantial amount of data not considered in clinical medicine could be added from basic immunology evidence, which could change the paradigms linked to what 'the anti-DNA antibody' is, in a pathogenic, classification or diagnostic context.
Collapse
|
38
|
Pawar RD, Goilav B, Xia Y, Zhuang H, Herlitz L, Reeves WH, Putterman C. Serum autoantibodies in pristane induced lupus are regulated by neutrophil gelatinase associated lipocalin. Clin Immunol 2014; 154:49-65. [PMID: 24971701 PMCID: PMC4119527 DOI: 10.1016/j.clim.2014.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/28/2022]
Abstract
The onset of autoantibodies in systemic autoimmunity can be the result of a breakdown in tolerance at multiple checkpoints. Genetic, hormonal, and immunological factors can combine with environmental influences to accelerate the onset of disease and aggravate disease outcome. Here, we describe a novel mechanism relating to the regulatory role of Neutrophil Gelatinase Associated Lipocalin (NGAL) in modulating the levels of autoantibodies in pristane induced lupus. Following a single injection of pristane intraperitoneally, NGAL expression was induced in both the serum and spleen. Furthermore, NGAL deficient mice were more susceptible to the induction of pristane stimulated autoimmunity, and displayed higher numbers of autoantibody secreting cells and increased expression of activation induced cytidine deaminase (AID) and other inflammatory mediators in the spleen. In contrast, kidney damage was milder in NGAL deficient mice, indicating that NGAL was detrimental in autoantibody mediated kidney disease. These studies indicate that NGAL plays differential roles in different tissues in the context of lupus, and suggest a previously unrecognized role for NGAL in adaptive immunity.
Collapse
Affiliation(s)
- Rahul D Pawar
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Beatrice Goilav
- The Division of Pediatric Nephrology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yumin Xia
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haoyang Zhuang
- The Division of Rheumatology & Clinical Immunology, University of Florida, Gainesville, FL 32611, USA
| | - Leal Herlitz
- The Department of Pathology, Columbia University Medical Center, NY 10032, USA
| | - Westley H Reeves
- The Division of Rheumatology & Clinical Immunology, University of Florida, Gainesville, FL 32611, USA
| | - Chaim Putterman
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
39
|
Vo A, Volpe BT, Tang CC, Schiffer WK, Kowal C, Huerta PT, Uluğ AM, Dewey SL, Eidelberg D, Diamond B. Regional brain metabolism in a murine systemic lupus erythematosus model. J Cereb Blood Flow Metab 2014; 34:1315-20. [PMID: 24824914 PMCID: PMC4126091 DOI: 10.1038/jcbfm.2014.85] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/16/2014] [Accepted: 04/22/2014] [Indexed: 01/18/2023]
Abstract
Systemic lupus erythematosus (SLE) is characterized by multiorgan inflammation, neuropsychiatric disorders (NPSLE), and anti-nuclear antibodies. We previously identified a subset of anti-DNA antibodies (DNRAb) cross-reactive with the N-methyl-D-aspartate receptor, present in 30% to 40% of patients, able to enhance excitatory post-synaptic potentials and trigger neuronal apoptosis. DNRAb+ mice exhibit memory impairment or altered fear response, depending on whether the antibody penetrates the hippocampus or amygdala. Here, we used 18F-fluorodeoxyglucose (FDG) microPET to plot changes in brain metabolism after regional blood-brain barrier (BBB) breach. In DNRAb+ mice, metabolism declined at the site of BBB breach in the first 2 weeks and increased over the next 2 weeks. In contrast, DNRAb- mice exhibited metabolic increases in these regions over the 4 weeks after the insult. Memory impairment was present in DNRAb+ animals with hippocampal BBB breach and altered fear conditioning in DNRAb+ mice with amygdala BBB breach. In DNRAb+ mice, we observed an inverse relationship between neuron number and regional metabolism, while a positive correlation was observed in DNRAb- mice. These findings suggest that local metabolic alterations in this model take place through different mechanisms with distinct time courses, with important implications for the interpretation of imaging data in SLE subjects.
Collapse
Affiliation(s)
- An Vo
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Bruce T Volpe
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Wynne K Schiffer
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Czeslawa Kowal
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Patricio T Huerta
- Laboratory of Immune & Neural Networks, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Aziz M Uluğ
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Stephen L Dewey
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| |
Collapse
|
40
|
The pathogenesis and diagnosis of systemic lupus erythematosus: still not resolved. Semin Immunopathol 2014; 36:301-11. [PMID: 24763531 DOI: 10.1007/s00281-014-0428-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/01/2014] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with various clinical manifestations affecting different tissues. A characteristic feature of SLE is the presence of autoantibodies against double-stranded (ds)DNA, histones and nucleosomes, and other chromatin components. SLE is a prototype type III hypersensitivity reaction. Local deposition of anti-nuclear antibodies in complex with released chromatin induces serious inflammatory conditions by activation of the complement system. The severe renal manifestation, lupus nephritis, is classified based on histological findings in renal biopsies. Apoptotic debris, including chromatin, is present in the extracellular matrix and circulation of patients with SLE. This may be due to an aberrant process of apoptosis and/or insufficient clearance of apoptotic cells/chromatin. The non-cleared apoptotic debris may lead to activation of both the innate and adaptive immune systems. In addition, an aberrant presentation of peptides by antigen-presenting cells, disturbed selection processes for lymphocytes, and deregulated lymphocyte responses may be involved in the development of autoimmunity. In the present review, we briefly will summarize current knowledge on the pathogenesis of SLE. We will also critically discuss and challenge central issues that need to be addressed in order to fully understand the pathogenic mechanisms involved in the development of SLE and in order to have an improved diagnosis for SLE. Disappointingly, in our opinion, there are still more questions than answers for the pathogenesis, diagnosis, and treatment of SLE.
Collapse
|
41
|
Szymula A, Rosenthal J, Szczerba BM, Bagavant H, Fu SM, Deshmukh US. T cell epitope mimicry between Sjögren's syndrome Antigen A (SSA)/Ro60 and oral, gut, skin and vaginal bacteria. Clin Immunol 2014; 152:1-9. [PMID: 24576620 DOI: 10.1016/j.clim.2014.02.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022]
Abstract
This study was undertaken to test the hypothesis that Sjogren's syndrome Antigen A (SSA)/Ro60-reactive T cells are activated by peptides originating from oral and gut bacteria. T cell hybridomas generated from HLA-DR3 transgenic mice recognized 3 regions on Ro60, with core epitopes mapped to amino acids 228-238, 246-256 and 371-381. BLAST analysis identified several mimicry peptides, originating from human oral, intestinal, skin and vaginal bacteria, as well as environmental bacteria. Amongst these, a peptide from the von Willebrand factor type A domain protein (vWFA) from the oral microbe Capnocytophaga ochracea was the most potent activator. Further, Ro60-reactive T cells were activated by recombinant vWFA protein and whole Escherichia coli expressing this protein. These results demonstrate that peptides derived from normal human microbiota can activate Ro60-reactive T cells. Thus, immune responses to commensal microbiota and opportunistic pathogens should be explored as potential triggers for initiating autoimmunity in SLE and Sjögren's syndrome.
Collapse
Affiliation(s)
- Agnieszka Szymula
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jacob Rosenthal
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Barbara M Szczerba
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Harini Bagavant
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Shu Man Fu
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Umesh S Deshmukh
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
42
|
Peterson LK, Pennington LF, Shaw LA, Brown M, Treacy EC, Friend SF, Hatlevik Ø, Rubtsova K, Rubtsov AV, Dragone LL. SLAP deficiency decreases dsDNA autoantibody production. Clin Immunol 2014; 150:201-9. [PMID: 24440645 DOI: 10.1016/j.clim.2013.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 11/18/2022]
Abstract
Src-like adaptor protein (SLAP) adapts c-Cbl, an E3 ubiquitin ligase, to activated components of the BCR signaling complex regulating BCR levels and signaling in developing B cells. Based on this function, we asked whether SLAP deficiency could decrease the threshold for tolerance and eliminate development of autoreactive B cells in two models of autoantibody production. First, we sensitized mice with a dsDNA mimetope that causes an anti-dsDNA response. Despite equivalent production of anti-peptide antibodies compared to BALB/c controls, SLAP(-/-) mice did not produce anti-dsDNA. Second, we used the 56R tolerance model. SLAP(-/-) 56R mice had decreased levels of dsDNA-reactive antibodies compared to 56R mice due to skewed light chain usage. Thus, SLAP is a critical regulator of B-cell development and function and its deficiency leads to decreased autoreactive B cells that are otherwise maintained by inefficient receptor editing or failed negative selection.
Collapse
Affiliation(s)
- Lisa K Peterson
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Luke F Pennington
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Laura A Shaw
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Meredith Brown
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Eric C Treacy
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Samantha F Friend
- Department of Pediatrics, University of Colorado Denver, 13001 E. 17th Place, Aurora, CO 80045, USA; Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Øyvind Hatlevik
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Kira Rubtsova
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Anatoly V Rubtsov
- Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Leonard L Dragone
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA; Department of Pediatrics, University of Colorado Denver, 13001 E. 17th Place, Aurora, CO 80045, USA; Department of Immunology, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA; Division of Rheumatology, Colorado Children's Hospital, 13123 E. 16th Ave., Aurora, CO 80045, USA.
| |
Collapse
|
43
|
Nikolova-Ganeva KA, Gesheva VV, Todorov TA, Voll RE, Vassilev TL. Targeted silencing of DNA-specific B cells combined with partial plasma cell depletion displays additive effects on delaying disease onset in lupus-prone mice. Clin Exp Immunol 2013; 174:221-8. [PMID: 23808414 DOI: 10.1111/cei.12164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2013] [Indexed: 11/29/2022] Open
Abstract
Targeting autoreactive B lymphocytes at any stage of their differentiation could yield viable therapeutic strategies for treating autoimmunity. All currently used drugs, including the most recently introduced biological agents, lack target specificity. Selective silencing of double-stranded DNA-specific B cells in animals with spontaneous lupus has been achieved previously by the administration of a chimeric antibody molecule that cross-links their DNA-reactive B cell immunoglobulin receptors with inhibitory FcγIIb (CD32) receptors. However, long-lived plasmacytes are resistant to this chimeric antibody as well as to all conventional treatments. Bortezomib (a proteasome inhibitor) depletes most plasma cells and has been shown recently to suppress disease activity in lupus mice. We hypothesized that the co-administration of non-toxic doses of bortezomib, that partially purge long-lived plasma cells, together with an agent that selectively silences DNA-specific B cells, should have additive effects in an autoantibody-mediated disease. Indeed, our data show that the simultaneous treatment of lupus-prone MRL/lpr mice with suboptimal doses of bortezomib plus the chimeric antibody resulted in the prevention or the delayed appearance of the disease manifestations as well as in a prolonged survival. The effect of the combination therapy was significantly stronger than that of the respective monotherapies and was comparable to that observed after cyclophosphamide administration.
Collapse
Affiliation(s)
- K A Nikolova-Ganeva
- Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | | | | |
Collapse
|
44
|
Quan J, Lakhanpal A, Reddy MM, Zaman S, Li QZ, German DC, Olsen NJ, Kodadek T, Karp DR. Discovery of biomarkers for systemic lupus erythematosus using a library of synthetic autoantigen surrogates. J Immunol Methods 2013; 402:23-34. [PMID: 24269750 DOI: 10.1016/j.jim.2013.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 12/28/2022]
Abstract
Antibodies to a wide range of self-antigens, including those directed against nucleic acids or nucleic acid-binding proteins are the essential biomarkers for diseases such as systemic lupus erythematosus (SLE). Highly complex libraries of nonamers consisting of N-substituted glycines (peptoids) were screened for compounds that bound IgG from patients with SLE and earlier, incomplete autoimmune syndromes. Peptoids were identified that could identify subjects with SLE and related syndromes with a high sensitivity (70%) and specificity (97.5%). Immobilized peptoids were used to isolate IgG from both healthy subjects and SLE patients that reacted with known RNA-binding proteins. In the case of SLE patients, the peptoid-purified IgG reacted with several autoantigens, suggesting that the peptoids are capable of interacting with multiple, structurally similar molecules. These results show that the measurement of IgG binding to peptoids can identify subjects with high levels of pathogenic autoantibodies.
Collapse
Affiliation(s)
- Jiexia Quan
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Akshai Lakhanpal
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - M Muralidhar Reddy
- The Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA; Opko Health, Inc., 555 Heritage Dr., Jupiter, FL 33458, USA
| | - Sayed Zaman
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Quan-Zhen Li
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dwight C German
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nancy J Olsen
- The Department of Medicine, Penn State M.S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Thomas Kodadek
- The Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - David R Karp
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; The Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
45
|
Franchin G, Son M, Kim SJ, Ben-Zvi I, Zhang J, Diamond B. Anti-DNA antibodies cross-react with C1q. J Autoimmun 2013; 44:34-9. [PMID: 23834843 DOI: 10.1016/j.jaut.2013.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that involves multiple organ systems and typically presents as a chronic inflammatory disease. Antibodies to double-stranded (ds) DNA are present in approximately 70% of patients and form nucleic acid containing immune complexes which activate dendritic cells through engagement of toll-like receptors, leading to a pro-inflammatory, pro-immunogenic milieu. In addition, anti-dsDNA antibodies deposit in kidneys to initiate glomerulonephritis. Antibodies to C1q have also been implicated in lupus nephritis and are found in 30-50% of patients. C1q is a known suppressor of immune activation and C1q deficiency is the strongest risk factor for SLE. We previously identified a subset of anti-DNA antibodies that binds the N-methyl-D-aspartate receptor. We now show that both mouse and human anti-DNA antibodies with this specificity bind C1q. These antibodies bind to Clq in glomeruli and exhibit decreased glomerular deposition in the absence of C1q. We propose that this subset of anti-DNA antibodies participates in lupus pathogenesis through direct targeting of C1q on glomeruli and also through removal of soluble C1q thereby limiting the ability of C1q to mediate immune homeostasis.
Collapse
Affiliation(s)
- Giovanni Franchin
- The Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Autoimmune diseases currently affect 5-7% of the world's population; in most diseases there are circulating autoantibodies. Brain-reactive antibodies are present in approximately 2-3% of the general population but do not usually contribute to brain pathology. These antibodies penetrate brain tissue only early in development or under pathologic conditions. This restriction on their pathogenicity and the lack of correlation between serum titers and brain pathology have, no doubt, contributed to a delayed appreciation of the contribution of autoantibodies in diseases of the central nervous system. Nonetheless, it is increasingly clear that antibodies can cause damage in the brain and likely initiate or aggravate multiple neurologic conditions; brain-reactive antibodies contribute to symptomatology in autoimmune disease, infectious disease, and malignancy.
Collapse
Affiliation(s)
- B Diamond
- Feinstein Institute for Medical Research, Manhasset, New York 11030, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
A large antibody repertoire is generated in developing B cells in the bone marrow. Before these B cells achieve immunocompetence, those expressing autospecificities must be purged. To that end, B cells within the bone marrow and just following egress from the bone marrow are subject to tolerance induction. Once B cells achieve immunocompetence, the antibody repertoire can be further diversified by somatic hypermutation of immunoglobulin genes in B cells that have been activated by antigen and cognate T cell help and have undergone a germinal center (GC) response. This process also leads to the generation of autoreactive B cells which must be again purged to protect the host. Thus, B cells within the GC and just following egress from the GC are also subject to tolerance induction. Available data suggest that B cell intrinsic processes triggered by signaling through the B cell receptor activate tolerance mechanisms at both time points. Recent data suggest that GC and post-GC B cells are also subject to B cell extrinsic tolerance mechanisms mediated through soluble and membrane-bound factors derived from various T cell subsets.
Collapse
|
48
|
Gesheva V, Szekeres Z, Mihaylova N, Dimitrova I, Nikolova M, Erdei A, Prechl J, Tchorbanov A. Generation of gene-engineered chimeric DNA molecules for specific therapy of autoimmune diseases. Hum Gene Ther Methods 2012; 23:357-65. [PMID: 23075110 PMCID: PMC4015069 DOI: 10.1089/hgtb.2012.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the development of self-reactive B and T cells and autoantibody production. In particular, double-stranded DNA-specific B cells play an important role in lupus progression, and their selective elimination is a reasonable approach for effective therapy of SLE. DNA-based vaccines aim at the induction of immune response against the vector-encoded antigen. Here, we are exploring, as a new DNA-based therapy of SLE, a chimeric DNA molecule encoding a DNA-mimotope peptide, and the Fv but not the immunogenic Fc fragment of an FcγRIIb-specific monoclonal antibody. This DNA construct was inserted in the expression vector pNut and used as a naked DNA vaccine in a mouse model of lupus. The chimeric DNA molecule can be expressed in eukaryotic cells and cross-links cell surface receptors on DNA-specific B cells, delivering an inhibitory intracellular signal. Intramuscular administration of the recombinant DNA molecule to lupus-prone MRL/lpr mice prevented increase in IgG anti-DNA antibodies and was associated with a low degree of proteinuria, modulation of cytokine profile, and suppression of lupus nephritis.
Collapse
Affiliation(s)
- Vera Gesheva
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Eilat D, Wabl M. B cell tolerance and positive selection in lupus. THE JOURNAL OF IMMUNOLOGY 2012; 189:503-9. [PMID: 22773662 DOI: 10.4049/jimmunol.1200848] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus is considered a prototype of systemic autoimmune diseases; however, despite considerable advances in recent years in the understanding of basic mechanisms in immunology, little progress has been made in elucidating the etiology and pathogenesis of this disease. This even holds for inbred mice, such as the lupus-prone New Zealand Black/New Zealand White F(1) mice, which are all genetically programmed to develop lupus at a predetermined age. This frustrating state of affairs calls for a fundamental change in our scientific thinking and the opening of new directions in lupus research. In this study, we suggest that intrinsic B cell tolerance mechanisms are not grossly impaired in lupus-prone mice, but that an unusually strong positive selection event recruits a small number of autoreactive B cells to the germinal centers. This event could be facilitated by nucleic acid-protein complexes that are created by somatic changes in the susceptible animal.
Collapse
Affiliation(s)
- Dan Eilat
- Department of Medicine, Hadassah University Hospital, Hebrew University Faculty of Medicine, Jerusalem, Israel
| | | |
Collapse
|
50
|
Wang L, Zhou D, Lee J, Niu H, Faust TW, Frattini S, Kowal C, Huerta PT, Volpe BT, Diamond B. Female mouse fetal loss mediated by maternal autoantibody. ACTA ACUST UNITED AC 2012; 209:1083-9. [PMID: 22565825 PMCID: PMC3371726 DOI: 10.1084/jem.20111986] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Systemic lupus erythematosus (SLE), a disease of women during childbearing years, is characterized by the production of double-stranded DNA antibodies. A subset of these antibodies, present in 40% of patients, cross-reacts with the NR2A and NR2B subunits of the N-methyl-d-aspartate receptor (NMDAR). In this study, we show that, in mouse models, these antibodies cause a loss of female fetus viability by inducing apoptosis of NR2A-expressing neurons within the brainstem late in fetal development; gender specificity derives from a time-dependent increased expression of NR2A in female brainstem or increased vulnerability of female fetal neurons to signaling through NR2A-containing NMDARs. This paradigm is consistent with available data on the sex ratio of live births of women with SLE. It represents a novel mechanism by which maternal autoantibodies can severely affect fetal health in a gender-specific fashion and raises the question of how many maternal antibodies affect brain development or exhibit gender-specific fetal effects.
Collapse
Affiliation(s)
- Li Wang
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, NorthShore–Long Island Jewish Health System, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|