1
|
Rekvig OP. Why is it so difficult to understand why we don't understand human systemic lupus erythematosus? Contemplating facts, conflicts, and impact of "the causality cascade paradigm". Front Immunol 2025; 15:1507792. [PMID: 39936150 PMCID: PMC11811100 DOI: 10.3389/fimmu.2024.1507792] [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: 10/08/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
In attempts to understand systemic lupus erythematosus (SLE), we find ourselves in the intellectual cross-point between nosology, pathogenicity-oriented science, philosophy, empiricism, and qualified conjectures. A vital consequence in science theory is that scientific hypotheses that are not critically investigated are in danger of being transformed into scientific dogmas. This statement has consequences for this study. Two central problematic aspects are discussed. For the first, we have to consider new selection principles for classification criteria-implying integration of the causality principle. Second, central historical data must be implemented if we aim to understand SLE. These data comprise famous descriptions of distinct, dynamically changing DNA structures linked to the genetic machinery. These unique structures have since their discoveries decades ago mostly been ignored in SLE research. Likewise, inconclusive dogmatic data indicate that different glomerular ligands are recognized by nephritogenic anti-dsDNA antibodies-exposed chromatin fragments or inherent membrane ligands. These incongruent models have not been comparatively and systematically investigated. Three research areas will be critically discussed: (i) selection and role of SLE classification criteria, a process that must imply the causality principle; (ii) definition and impact of anti-dsDNA structure-specific antibodies; (iii) incongruent pathogenic models that account for lupus nephritis. A precise and critically important question is if SLE itself is a response to a dominant unified cause that initiates a cascade of downstream effects (criteria) or if SLE represents combined responses to a random interplay of multiple cause-effect events. These principally different explanations are formally not excluded or accepted today. Currently, SLE may be regarded as a disease with phenotypic diversity, independently segregated manifestations with unresolved etiologies that are not unique to a single SLE phenotype. The focus for the present discussion is basically how we, by critical hypotheses, can re-consider science-based selection of SLE classification criteria in order to delimitate and rationalize SLE. Classification criteria, autoimmunity, DNA structures, and anti-dsDNA antibodies are integrated aspects in this discussion.
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
|
2
|
Sener S, Batu ED, Unal D, Basaran O, Saribas Z, Bilginer Y, Sener B, Ozen S. Examination of antinuclear antibody staining patterns and titers in patients with childhood-onset systemic lupus erythematosus. Lupus 2025; 34:71-78. [PMID: 39603714 DOI: 10.1177/09612033241305198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
OBJECTIVE Antinuclear antibodies (ANA) staining patterns can provide useful information in systemic lupus erythematosus (SLE). In our study, we examined the frequency of ANA staining patterns in disease-related features in childhood-onset SLE patients. METHODS ANA and its staining patterns were assessed in childhood-onset SLE patients. RESULTS Two hundred twenty-three patients were included (F/M = 3/1). Their median age at diagnosis was 14.3 (11.9-16.1) years. The anti-cell (AC)-4/5 (fine or large speckled) pattern was the most common nuclear ANA pattern (75.8%), while the AC-19 (dense fine speckled) pattern was the most frequently detected cytoplasmic ANA pattern (13.1%). The AC-4/5 (fine or large speckled) patterns were notably seen in fever, acute and chronic cutaneous lupus, arthritis, serositis, hematologic involvement, renal involvement, neuropsychiatric involvement, gastrointestinal involvement, and cardiopulmonary involvement (all p < .001). Conversely, the AC-1 (homogeneous) pattern was significantly detected in oral/nasal ulcers and non-scarring alopecia (both p < .001). Regarding the laboratory features, the AC-4/5 (fine or large speckled) patterns exhibited a predominant seen in autoimmune hemolytic anemia, leukopenia, thrombocytopenia, elevated ESR and CRP, hypocomplementemia, direct Coombs, anti-Smith (Sm), anti-SSA and SS-B, anti-ribonucleoprotein (RNP), anti-histone, anti-ribosomal P, lupus anticoagulant, anti-cardiolipin immunoglobulin (Ig)M/IgG, and anti-β2-glycoprotein IgM/IgG positivities (all p < .001). In contrast, the AC-1 (homogeneous) pattern was detected in anti-double-stranded (ds) DNA and anti-histone positivity (both p < .001). CONCLUSION Our study showed that AC-4/5 and AC-1 patterns of ANA are frequently detected in many clinical and serological features of childhood-onset SLE patients. However, further studies are needed in larger populations to verify these results.
Collapse
Affiliation(s)
- Seher Sener
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Ezgi Deniz Batu
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Dilara Unal
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Ozge Basaran
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Zeynep Saribas
- Department of Medical Microbiology, Hacettepe University, Ankara, Turkey
| | - Yelda Bilginer
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Burcin Sener
- Department of Medical Microbiology, Hacettepe University, Ankara, Turkey
| | - Seza Ozen
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
3
|
Spencer J, Jain S. Could tolerance to DNA be broken in the gut in systemic lupus erythematosus? Immunol Lett 2024; 270:106937. [PMID: 39490628 DOI: 10.1016/j.imlet.2024.106937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/14/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The bacteria in the human colon outnumber the total number of nucleated cells in the human body by approximately 10:1. The DNA that the bacteria contain is enriched around 20-fold in immune stimulatory CpG motifs compared to the DNA of host cells. In addition, this DNA can have alternative more immunogeneic DNA structures and it may be presented to the immune system alongside other proinflammatory bacterial innate ligands such as LPS. To ensure that this immunostimulatory combination is not pathogenic, the luminal boundary of host tissues in the human gastrointestinal tract is protected by cells secreting bactericides together with the secreted enzyme DNASE1L3 that can break down bacterial DNA. Cells with RNA encoding DNASE1L3 are particularly abundant in the gut-associated lymphoid tissue where bacteria are specifically sampled into the body, alongside B cells noted for their T independent function. Importantly, individuals with loss of function mutations in DNASE1L3 develop anti-DNA antibodies and lupus symptoms. In this review, we explore the possibility that a perfect storm might break tolerance to DNA: when bacterial DNA from microbiota that is not digested by DNASE1L3 directly encounters B cells that are not necessarily restricted by T cell dependence.
Collapse
Affiliation(s)
- Jo Spencer
- School of Immunology and Microbial Sciences, King's College London, London, UK.
| | - Sahil Jain
- School of Immunology and Microbial Sciences, King's College London, London, UK; Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
4
|
Stabach PR, Sims D, Gomez-Bañuelos E, Zehentmeier S, Dammen-Brower K, Bernhisel A, Kujawski S, Lopez SG, Petri M, Goldman DW, Lester ER, Le Q, Ishaq T, Kim H, Srivastava S, Kumar D, Pereira JP, Yarema KJ, Koumpouras F, Andrade F, Braddock DT. A dual-acting DNASE1/DNASE1L3 biologic prevents autoimmunity and death in genetic and induced lupus models. JCI Insight 2024; 9:e177003. [PMID: 38888971 PMCID: PMC11383374 DOI: 10.1172/jci.insight.177003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
A defining feature of systemic lupus erythematosus (SLE) is loss of tolerance to self-DNA, and deficiency of DNASE1L3, the main enzyme responsible for chromatin degradation in blood, is also associated with SLE. This association can be found in an ultrarare population of pediatric patients with DNASE1L3 deficiency who develop SLE, adult patients with loss-of-function variants of DNASE1L3 who are at a higher risk for SLE, and patients with sporadic SLE who have neutralizing autoantibodies against DNASE1L3. To mitigate the pathogenic effects of inherited and acquired DNASE1L3 deficiencies, we engineered a long-acting enzyme biologic with dual DNASE1/DNASE1L3 activity that is resistant to DNASE1 and DNASE1L3 inhibitors. Notably, we found that the biologic prevented the development of lupus in Dnase1-/-Dnase1L3-/- double-knockout mice and rescued animals from death in pristane-induced lupus. Finally, we confirmed that the human isoform of the enzyme biologic was not recognized by autoantibodies in SLE and efficiently degraded genomic and mitochondrial cell-free DNA, as well as microparticle DNA, in SLE plasma. Our findings suggest that autoimmune diseases characterized by aberrant DNA accumulation, such as SLE, can be effectively treated with a replacement DNASE tailored to bypass pathogenic mechanisms, both genetic and acquired, that restrict DNASE1L3 activity.
Collapse
Affiliation(s)
- Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dominique Sims
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Eduardo Gomez-Bañuelos
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kris Dammen-Brower
- Translational Tissue Engineering Center and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Andrew Bernhisel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sophia Kujawski
- Department of Rheumatology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sam G. Lopez
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel W. Goldman
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ethan R. Lester
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Quan Le
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tayyaba Ishaq
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hana Kim
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Shivani Srivastava
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Deepika Kumar
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joao P. Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kevin J. Yarema
- Translational Tissue Engineering Center and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fotios Koumpouras
- Department of Rheumatology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Felipe Andrade
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Pisetsky DS, Herbert A. The role of DNA in the pathogenesis of SLE: DNA as a molecular chameleon. Ann Rheum Dis 2024; 83:830-837. [PMID: 38749573 PMCID: PMC11168871 DOI: 10.1136/ard-2023-225266] [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: 11/10/2023] [Accepted: 04/11/2024] [Indexed: 06/14/2024]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterised by antibodies to DNA (anti-DNA) and other nuclear macromolecules. Anti-DNA antibodies are markers for classification and disease activity and promote pathogenesis by forming immune complexes that deposit in the tissue or stimulate cytokine production. Studies on the antibody response to DNA have focused primarily on a conformation of DNA known as B-DNA, the classic right-handed double helix. Among other conformations of DNA, Z-DNA is a left-handed helix with a zig-zag backbone; hence, the term Z-DNA. Z-DNA formation is favoured by certain base sequences, with the energetically unfavourable flip from B-DNA to Z-DNA dependent on conditions. Z-DNA differs from B-DNA in its immunogenicity in animal models. Furthermore, anti-Z-DNA antibodies, but not anti-B-DNA antibodies, can be present in otherwise healthy individuals. In SLE, antibodies to Z-DNA can occur in association with antibodies to B-DNA as a cross-reactive response, rising and falling together. While formed transiently in chromosomal DNA, Z-DNA is stably present in bacterial biofilms; biofilms can provide protection against antibiotics and other challenges including elements of host defence. The high GC content of certain bacterial DNA also favours Z-DNA formation as do DNA-binding proteins of bacterial or host origin. Together, these findings suggest that sources of Z-DNA can enhance the immunogenicity of DNA and, in SLE, stimulate the production of cross-reactive antibodies that bind both B-DNA and Z-DNA. As such, DNA can act as a molecular chameleon that, when stabilised in the Z-DNA conformation, can drive autoimmunity.
Collapse
Affiliation(s)
- David S Pisetsky
- Duke University Medical Center, Durham, North Carolina, USA
- Medical Research, Durham VA Health Care System, Durham, North Carolina, USA
| | - Alan Herbert
- InsideOutBio Inc, Charlestown, Massachusetts, USA
| |
Collapse
|
6
|
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
|
7
|
Bruschi M, Angeletti A, Prunotto M, Meroni PL, Ghiggeri GM, Moroni G, Sinico RA, Franceschini F, Fredi M, Vaglio A, Cavalli A, Scapozza L, Patel JJ, Tan JC, Lo KC, Cavagna L, Petretto A, Pratesi F, Migliorini P, Locatelli F, Pazzola G, Pesce G, Giannese D, Manfredi A, Ramirez GA, Esposito P, Murdaca G, Negrini S, Bui F, Trezzi B, Emmi G, Cavazzana I, Binda V, Fenaroli P, Pisan I, Montecucco C, Santoro D, Scolari F, Mescia F, Volpi S, Mosca M, Tincani A, Ravelli A, Murtas C, Candiano G, Caridi G, La Porta E, Verrina E. A critical view on autoantibodies in lupus nephritis: Concrete knowledge based on evidence. Autoimmun Rev 2024; 23:103535. [PMID: 38552995 DOI: 10.1016/j.autrev.2024.103535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/07/2024]
Abstract
Deposition of autoantibodies in glomeruli is a key factor in the development of lupus nephritis (LN). For a long time, anti-dsDNA and anti-C1q antibodies were thought to be the main cause of the kidney damage. However, recent studies have shown that the list of autoantibidies that have renal tropism and deposit in the kidney in LN is increasing and the link between anti-dsDNA and renal pathology is weak due to potential confounders. Aspecific bindings of dsDNA with cationic antibodies and of anti-dsDNA with several renal antigens such as actinin, laminin, entactin, and annexinA2 raised doubts about the specific target of these antibodies in the kidney. Moreover, the isotype of anti-dsDNA in SLE and LN has never received adequate interest until the recent observation that IgG2 are preponderant over IgG1, IgG3 and IgG4. Based on the above background, recent studies investigated the involvement of anti-dsDNA IgG2 and of other antibodies in LN. It was concluded that circulating anti-dsDNA IgG2 levels do not distinguish between LN versus non-renal SLE, and, in patients with LN, their levels do not change over time. Circulating levels of other antibodies such as anti-ENO1 and anti-H2 IgG2 were, instead, higher in LN vs non-renal SLE at the time of diagnosis and decreased following therapies. Finally, new classes of renal antibodies that potentially modify the anti-inflammatory response in the kidney are emerging as new co-actors in the pathogenetic scenario. They have been defined as 'second wave antibodies' for the link with detoxifying mechanisms limiting the oxidative stress in glomeruli that are classically stimulated in a second phase of inflammation. These findings have important clinical implications that may modify the laboratory approach to LN. Serum levels of anti-ENO1 and anti-H2 IgG2 should be measured in the follow up of patients for designing the length of therapies and identify those patients who respond to treatments. Anti-SOD2 could help to monitor and potentiate the anti-inflammatory response in the kidney.
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
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Pier Luigi Meroni
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano-Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy.
| | - Gian Marco Ghiggeri
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy; Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Gabriella Moroni
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | | | - Franco Franceschini
- Rheumatology and Clinical Immunology, ASST SpedaliCivili and Università of Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology, ASST SpedaliCivili and Università of Brescia, Italy
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, and Nephrology and Dialysis Unit, Meyer Children's Hospital, Firenze, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Leonardo Scapozza
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | | | | | - Ken C Lo
- Nimble Therapeutics, Madison, WI, USA
| | - Lorenzo Cavagna
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Andrea Petretto
- Core Facilities-Proteomics Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, 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
- Nephrology and Dialysis, Arciospedale Santa Maria nuova, Reggio Emilia, Italy
| | | | - Angelo Manfredi
- Unit of Internal Medicine and Immunology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giuseppe A Ramirez
- Unit of Internal Medicine and Immunology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Pasquale Esposito
- Division of Nephrology, University of Genoa and Policlinico San Martino, Genova, Italy
| | | | - Simone Negrini
- Department of Internal Medicine, University of Genoa, Italy
| | - Federica Bui
- Division of Nephrology, University of Genoa and Policlinico San Martino, Genova, Italy
| | - Barbara Trezzi
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Giacomo Emmi
- Lupus Clinic Department of biomedicine, University of Florence, University Hospital Careggi, Florence, Italy
| | - Ilaria Cavazzana
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Valentina Binda
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, and Nephrology and Dialysis Unit, Meyer Children's Hospital, Firenze, Italy
| | - Paride Fenaroli
- Nephrology Unit, University Hospital, University of Parma, Parma, Italy
| | - Isabella Pisan
- 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, ASST SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Federica Mescia
- Division of Nephrology and Dialysis, ASST SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Stefano Volpi
- Division of Paediatric Rheumatology and Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marta Mosca
- Rheumatologu Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Angela Tincani
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Angelo Ravelli
- Division of Paediatric Rheumatology and Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Corrado Murtas
- Nephrology and Dialysis Unit, Ospedale Belcolle, 01100 Viterbo, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluca Caridi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Edoardo La Porta
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Verrina
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
8
|
Pisetsky DS, Gedye MJ, David LA, Spencer DM. The Binding Properties of Antibodies to Z-DNA in the Sera of Normal Healthy Subjects. Int J Mol Sci 2024; 25:2556. [PMID: 38473808 PMCID: PMC10931986 DOI: 10.3390/ijms25052556] [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: 12/29/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Antibodies to DNA are a diverse set of antibodies that bind sites on DNA, a polymeric macromolecule that displays various conformations. In a previous study, we showed that sera of normal healthy subjects (NHS) contain IgG antibodies to Z-DNA, a left-handed helix with a zig-zig backbone. Recent studies have demonstrated the presence of Z-DNA in bacterial biofilms, suggesting a source of this conformation to induce responses. To characterize further antibodies to Z-DNA, we used an ELISA assay with brominated poly(dGdC) as a source of Z-DNA and determined the isotype of these antibodies and their binding properties. Results of these studies indicate that NHS sera contain IgM and IgA as well as IgG anti-Z-DNA antibodies. As shown by the effects of ionic strength in association and dissociation assays, the anti-Z-DNA antibodies bind primarily by electrostatic interactions; this type of binding differs from that of induced anti-Z-DNA antibodies from immunized animals which bind by non-ionic interactions. Furthermore, urea caused dissociation of NHS anti-Z-DNA at molar concentrations much lower than those for the induced antibodies. These studies also showed IgA anti-Z-DNA antibodies in fecal water. Together, these studies demonstrate that antibodies to Z-DNA occur commonly in normal immunity and may arise as a response to Z-DNA of bacterial origin.
Collapse
Affiliation(s)
- David S. Pisetsky
- Medical Research Service, Durham Veterans Administration Medical Center, Durham, NC 27705, USA;
- Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Matthew J. Gedye
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; (M.J.G.); (L.A.D.)
| | - Lawrence A. David
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; (M.J.G.); (L.A.D.)
- Duke Microbiome Center, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Diane M. Spencer
- Medical Research Service, Durham Veterans Administration Medical Center, Durham, NC 27705, USA;
- Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
9
|
Rekvig OP. The greatest contribution to medical science is the transformation from studying symptoms to studying their causes-the unrelenting legacy of Robert Koch and Louis Pasteur-and a causality perspective to approach a definition of SLE. Front Immunol 2024; 15:1346619. [PMID: 38361929 PMCID: PMC10867267 DOI: 10.3389/fimmu.2024.1346619] [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: 11/29/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
The basic initiative related to this study is derived from the fact that systemic lupus erythematosus (SLE) is a unique and fertile system science subject. We are, however, still far from understanding its nature. It may be fair to indicate that we are spending more time and resources on studying the complexity of classified SLE than studying the validity of classification criteria. This study represents a theoretical analysis of current instinctual SLE classification criteria based on "the causality principle." The discussion has its basis on the radical scientific traditions introduced by Robert Koch and Louis Pasteur. They announced significant changes in our thinking of disease etiology through the implementation of the modern version of "the causality principle." They influenced all aspects of today's medical concepts and research: the transformation of medical science from studies of symptoms to study their causes, relevant for monosymptomatic diseases as for syndromes. Their studies focused on bacteria as causes of infectious diseases and on how the immune system adapts to control and prevent contagious spreading. This is the most significant paradigm shift in the modern history of medicine and resulted in radical changes in our view of the immune system. They described acquired post-infection immunity and active immunization by antigen-specific vaccines. The paradigm "transformation" has a great theoretical impact also on current studies of autoimmune diseases like SLE: symptoms and their cause(s). In this study, the evolution of SLE classification and diagnostic criteria is discussed from "the causality principle" perspective, and if contemporary SLE classification criteria are as useful as believed today for SLE research. This skepticism is based on the fact that classification criteria are not selected based on cogent causal strategies. The SLE classification criteria do not harmonize with Koch's and Pasteur's causality principle paradigms and not with Witebsky's Koch-derived postulates for autoimmune and infectious diseases. It is not established whether the classification criteria can separate SLE as a "one disease entity" from "SLE-like non-SLE disorders"-the latter in terms of SLE imitations. This is discussed here in terms of weight, rank, and impact of the classification criteria: Do they all originate from "one basic causal etiology"? Probably not.
Collapse
Affiliation(s)
- Ole Petter Rekvig
- Section for Autoimmunity, Fürst Medical Laboratory, Oslo, Norway
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
10
|
Jin H, Arase H. Neoself Antigens Presented on MHC Class II Molecules in Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:51-65. [PMID: 38467972 DOI: 10.1007/978-981-99-9781-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Major histocompatibility complex (MHC) class II molecules play a crucial role in immunity by presenting peptide antigens to helper T cells. Immune cells are generally tolerant to self-antigens. However, when self-tolerance is broken, immune cells attack normal tissues or cells, leading to the development of autoimmune diseases. Genome-wide association studies have shown that MHC class II is the gene most strongly associated with the risk of most autoimmune diseases. When misfolded self-antigens, called neoself antigens, are associated with MHC class II molecules in the endoplasmic reticulum, they are transported by the MHC class II molecules to the cell surface without being processed into peptides. Moreover, neoself antigens that are complexed with MHC class II molecules of autoimmune disease risk alleles exhibit distinct antigenicities compared to normal self-antigens, making them the primary targets of autoantibodies in various autoimmune diseases. Elucidation of the immunological functions of neoself antigens presented on MHC class II molecules is crucial for understanding the mechanism of autoimmune diseases.
Collapse
Affiliation(s)
- Hui Jin
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan.
| |
Collapse
|
11
|
Novikova TS, Ermakov EA, Kostina EV, Sinyakov AN, Sizikov AE, Nevinsky GA, Buneva VN. Hydrolysis of Oligodeoxyribonucleotides on the Microarray Surface and in Solution by Catalytic Anti-DNA Antibodies in Systemic Lupus Erythematosus. Curr Issues Mol Biol 2023; 45:9887-9903. [PMID: 38132463 PMCID: PMC10742339 DOI: 10.3390/cimb45120617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Anti-DNA antibodies are known to be classical serological hallmarks of systemic lupus erythematosus (SLE). In addition to high-affinity antibodies, the autoantibody pool also contains natural catalytic anti-DNA antibodies that recognize and hydrolyze DNA. However, the specificity of such antibodies is uncertain. In addition, DNA binding to a surface such as the cell membrane, can also affect its recognition by antibodies. Here, we analyzed the hydrolysis of short oligodeoxyribonucleotides (ODNs) immobilized on the microarray surface and in solution by catalytic anti-DNA antibodies from SLE patients. It has been shown that IgG antibodies from SLE patients hydrolyze ODNs more effectively both in solution and on the surface, compared to IgG from healthy individuals. The data obtained indicate a more efficient hydrolysis of ODNs in solution than immobilized ODNs on the surface. In addition, differences in the specificity of recognition and hydrolysis of certain ODNs by anti-DNA antibodies were revealed, indicating the formation of autoantibodies to specific DNA motifs in SLE. The data obtained expand our understanding of the role of anti-DNA antibodies in SLE. Differences in the recognition and hydrolysis of surface-tethered and dissolved ODNs need to be considered in DNA microarray applications.
Collapse
Affiliation(s)
- Tatiana S. Novikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Evgeny A. Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Elena V. Kostina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexander N. Sinyakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexey E. Sizikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Sciences, 630099 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Valentina N. Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
12
|
Rekvig OP. SLE classification criteria: Is "The causality principle" integrated and operative - and do the molecular and genetical network, on which criteria depend on, support the definition of SLE as "a one disease entity" - A theoretical discussion. Autoimmun Rev 2023; 22:103470. [PMID: 37884202 DOI: 10.1016/j.autrev.2023.103470] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Molecular and cellular aspects of the autoimmune pathophysiology in SLE is linked to the "The causality principle". SLE Classification Criteria identify per definition disease measures (here: synonymous with classification criteria), but not diagnostic criteria within a classical framework. These two mostly theoretical criteria collections represent a salient conflict between phenomenology and the causality principle - between disease measures and molecular interactions that promote such measures, in other words their cause(s). Essentially, each criterion evolves from immunogenic and inflammatory signals - some are interconnected, some are not. Disparate signals instigated by disparate causes. These may promote clinically heterogenous SLE cohorts with respect to organ affection, autoimmunity, and disease course. There is today no concise measures or arguments that settle whether SLE cohorts evolve from one decisive etiological factor (homogenous cohorts), or if disparate patho-biological factors promote SLE (heterogenous cohorts). Current SLE cohorts are not ideal substrates to serve as study objects if the research aims are to describe etiology, and molecular interactions that cause - and link - primary and secondary pathophysiological events together - events that account for early and progressive SLE. We have to develop SLE criteria allowing us to identify definable categories of SLE in order to describe etiology, pathophysiology and diagnostic criteria of delimitated SLE versions. In this regard, the causality principle is central to define dominant etiologies of individual SLE categories, and subsequent and consequent down-stream diagnostic disease measures. In this sense, we may whether we like it or not identify different SLE categories like "genuine SLE" and "SLE-like non-SLE" syndromes. Many aspects of this problem are thoroughly discussed in this study.
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
|
13
|
Rojo R, Calvo Alén J, Prada Á, Valor S, Roy G, López-Hoyos M, Cervera R, Sánchez Mateos P, Jurado Roger A. Recommendations for the use of anti-dsDNA autoantibodies in the diagnosis and follow-up of systemic lupus erythematosus - A proposal from an expert panel. Autoimmun Rev 2023; 22:103479. [PMID: 37967782 DOI: 10.1016/j.autrev.2023.103479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Anti-dsDNA autoantibodies are listed as one of the classification criteria for systemic lupus erythematosus (SLE) and are relatively effective indicators for monitoring disease activity and treatment response. Therefore, clinicians rely on them to diagnose and adjust medication and treatment strategies for SLE patients. However, the use of anti-dsDNA antibodies is not free from controversy. Part of this controversy stems from the fact that anti-dsDNA antibodies are found in several disorders, besides SLE. In addition to this, anti-dsDNA antibodies are a heterogeneous group of antibodies, and their determination still lacks proper standardization. Moreover, anti-dsDNA testing specificity and diagnostic performance change depending on the population under study. These and other issues result in inconsistency and encumber the clinical use of anti-dsDNA antibodies. A panel of medical laboratory and clinical experts on SLE discussed such issues based on their clinical experience in a first meeting, establishing a series of recommendations. The proceedings of this first meeting, plus an exhaustive review of the literature, were used to compose a paper draft. The panel subsequently discussed and refined this draft in a second meeting, the result of which is this paper. This document is relevant to clinical laboratories as it guides to improving diagnosis and monitoring of SLE. Simultaneously, it will help laboratories compile more informative reports, not limited to a mere number. It is also relevant to clinical doctors who wish to better understand laboratory methods so that they can do a more efficient, better-aimed laboratory test ordering.
Collapse
Affiliation(s)
- Ricardo Rojo
- Specialist Consultant at the Immunology Department of the University Hospital of A Coruña, Spain
| | - Jaime Calvo Alén
- Head of the Rheumatology Department at the Araba University Hospital, Vitoria, Spain
| | - Álvaro Prada
- Head of Section at the Immunology Laboratory of the University Hospital of Donostia, Spain
| | | | - Garbiñe Roy
- Head of the Autoimmunity Section at the Immunology Department of the Ramón y Cajal University Hospital, Madrid, Spain
| | - Marcos López-Hoyos
- Head of the Immunology Department at the Marqués de Valdecilla-IDIVAL University Hospital, Santander. Full Professor, Molecular Biology Department at the University of Cantabria, Santander, Spain.
| | - Ricard Cervera
- Head of the Department of Autoimmune Diseases, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Paloma Sánchez Mateos
- Full Professor at the Complutense University, and Specialist Consultant at the Immunology Department of the Gregorio Marañón General University Hospital, Madrid, Spain
| | - Aurora Jurado Roger
- Head of Section at the Immunology and Allergology Department of the Reina Sofía University Hospital, Córdoba, Spain
| |
Collapse
|
14
|
Kubota T. An Emerging Role for Anti-DNA Antibodies in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:16499. [PMID: 38003689 PMCID: PMC10671047 DOI: 10.3390/ijms242216499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Anti-DNA antibodies are hallmark autoantibodies produced in systemic lupus erythematosus (SLE), but their pathogenetic role is not fully understood. Accumulating evidence suggests that some anti-DNA antibodies enter different types of live cells and affect the pathophysiology of SLE by stimulating or impairing these cells. Circulating neutrophils in SLE are activated by a type I interferon or other stimuli and are primed to release neutrophil extracellular traps (NETs) on additional stimulation. Anti-DNA antibodies are also involved in this process and may induce NET release. Thereafter, they bind and protect extracellular DNA in the NETs from digestion by nucleases, resulting in increased NET immunogenicity. This review discusses the pathogenetic role of anti-DNA antibodies in SLE, mainly focusing on recent progress in the two research fields concerning antibody penetration into live cells and NETosis.
Collapse
Affiliation(s)
- Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Tsuchiura 300-0051, Ibaraki, Japan
| |
Collapse
|
15
|
Nieto-Aristizábal I, España LJ, Ortega J, Ruiz-Ordoñez I, Vivas ÁJ, Castaño GL, Castillo C, Bautista-Vargas M, Suárez A, Cañas CA, Tobón GJ. Diagnostic Performance of Anti-dsDNA Tests by Indirect Immunofluorescence and Enzyme-linked Immunosorbent Assay in Patients with Systemic Lupus Erythematosus. J Appl Lab Med 2023; 8:713-725. [PMID: 37018046 DOI: 10.1093/jalm/jfad006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/17/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Several laboratory techniques for anti double-stranded (ds) DNA detection in systemic lupus erythematosus (SLE) are available, with variable diagnostic performance. We aimed to evaluate anti-dsDNA's diagnostic performance by indirect immunofluorescence (IIF) and enzyme-linked immunosorbent assay (EIA). METHODS We conducted a single-center retrospective (2015 to 2020) study. Patients with anti-dsDNA tests by IIF and EIA were included. We evaluated the indications, applications, concordance, positive predictive value (PPV) of anti-dsDNA to confirm SLE diagnosis or flares, and associations of disease manifestations with positivity with each technique. RESULTS A total of 1368 reports of anti-dsDNA tests by IIF and EIA and the corresponding medical records of the patients were analyzed. The main indication for anti-dsDNA testing was to help in the diagnosis of SLE in 890 (65%) of the samples, and the main application after obtaining the results was SLE exclusion in 782 (57.2%) cases. The combination with the highest frequency was the negativity result by both techniques in 801 (58.5%) cases (Cohen kappa 0.57). Both methods were positive in 300 patients with SLE (Cohen kappa 0.42). The PPVs of anti-dsDNA tests to confirm diagnosis/flare was 79.64% (95% CI, 75.35-83.35) by EIA, 78.75% (95% CI, 74.27-82.62) by IIF, and 82% (95% CI, 77.26-85.93) when both were positive. CONCLUSIONS Anti-dsDNA detection by IIF and EIA are complementary and may indicate different clinical patterns in patients with SLE. The detection of anti-dsDNA antibodies by both techniques has a higher PPV than either separately for confirming SLE diagnosis or flares. These results highlight the need for evaluating both methods in clinical practice.
Collapse
Affiliation(s)
- Ivana Nieto-Aristizábal
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Cra. 98 No. 18-49, Cali 760032, Colombia
| | | | - Jorge Ortega
- Universidad Icesi, Facultad de Ciencias de la Salud, Cali, Colombia
| | - Ingrid Ruiz-Ordoñez
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Cra. 98 No. 18-49, Cali 760032, Colombia
| | - Álvaro J Vivas
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Cra. 98 No. 18-49, Cali 760032, Colombia
| | - Gloria L Castaño
- Fundación Valle del Lili, Laboratorio Clínico, Cra. 98 No. 18-49, Cali 760032, Colombia
| | - Carmen Castillo
- Fundación Valle del Lili, Laboratorio Clínico, Cra. 98 No. 18-49, Cali 760032, Colombia
| | | | - Ana Suárez
- Universidad Icesi, Facultad de Ciencias de la Salud, Cali, Colombia
| | - Carlos A Cañas
- Universidad Icesi, CIRAT: Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Fundación Valle del Lili, Unidad de Reumatología, Cra. 98 No. 18-49, Cali 760032, Colombia
| | - Gabriel J Tobón
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Cra. 98 No. 18-49, Cali 760032, Colombia
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University, Springfield, IL 62901, United States
| |
Collapse
|
16
|
Tew DJ, Hebert JM, Schmier BJ. Discovery and properties of a monoclonal antibody targeting 8-oxoA, an oxidized adenine lesion in DNA and RNA. Redox Biol 2023; 62:102658. [PMID: 36989571 PMCID: PMC10074937 DOI: 10.1016/j.redox.2023.102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/17/2023] Open
Abstract
8-oxoA, a major oxidation product of adenosine, is a mispairing, mutagenic lesion that arises in DNA and RNA when •OH radicals or one-electron oxidants attack the C8 adenine atom or polymerases misincorporate 8-oxo(d)ATP. The danger of 8-oxoA is underscored by the existence of dedicated cellular repair machinery that explicitly excise it from DNA, the attenuation of translation induced by 8-oxoA-mRNA or damaged ribosomes, and its potency as a TLR7 agonist. Here we present the discovery, purification, and biochemical characterization of a new mouse IgGk1 monoclonal antibody (6E4) that specifically targets 8-oxoA. Utilizing an AchE-based competitive ELISA assay, we demonstrate the selectivity of 6E4 for 8-oxoA over a plethora of canonical and chemically modified nucleosides including 8-oxoG, A, m6A, 2-oxoA, and 5-hoU. We further show the ability of 6E4 to exclusively recognize 8-oxoA in nucleoside triphosphates (8-oxoATP) and DNA/RNA oligonucleotides containing a single 8-oxoA. 6E4 also binds 8-oxoA in duplex DNA/RNA antigens where the lesion is either paired correctly or base mismatched. Our findings define the 8-oxoAde nucleobase as the critical epitope and indicate mAb 6E4 is ideally suited for a broad range of immunological applications in nucleic acid detection and quality control.
Collapse
|
17
|
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
|
18
|
Rekvig OP. SLE classification criteria: Science-based icons or algorithmic distractions – an intellectually demanding dilemma. Front Immunol 2022; 13:1011591. [PMID: 36248792 PMCID: PMC9555175 DOI: 10.3389/fimmu.2022.1011591] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
It is, so to say, not a prerogative authority assigned to SLE classification criteria that allow them to declare something definitively important about SLE. This is particularly true as criteria-based classification processes overrule the highly needed evolution of concise diagnostic criteria. It is classification criteria that allocate SLE patients into cohorts intended to describe the nature of their disease. Therefore, all major SLE classification criteria since the 1971 preliminary criteria usurp the role of diagnostic criteria. Today´s practice silently accept that the SLE classification process “diagnose” SLE patients despite the fact that classification criteria are not accepted as diagnostic criteria! This is a central paradox in contemporary SLE research strategies. Contemporary SLE cohorts are designed to investigate SLE´s etiological features. However, each cohort that is categorized by classification criteria has one central inherent problem. From theoretical and practical arguments, they embody multiple distinct clinical phenotypes. This raises the critical and principal question if phenotypically heterogenic SLE cohorts are useful to identify basic SLE-specific etiology(ies) and disease process(es). In times to come, we must prioritize development of firm diagnostic criteria for SLE, as the classification criteria have not contributed to reduce the enigmatic character of the syndrome. No radical improvements are visible in the horizon that may lead to concise investigations of SLE in well-defined homogenous SLE cohorts. We must develop new strategies where studies of phenotypically standardized cohorts of SLE must be central elements. Problems related to contemporary SLE classification criteria are contemplated, analyzed, and critically discussed in this study.
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
- *Correspondence: Ole Petter Rekvig,
| |
Collapse
|
19
|
Al-Mughales JA. Anti-Nuclear Antibodies Patterns in Patients With Systemic Lupus Erythematosus and Their Correlation With Other Diagnostic Immunological Parameters. Front Immunol 2022; 13:850759. [PMID: 35359932 PMCID: PMC8964090 DOI: 10.3389/fimmu.2022.850759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/18/2022] [Indexed: 11/20/2022] Open
Abstract
Background Antinuclear antibodies (ANA) are major immunodiagnostic tools in systemic lupus erythematosus (SLE); however, their clinical and pathogenic roles are not yet elucidated and are a subject of controversy. Objectives The aim of the study is to explore the pathogenic significance of ANA patterns among SLE patients, by analyzing their association with ANA titers, complement levels and other pathogenic immune markers, namely, anti-double-stranded DNA (anti-dsDNA), complements C3 and C4, rheumatoid factor (RF), anticardiolipin antibodies IgG (ACL IgG) and IgM (ACL IgM), Beta-2 Glycoprotein 1 Antibodies (β2-GP) IgG (β2-IgM) and IgM (β2-IgM), and lupus anticoagulant (LA). Method A comparative cross-sectional study was conducted among 495 SLE patients, who were diagnosed and classified by consultant rheumatologists according to the new European League Against Rheumatism (EULAR)/American College of Rheumatology (ACR) 2019 criteria. SLE immunodiagnostic profiles were analyzed including the following parameters: ANA antibody titers and staining patterns, anti-dsDNA, C3 and C4 levels, aCL, and anti-β2-GP and LA. Result The most frequently observed ANA patterns were the speckled (52.1%) and homogeneous (35.2%) patterns, while other patterns were rare representing less than 7% of the patients each. ANA titers were highest in patients with mixed pattern followed by the speckled pattern. Of all the investigated patterns, the peripheral pattern showed the most pathogenic immune profile, namely, highest levels of anti-dsDNA, lowest levels of C4, and highest levels of aCL and β2-GP IgG and IgM. Conclusion This retrospective study showed that speckled followed by homogeneous ANA patterns were predominant accounting for 52.1 and 35.2% of the patients. The ANA pattern showed several associations with other immune markers that are documented to have significant clinical implications in SLE. Peripheral, mixed, and speckled patterns were associated with higher profiles of immune markers indicative of a potential prognostic value of these patterns in SLE.
Collapse
Affiliation(s)
- Jamil A. Al-Mughales
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Laboratory Medicine, Diagnostic Immunology Division, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
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
|
21
|
Infantino M, Nagy E, Bizzaro N, Fischer K, Bossuyt X, Damoiseaux J. Anti-dsDNA antibodies in the classification criteria of systemic lupus erythematosus. J Transl Autoimmun 2022; 5:100139. [PMID: 35028552 PMCID: PMC8741517 DOI: 10.1016/j.jtauto.2021.100139] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022] Open
Abstract
Anti-double stranded DNA (dsDNA) antibodies play an important role in the diagnosis, classification and management of systemic lupus erythematosus (SLE), an autoimmune disease characterized by heterogeneous clinical manifestations and a wide range of autoantibodies, which makes the diagnosis quite challenging. In the absence of diagnostic criteria, classification criteria have been used for many decades. The first classification criteria for SLE were formulated in 1971 by the American College of Rheumatology (ACR), followed by two revisions in 1982 and 1997. In order to improve their clinical performance and to reflect new knowledge on autoantibodies, new classification criteria for SLE were issued in 2012 by the Systemic Lupus International Collaborating Clinics (SLICC). These criteria proposed to classify only patients that have at least one immunologic criterion, overcoming SLE classification based solely on clinical manifestations. In 2019, the European League Against Rheumatism (EULAR)/ACR proposed new criteria that aimed to maintain the high specificity of the ACR criteria with a sensitivity close to the SLICC 2012 criteria. These 2019 criteria reinforced the importance of autoantibodies in SLE diagnosis, assigning the highest score (6 points) to anti-dsDNA antibodies in the fully weighted scoring of the disease. The current criteria require the use of an anti-dsDNA assay with at least 90% specificity, such as the Crithidia luciliae immunofluorescence test (CLIFT) or FARR assay. However, the criteria do not comment on all the tests currently widely used in clinical laboratories. Neither do they consider the technological evolutions, nor standardization issues. Since strict adherence to any of the classification criteria, including the serological items, could lead to possible misclassification of SLE and/or delayed diagnosis, test characteristics of the distinct immunoassays should be taken into consideration.
Collapse
Affiliation(s)
- Maria Infantino
- Immunology and Allergy Laboratory, San Giovanni di Dio Hospital, Florence, Italy
| | - Eszter Nagy
- National Institute of Locomotor Diseases and Disabilities, Budapest, Hungary.,Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, San Antonio Hospital, Tolmezzo, Italy
| | - Katarzyna Fischer
- Individual Laboratory for Rheumatologic Diagnostics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
22
|
Pisetsky DS, Shaffer R, Armstrong DD, Spencer DM. The interaction of anti-DNA antibodies with DNA antigen: Evidence for hysteresis for high avidity binding. Clin Immunol 2021; 231:108848. [PMID: 34492381 DOI: 10.1016/j.clim.2021.108848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/11/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Antibodies to DNA (anti-DNA) are the serological hallmark of systemic lupus erythematosus. Previous studies have indicated that the phosphodiester backbone is the main antigenic target, with electrostatic interactions important for high avidity. To define further these interactions, the effects of ionic strength on anti-DNA binding of SLE plasmas were assessed in association and dissociation assays by ELISA. As these studies demonstrated, increasing ionic strength to a concentration of 1000 mM NaCl reduced antibody binding although the extent of the reduction varied among samples. In dissociation assays, differences among plasmas were also observed. For one of the plasmas, binding to DNA displayed resistance to dissociation by increasing ionic strength even though these concentrations limited binding in association assays. Time course studies showed a gradual change in binding interactions. These studies indicate that anti-DNA binding can involve both electrostatic and non-electrostatic interactions, with binding in some plasmas showing evidence of hysteresis.
Collapse
Affiliation(s)
- David S Pisetsky
- Medical Research Service, Durham Veterans Administration Medical Center, Durham, NC 27705, USA; Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | - Diane M Spencer
- Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
23
|
González Rodríguez C, Aparicio Hernández M, Alarcón Torres I. Update and clinical management of anti-DNA auto-antibodies. ADVANCES IN LABORATORY MEDICINE 2021; 2:313-331. [PMID: 37362416 PMCID: PMC10197362 DOI: 10.1515/almed-2021-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/19/2020] [Indexed: 06/28/2023]
Abstract
Anti-deoxyribonucleic acid (DNA) antibodies in the clinical laboratory are intimately linked to the diagnosis and monitoring of systemic lupus erythematosus (SLE); however, the characteristics of the analytical methods and the properties of the antibodies themselves are heterogeneous. To review the definition and properties of anti-double-stranded anti-DNA (anti-dsDNA) antibodies, the adequacy of analytical methods, and the clinical requirements for this biomarker. Through PubMed we searched the existing literature with the terms anti-dsDNA, editorial, review, guideline, meta-analysis and SLE. The last search, anti-dsDNA and SLE restricted to the last two years. Information was expanded through related articles and those published in official state bodies related to anti-dsDNA and SLE. Clinical laboratory methods for anti-dsDNA analysis and their characteristics are analyze. The clinical utility of anti-dsDNA in its diagnostic, clinical association and follow-up aspects of SLE is reviewed. There is wide variability in analytical methods and deficits in standardization persist. They are part of the current SLE classification criteria and are used as markers in the follow-up of the disease. Their diagnostic usefulness improves when they are determined in antinuclear antibody (ANA)-positive patients. In follow-up, quantification is of interest, preferably with the same analytical method (given the deficits in standardization).
Collapse
Affiliation(s)
| | - MªBelén Aparicio Hernández
- Servicio Bioquímica Clínica y Análisis Cínicos, Complejo Asistencial Universitario Salamanca, Salamanca, Spain
| | | |
Collapse
|
24
|
Abd El Monem Teama M, Adham El-Mohamdy M, Abdellah Abdullah Mahmoud F, Mohammed Badr F. Autoantibody Profile of Egyptian Juvenile Systemic Lupus Erythematosus Patients and Its Association with Clinical Characteristics and Disease Activity. Open Access Rheumatol 2021; 13:201-212. [PMID: 34295197 PMCID: PMC8291800 DOI: 10.2147/oarrr.s317315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/02/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE This study was conducted to estimate the frequency of anti-nuclear antibodies (ANAs), anti-dsDNA, and anti-extractable nuclear antigen (ENA) antibodies in juvenile systemic lupus erythematosus (JSLE) patients and their association with different clinical manifestations and disease activity. PATIENTS AND METHODS A cross-sectional study that includes 100 JSLE patients from Ain Shams University Hospital was conducted. All subjects underwent history taking, clinical examination, assessment of disease activity based on the SLE disease activity index (SLEDAI), laboratory investigations, and tests for autoantibodies, namely ANA, anti-dsDNA, and anti-ENA antibodies, including anti-Ro (SSA), anti-La (SSB), anti-Smith (Sm), and anti-U1-ribonucleoprotein (U1-RNP). RESULTS The most common clinical features were polyarthralgia (71%), haematological manifestations (65%), malar rash (54%), and nephritis (51%), respectively. All patients had positive ANA (100%), while anti-dsDNA frequency was 83%. The most common anti-ENA antibodies were anti-RNP (41%), anti-Sm (31%), anti-SSA (27%), and anti-SSB (20%), respectively. Anti-RNP had a clinical association with oral ulcer, Raynaud' phenomena, haematological, neuropsychiatric and thromboembolic manifestations. Meanwhile, anti-Sm had a significant association with serositis, mucocutaneous, constitutional, and neuropsychiatric manifestations. Anti-SSA was associated with mucocutaneous, musculoskeletal, Raynaud' phenomena, renal, haematological and cardiac manifestations, while anti-SSB was significantly associated with malar rash, serositis, thromboembolic, musculoskeletal, and neuropsychiatric manifestations. Concerning SLEADI score, anti-dsDNA antibody was significantly associated with moderate disease activity score (p=0.032) while anti-SSA significantly associated with high disease activity (p=0.045). Both anti-SSB and anti-Sm were significantly associated with both moderate and high disease activities, meanwhile anti-U1-RNP was associated with moderate disease activity (p=0.014). CONCLUSION Anti-dsDNA and anti-ENAs antibodies were frequently found in JSLE patients (83%, 63%), respectively. They were significantly associated with variable clinical manifestations and could be used as predictors for assessment of disease activity.
Collapse
Affiliation(s)
- Mohammed Abd El Monem Teama
- Internal Medicine Department, Division of Rheumatology and Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Fatma Mohammed Badr
- Internal Medicine Department, Division of Rheumatology and Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
25
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Bruschi M, Moroni G, Sinico RA, Franceschini F, Fredi M, Vaglio A, Cavagna L, Petretto A, Pratesi F, Migliorini P, Locatelli F, Pazzola G, Pesce G, Bagnasco M, Manfredi A, Ramirez GA, Esposito P, Murdaca G, Negrini S, Cipriani L, Trezzi B, Emmi G, Cavazzana I, Binda V, d’Alessandro M, Fenaroli P, Pisani I, Garibotto G, Montecucco C, Santoro D, Scolari F, Volpi S, Mosca M, Tincani A, Candiano G, Prunotto M, Verrina E, Angeletti A, Ravelli A, Ghiggeri GM. Serum IgG2 antibody multi-composition in systemic lupus erythematosus and in lupus nephritis (Part 2): prospective study. Rheumatology (Oxford) 2021; 60:3388-3397. [PMID: 33351137 PMCID: PMC8516512 DOI: 10.1093/rheumatology/keaa793] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Circulating anti-ENO1 and anti-H2A IgG2 have been identified as specific signatures of LN in a cross-over approach. We sought to show whether the same antibodies identify selected population of patients with LN with potentially different clinical outcomes. METHODS Here we report the prospective analysis over 36 months of circulating IgG2 levels in patients with newly diagnosed LN (n=91) and SLE (n=31) and in other patients with SLE recruited within 2 years from diagnosis (n=99). Anti-podocyte (ENO1), anti-nucleosome (DNA, histone 2 A, histone 3) and anti-circulating proteins (C1q, AnnexinA1-ANXA1) IgG2 antibodies were determined by home-made techniques. RESULTS LN patients were the main focus of the study. Anti-ENO1, anti-H2A and anti-ANXA1 IgG2 decreased in parallel to proteinuria and normalized within 12 months in the majority of patients while anti-dsDNA IgG2 remained high over the 36 months. Anti-ENO1 and anti-H2A had the highest association with proteinuria (Heat Map) and identified the highest number of patients with high proteinuria (68% and 71% respectively) and/or with reduced estimated glomerula filtration rate (eGFR) (58% for both antibodies) compared with 23% and 17% of anti-dsDNA (agreement analysis). Anti-ENO1 positive LN patients had higher proteinuria than negative patients at T0 and presented the maximal decrement within 12 months. CONCLUSIONS Anti-ENO1, anti-H2A and anti-ANXA1 antibodies were associated with high proteinuria in LN patients and Anti-ENO1 also presented the maximal reduction within 12 months that paralleled the decrease of proteinuria. Anti-dsDNA were not associated with renal outcome parameters. New IgG2 antibody signatures should be utilized as tracers of personalized therapies in LN. TRIAL REGISTRATION The Zeus study was registered at https://clinicaltrials.gov (study number: NCT02403115).
Collapse
Affiliation(s)
- Maurizio Bruschi
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gabriella Moroni
- Division of Nephrology and Dialysis, Fondazione IRCCS Ca' Granda Ospedale Maggiore, Milano, Italy
| | | | - Franco Franceschini
- Rheumatology and Clinical Immunology, ASST SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology, ASST SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences ‘Mario Serio’, University of Firenze, Firenze, Italy
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Firenze, Italy
| | - Lorenzo Cavagna
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Andrea Petretto
- Core Facilities-Proteomics Laboratory, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, 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, Genoa, Italy
| | - Marcello Bagnasco
- Medical and Radiometabolic Therapy Unit, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Angelo Manfredi
- Unit of Internal Medicine and Immunology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giuseppe A Ramirez
- Unit of Internal Medicine and Immunology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Pasquale Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Simone Negrini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Leda Cipriani
- Division of Nephrology, University of Genoa and Policlinico San Martino, Genova, 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 SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Valentina Binda
- Division of Nephrology and Dialysis, Fondazione IRCCS Ca' Granda Ospedale Maggiore, Milano, Italy
| | - Matteo d’Alessandro
- Division of Nephrology, Dialysis and Transplantation, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paride Fenaroli
- Nephrology Unit, University Hospital, University of Parma, Parma, Italy
| | - Isabella Pisani
- Nephrology Unit, University Hospital, University of Parma, Parma, Italy
| | - Giacomo Garibotto
- Division of Nephrology, University of Genoa and Policlinico San Martino, Genova, 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 Paediatric Rheumatology Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marta Mosca
- Rheumatologu Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology, ASST SpedaliCivili and Università of Brescia, Brescia, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Prunotto
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Enrico Verrina
- Division of Nephrology, Dialysis and Transplantation, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Angeletti
- Division of Nephrology, Dialysis and Transplantation, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Angelo Ravelli
- Division of Paediatric Rheumatology Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Division of Nephrology, Dialysis and Transplantation, Scientific Institute for Research and Health Care, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
27
|
Tsuji H, Ohmura K, Jin H, Naito R, Arase N, Kohyama M, Suenaga T, Sakakibara S, Kochi Y, Okada Y, Yamamoto K, Kikutani H, Morinobu A, Mimori T, Arase H. Anti-dsDNA antibodies recognize DNA presented on HLA class II molecules of systemic lupus erythematosus risk alleles. Arthritis Rheumatol 2021; 74:105-111. [PMID: 34164946 DOI: 10.1002/art.41897] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/08/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Specific HLA class II alleles are associated with susceptibility to systemic lupus erythematosus (SLE). The role of HLA class II molecules in SLE pathogenesis remains unclear, although anti-DNA antibodies are specific to SLE and correlate to disease activity. We previously demonstrated that misfolded proteins bound to HLA class II molecules are specific targets for the autoantibodies produced in autoimmune diseases. We hypothesized that DNA binds to HLA class II molecules in a manner similar to that of misfolded proteins and that DNA bound to HLA class II molecules is involved in SLE pathogenicity. METHODS We analyzed the binding of DNA to HLA class II molecules, as well as the response of cells expressing anti-DNA B cell receptors (BCR) to cells expressing the DNA/HLA class II complex. RESULTS Efficient binding of DNA to HLA class II molecules was observed in risk alleles of SLE, such as HLA-DRB1*15:01. The efficiency of DNA binding to each HLA-DR allele was positively associated with the risk of SLE conferred by the HLA-DR allele. In addition, reporter cells carrying anti-DNA BCRs were activated by cells expressing DNA/HLA class II complexes. CONCLUSION DNA bound to HLA class II molecules is involved in SLE pathogenesis.
Collapse
Affiliation(s)
- Hideaki Tsuji
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, 606-8507, Japan.,Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, 606-8507, Japan
| | - Hui Jin
- Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Naito
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, 606-8507, Japan.,Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriko Arase
- Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Dermatology, Osaka University Graduate School of Medicine, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masako Kohyama
- Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tadahiro Suenaga
- Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Microbiology, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, 960-1295, Japan
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.,Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yukinori Okada
- Laboratory of Statistical Immunology, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Hitoshi Kikutani
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, 606-8507, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, 606-8507, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
28
|
Hartl J, Serpas L, Wang Y, Rashidfarrokhi A, Perez OA, Sally B, Sisirak V, Soni C, Khodadadi-Jamayran A, Tsirigos A, Caiello I, Bracaglia C, Volpi S, Ghiggeri GM, Chida AS, Sanz I, Kim MY, Belmont HM, Silverman GJ, Clancy RM, Izmirly PM, Buyon JP, Reizis B. Autoantibody-mediated impairment of DNASE1L3 activity in sporadic systemic lupus erythematosus. J Exp Med 2021; 218:e20201138. [PMID: 33783474 PMCID: PMC8020718 DOI: 10.1084/jem.20201138] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Antibodies to double-stranded DNA (dsDNA) are prevalent in systemic lupus erythematosus (SLE), particularly in patients with lupus nephritis, yet the nature and regulation of antigenic cell-free DNA (cfDNA) are poorly understood. Null mutations in the secreted DNase DNASE1L3 cause human monogenic SLE with anti-dsDNA autoreactivity. We report that >50% of sporadic SLE patients with nephritis manifested reduced DNASE1L3 activity in circulation, which was associated with neutralizing autoantibodies to DNASE1L3. These patients had normal total plasma cfDNA levels but showed accumulation of cfDNA in circulating microparticles. Microparticle-associated cfDNA contained a higher fraction of longer polynucleosomal cfDNA fragments, which bound autoantibodies with higher affinity than mononucleosomal fragments. Autoantibodies to DNASE1L3-sensitive antigens on microparticles were prevalent in SLE nephritis patients and correlated with the accumulation of cfDNA in microparticles and with disease severity. DNASE1L3-sensitive antigens included DNA-associated proteins such as HMGB1. Our results reveal autoantibody-mediated impairment of DNASE1L3 activity as a common nongenetic mechanism facilitating anti-dsDNA autoreactivity in patients with severe sporadic SLE.
Collapse
Affiliation(s)
- Johannes Hartl
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Lee Serpas
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Yueyang Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Oriana A. Perez
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Benjamin Sally
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Vanja Sisirak
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Le Centre national de la recherche scientifique - unité mixte de recherche 5164, ImmunoConcEpt, Universite ´de Bordeaux, Bordeaux, France
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY
| | - Ivan Caiello
- Division of Rheumatology, Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Claudia Bracaglia
- Division of Rheumatology, Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Stefano Volpi
- Centro per le Malattie Autoinfiammatorie e Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università degli Studi di Genova, Genoa, Italy
| | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Giannina Gaslini, Genoa, Italy
| | - Asiya Seema Chida
- Division of Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA
| | - Mimi Y. Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - H. Michael Belmont
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Gregg J. Silverman
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Robert M. Clancy
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Peter M. Izmirly
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jill P. Buyon
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
29
|
Granito A, Muratori L, Tovoli F, Muratori P. Diagnostic role of anti-dsDNA antibodies: do not forget autoimmune hepatitis. Nat Rev Rheumatol 2021; 17:244. [PMID: 33462415 DOI: 10.1038/s41584-021-00573-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Alessandro Granito
- Division of Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
- Center for the Study and Treatment of Autoimmune Diseases of the Liver and Biliary System, University of Bologna, Bologna, Italy.
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy.
| | - Luigi Muratori
- Division of Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Center for the Study and Treatment of Autoimmune Diseases of the Liver and Biliary System, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Francesco Tovoli
- Division of Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Paolo Muratori
- Division of Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Center for the Study and Treatment of Autoimmune Diseases of the Liver and Biliary System, University of Bologna, Bologna, Italy
- Department of the Science of Quality of Life, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
30
|
Xia Y, Fang X, Dai X, Li M, Jin L, Tao J, Li X, Wang Y, Li X. Iguratimod ameliorates nephritis by modulating the Th17/Treg paradigm in pristane-induced lupus. Int Immunopharmacol 2021; 96:107563. [PMID: 33812258 DOI: 10.1016/j.intimp.2021.107563] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Iguratimod, an anti-rheumatic drug, has been widely used in the treatment of rheumatoid arthritis, but is still at an investigative stage for treatment of systemic lupus erythematosus (SLE). We examined the therapeutic effects of iguratimod and the mechanism underlying the efficacy in murine lupus model. METHODS Pristane-induced lupus model of BALB/c mice (PI mice) were treated with iguratimod and mycophenolate mofetil. Proteinuria, anti-dsDNA antibodies and immunoglobulins production were measured. Renal pathology was evaluated. The percentage of Th17 and Treg cells in spleen and the expression of cytokines and mRNAs related to Th17 and Treg cells was analyzed. RESULTS Iguratimod attenuated the severity of nephritis in PI mice in a dose-dependent manner. Proteinuria was continuously decreased and pathology of glomerulonephritis and tubulonephritis was significantly reduced along with reduction of glomerular immune complex deposition. Also, serum anti-dsDNA and total IgG and IgM levels were reduced by iguratimod in mice. It is worth mentioning that the efficacy of the 30 mg/kg/d iguratimod dose is comparable to, or even better than, 100 mg kg/d of mycophenolate mofetil. Furthermore, the percentage of Th17 cells was found decreased and the percentage of Treg cells increased. ROR-γt mRNA and serum cytokines (IL-17A and IL-22) of Th17 cells decreased accordingly. By contrast, Foxp3 mRNA and cytokines (TGF-β and IL-10) of Treg cells increased. CONCLUSION Iguratimod ameliorates nephritis of SLE and modulates the Th17/Treg ratio in murine nephritis of SLE, suggesting that Iguratimod could be an effective drug in treatment of SLE.
Collapse
Affiliation(s)
- Yuan Xia
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan Fang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaojuan Dai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Manyun Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Li Jin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaomei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yiping Wang
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Xiangpei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
31
|
Takata H, Shimizu T, Kawaguchi Y, Ueda H, Elsadek NE, Ando H, Ishima Y, Ishida T. Nucleic acids delivered by PEGylated cationic liposomes in systemic lupus erythematosus-prone mice: A possible exacerbation of lupus nephritis in the presence of pre-existing anti-nucleic acid antibodies. Int J Pharm 2021; 601:120529. [PMID: 33781884 DOI: 10.1016/j.ijpharm.2021.120529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/04/2021] [Accepted: 03/21/2021] [Indexed: 12/18/2022]
Abstract
Nucleic acid-based therapy with plasmid DNA (pDNA) and small interfering RNA (siRNA) have received recent attention for their ability to modulate the cellular expression of genes and proteins. Polyethylene glycol-modified (PEGylated) cationic nanoparticles have been used as non-viral vectors for the in vivo delivery of these nucleic acids. We have reported that PEGylated cationic liposomes (PCL) including pDNA or siRNA induce anti-PEG antibodies upon repeated intravenous injection, leading to the formation of immune complexes and enhanced clearance from the blood of subsequent doses. However, the issue surrounding the association of nucleic acids with PCL whether induces anti-nucleic acid antibodies has not been studied. Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with the character of end-organ damage and the presence of anti-nuclear antibodies. We used a healthy mouse and an SLE mouse model to test the hypothesis that nucleic acids associated with PCL induce anti-nuclear antibodies and then induce SLE and exacerbate SLE symptoms. We report here that pDNA or siRNA associated with PCL (pDNA/PCL or siRNA/PCL) induced anti-DNA or RNA antibodies, respectively, in healthy mice. Repeated injections did not, however, cause SLE-like symptoms in the healthy mice. In addition, in SLE-prone mice with pre-existing anti-nuclear antibodies, pDNA/PCL were deposited on the kidneys and exacerbated lupus nephritis subsequent to the formation of immune complexes. These results may imply that nucleic acids associated with PCL do not contribute to the onset of SLE in healthy individuals who lack anti-nuclear antibodies, but nucleic acids may exacerbate the symptoms in SLE patients who have pre-existing anti-nuclear antibodies.
Collapse
Affiliation(s)
- Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Hiro Ueda
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Nehal E Elsadek
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
32
|
Schmetzer O, Lakin E, Roediger B, Duchow A, Asseyer S, Paul F, Siebert N. Anti-aquaporin 4 IgG Is Not Associated With Any Clinical Disease Characteristics in Neuromyelitis Optica Spectrum Disorder. Front Neurol 2021; 12:635419. [PMID: 33776892 PMCID: PMC7994757 DOI: 10.3389/fneur.2021.635419] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Neuromyelitis optica spectrum disorder (NMOSD) is a clinically defined, inflammatory central nervous system (CNS) disease of unknown cause, associated with humoral autoimmune findings such as anti-aquaporin 4 (AQP4)-IgG. Recent clinical trials showed a benefit of anti-B cell and anti-complement-antibodies in NMOSD, suggesting relevance of anti-AQP4-IgG in disease pathogenesis. Objective: AQP4-IgG in NMOSD is clearly defined, yet up to 40% of the patients are negative for AQP4-IgG. This may indicate that AQP4-IgG is not disease-driving in NMOSD or defines a distinct patient endotype. Methods: We established a biobank of 63 clinically well-characterized NMOSD patients with an extensive annotation of 351 symptoms, patient characteristics, laboratory results and clinical scores. We used phylogenetic clustering, heatmaps, principal component and longitudinal causal interference analyses to test for the relevance of anti-AQP4-IgG. Results: Anti-AQP4-IgG was undetectable in 29 (46%) of the 63 NMOSD patients. Within anti-AQP4-IgG-positive patients, anti-AQP4-IgG titers did not correlate with clinical disease activity. Comparing anti-AQP4-IgG-positive vs. -negative patients did not delineate any clinically defined subgroup. However, anti-AQP4-IgG positive patients had a significantly (p = 0.022) higher rate of additional autoimmune diagnoses. Conclusion: Our results challenge the assumption that anti-AQP4-IgG alone plays a disease-driving role in NMOSD. Anti-AQP4-IgG might represent an epiphenomenon associated with NMOSD, may represent one of several immune mechanisms that collectively contribute to the pathogenesis of this disease or indeed, anti-AQP4-IgG might be the relevant factor in only a subgroup of patients.
Collapse
Affiliation(s)
- Oliver Schmetzer
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Elisa Lakin
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ben Roediger
- Novartis Institutes for Biomedical Research - Autoimmunity, Transplantation and Inflammation, Basel, Switzerland
| | - Ankelien Duchow
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Susanna Asseyer
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nadja Siebert
- Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, NeuroCure Clinical Research Center (NCRC) and Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
33
|
Fan R, Zhang Y, Xu Y, Tong J, Chen Z, Gu M, Fan W, Chen Y, Peng F, Jiang Y. Serum antinuclear antibodies associate with worse prognosis in AQP4-positive neuromyelitis optica spectrum disorder. Brain Behav 2021; 11:e01865. [PMID: 33319460 PMCID: PMC7821584 DOI: 10.1002/brb3.1865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 08/31/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Studies have demonstrated that antinuclear antibodies (ANAs) may be correlated with neuromyelitis optica spectrum disorder (NMOSD). However, the conflicting results of studies about the value of ANAs in AQP4 antibody-positive NMOSD patients need to be further investigated. MATERIAL Case data were collected from 143 patients with AQP4 antibody-positive NMOSD. Patients were divided into two groups based on the ANA test results. The analysis of clinical characteristics, laboratory tests, and MRI examination results were compared between two groups: the NMOSD patients with ANA (+) and with ANA (-). RESULTS Disease duration of NMOSD is shorter in the ANA (+) patients with EDSS < 4 than in the ANA (-) patients (12.05 ± 16.73 versus 29.43 ± 41.03, p-value = .013). The median time from disease onset to an EDSS score of 4.0 is significantly longer in the ANA (-) NMOSD patients than in the ANA (+) patients (48.2 months versus 24 months, p = .04). In addition, ANA (RR, 2.234; 95% CI, 1.078-4.629; p-value = .031) can predict the severity of NMOSD. CONCLUSIONS Antinuclear antibodies seem to be associated with more severe disease activity in NMOSD patients.
Collapse
Affiliation(s)
- Rong Fan
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuefeng Zhang
- Department of Neurology, Guangzhou Brain Hospital, Guangzhou, China
| | - Yunqi Xu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiayi Tong
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhigang Chen
- Department of Neurology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Meifeng Gu
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenkui Fan
- Department of Nephrology, The Second People's Hospital of Foshan, Foshan, China
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Fuhua Peng
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Jiang
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Rekvig OP. Autoimmunity and SLE: Factual and Semantic Evidence-Based Critical Analyses of Definitions, Etiology, and Pathogenesis. Front Immunol 2020; 11:569234. [PMID: 33123142 PMCID: PMC7573073 DOI: 10.3389/fimmu.2020.569234] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
One cannot discuss anti-dsDNA antibodies and lupus nephritis without discussing the nature of Systemic lupus erythematosus (SLE). SLE is insistently described as a prototype autoimmune syndrome, with anti-dsDNA antibodies as a central biomarker and a pathogenic factor. The two entities, "SLE" and "The Anti-dsDNA Antibody," have been linked in previous and contemporary studies although serious criticism to this mutual linkage have been raised: Anti-dsDNA antibodies were first described in bacterial infections and not in SLE; later in SLE, viral and parasitic infections and in malignancies. An increasing number of studies on classification criteria for SLE have been published in the aftermath of the canonical 1982 American College of Rheumatology SLE classification sets of criteria. Considering these studies, it is surprising to observe a nearby complete absence of fundamental critical/theoretical discussions aimed to explain how and why the classification criteria are linked in context of etiology, pathogenicity, or biology. This study is an attempt to prioritize critical comments on the contemporary definition and classification of SLE and of anti-dsDNA antibodies in context of lupus nephritis. Epidemiology, etiology, pathogenesis, and measures of therapy efficacy are implemented as problems in the present discussion. In order to understand whether or not disparate clinical SLE phenotypes are useful to determine its basic biological processes accounting for the syndrome is problematic. A central problem is discussed on whether the clinical role of anti-dsDNA antibodies from principal reasons can be accepted as a biomarker for SLE without clarifying what we define as an anti-dsDNA antibody, and in which biologic contexts the antibodies appear. In sum, this study is an attempt to bring to the forum critical comments on the contemporary definition and classification of SLE, lupus nephritis and anti-dsDNA antibodies. Four concise hypotheses are suggested for future science at the end of this analytical study.
Collapse
Affiliation(s)
- Ole Petter Rekvig
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
- Fürst Medical Laboratory, Oslo, Norway
| |
Collapse
|
35
|
Natural Catalytic IgGs Hydrolyzing Histones in Schizophrenia: Are They the Link between Humoral Immunity and Inflammation? Int J Mol Sci 2020; 21:ijms21197238. [PMID: 33008051 PMCID: PMC7582518 DOI: 10.3390/ijms21197238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 12/21/2022] Open
Abstract
Schizophrenia is known to be accompanied not only with an imbalance in the neurotransmitter systems but also with immune system dysregulation and chronic low-grade inflammation. Extracellular histones and nucleosomes as damage-associated molecular patterns (DAMPs) trigger systemic inflammatory and toxic reactions by activating Toll-like receptors. In this work, we obtained the first evidence that polyclonal IgGs of patients with schizophrenia effectively hydrolyze five histones (H1, H2a, H2b, H3, and H4). Several strict criteria were used to demonstrate that histone-hydrolyzing activity is a property of the analyzed IgGs. The IgGs histone-hydrolyzing activity level, depending on the type of histone (H1–H4), was statistically significantly 6.1–20.2 times higher than that of conditionally healthy donors. The investigated biochemical properties (pH and metal ion dependences, kinetic characteristics) of these natural catalytic IgGs differed markedly from canonical proteases. It was previously established that the generation of natural catalytic antibodies is an early and clear sign of impaired humoral immunity. One cannot, however, exclude that histone-hydrolyzing antibodies may play a positive role in schizophrenia pathogenesis because histone removal from circulation or the inflamed area minimizes the inflammatory responses. Thus, it can be assumed that histone-hydrolyzing antibodies are a link between humoral immunity and inflammatory responses in schizophrenia.
Collapse
|
36
|
Pisetsky DS, Lipsky PE. New insights into the role of antinuclear antibodies in systemic lupus erythematosus. Nat Rev Rheumatol 2020; 16:565-579. [PMID: 32884126 DOI: 10.1038/s41584-020-0480-7] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2020] [Indexed: 01/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by antinuclear antibodies (ANAs) that form immune complexes that mediate pathogenesis by tissue deposition or cytokine induction. Some ANAs bind DNA or associated nucleosome proteins, whereas other ANAs bind protein components of complexes of RNA and RNA-binding proteins (RBPs). Levels of anti-DNA antibodies can fluctuate widely, unlike those of anti-RBP antibodies, which tend to be stable. Because anti-DNA antibody levels can reflect disease activity, repeat testing is common; by contrast, a single anti-RBP antibody determination is thought to suffice for clinical purposes. Experience from clinical trials of novel therapies has provided a new perspective on ANA expression during disease, as many patients with SLE are ANA negative at screening despite previously testing positive. Because trial results suggest that patients who are ANA negative might not respond to certain agents, screening strategies now involve ANA and anti-DNA antibody testing to identify patients with so-called 'active, autoantibody-positive SLE'. Evidence suggests that ANA responses can decrease over time because of the natural history of disease or the effects of therapy. Together, these findings suggest that, during established disease, more regular serological testing could illuminate changes relevant to pathogenesis and disease status.
Collapse
Affiliation(s)
- David S Pisetsky
- Departments of Medicine and Immunology, Duke University Medical Center and Medical Research Service, Veterans Administration Medical Center, Durham, NC, USA.
| | | |
Collapse
|
37
|
Gordon RE, Nemeth JF, Singh S, Lingham RB, Grewal IS. Harnessing SLE Autoantibodies for Intracellular Delivery of Biologic Therapeutics. Trends Biotechnol 2020; 39:298-310. [PMID: 32807530 DOI: 10.1016/j.tibtech.2020.07.003] [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: 06/04/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Intracellular delivery of therapeutic antibodies is highly desirable but remains a challenge for biomedical research and the pharmaceutical industry. Approximately two-thirds of disease-associated targets are found inside the cell. Difficulty blocking these targets with available drugs creates a need for technology to deliver highly specific therapeutic antibodies intracellularly. Historically, antibodies have not been believed to traverse the cell membrane and neutralize intracellular targets. Emerging evidence has revealed that anti-DNA autoantibodies found in systemic lupus erythematosus (SLE) patients can penetrate inside the cell. Harnessing this technology has the potential to accelerate the development of drugs against intracellular targets. Here, we dissect the mechanisms of the intracellular localization of SLE antibodies and discuss how to apply these insights to engineer successful cell-penetrating antibody drugs.
Collapse
Affiliation(s)
- Renata E Gordon
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Jennifer F Nemeth
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Sanjaya Singh
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Russell B Lingham
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Iqbal S Grewal
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA.
| |
Collapse
|
38
|
Pisetsky DS. Evolving story of autoantibodies in systemic lupus erythematosus. J Autoimmun 2020; 110:102356. [PMID: 31810857 PMCID: PMC8284812 DOI: 10.1016/j.jaut.2019.102356] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 01/02/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by antinuclear antibody (ANA) production. ANAs bind to DNA, RNA and complexes of proteins and nucleic acids and are important markers for diagnosis and activity. According to current models, ANAs originate from antigen-driven processes; nevertheless, antibody responses to both DNA and RNA binding proteins display features unexpected in terms of current paradigms for antigenicity. These differences may reflect disturbances in both B and T cells critical for autoreactivity. Clinically, ANA testing has new uses for determining classification as well as assessing eligibility for clinical trials. Studies of patients with established disease show frequent seronegativity. In this setting, seronegativity may indicate a stage of disease called post-autoimmunity in which the natural history of disease or effects of immunosuppressive therapies modifies responses. The new uses of ANA testing highlight the importance of understanding autoantigenicity and developing sensitive and informative assays for clinical assessments.
Collapse
Affiliation(s)
- David S Pisetsky
- Division of Rheumatology and Immunology, Duke University Medical Center and Medical Research Service, Durham Veterans Administration Medical Center, Durham, NC, USA.
| |
Collapse
|
39
|
Ghiggeri GM, D’Alessandro M, Bartolomeo D, Degl’Innocenti ML, Magnasco A, Lugani F, Prunotto M, Bruschi M. An Update on Antibodies to Necleosome Components as Biomarkers of Sistemic Lupus Erythematosus and of Lupus Flares. Int J Mol Sci 2019; 20:ijms20225799. [PMID: 31752186 PMCID: PMC6888059 DOI: 10.3390/ijms20225799] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/23/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with variable clinical expression. It is a potentially devastating condition affecting mostly women and leading to clinically unpredictable outcomes. Remission and flares may, in fact, alternate over time and a mild involvement limited to few articular sites may be followed by severe and widespread organ damage. SLE is the prototype of any autoimmune condition and has, for this reason, attracted the interest of basic immunologists. Therapies have evolved over time and clinical prognosis has, in parallel, been improved. What clinicians still lack is the possibility to use biomarkers of the disease as predictors of outcome and, in this area, several studies are trying to find solutions. Circulating autoantibodies are clearly a milestone of clinical research and the concrete possibility is to integrate, in the future, classical markers of activation (like C3) with target organ autoantibodies. Anti-dsDNA antibodies represent a basic point in any predictive attempt in SLE and should be considered the benchmark for any innovative proposal in the wide field of target organ pathologies related to SLE. DNA is part of the nucleosome that is the basic unit of chromatin. It consists of DNA wrapped around a histone octamer made of 2 copies each of Histone 2A, 2B, 3, and 4. The nucleosome has a plastic organization that varies over time and has the potential to stimulate the formation of antibodies directed to the whole structure (anti-nucleosome) or its parts (anti-dsDNA and anti-Histones). Here, we present an updated review of the literature on antibodies directed to the nucleosome and the nucleosome constituents, i.e., DNA and Histones. Wetriedto merge the data first published more than twenty years ago with more recent results to create a balanced bridge between old dogma and more recent research that could serve as a stimulus to reconsider mechanisms for SLE. The formation of large networks would provide the chance of studying large cohorts of patients and confirm what already presented in small sample size during the last years.
Collapse
Affiliation(s)
- Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, Istituto G. Gaslini, Largo G. Gaslini 5, 16147 Genoa, Italy (D.B.); (A.M.)
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS IstitutoGianninaGaslini, 16147 Genoa, Italy; (F.L.); (M.B.)
- Correspondence: ; Tel.: (+39)-010-380742; Fax: (+39)-010-395214
| | - Matteo D’Alessandro
- Division of Nephrology, Dialysis and Transplantation, Istituto G. Gaslini, Largo G. Gaslini 5, 16147 Genoa, Italy (D.B.); (A.M.)
| | - Domenico Bartolomeo
- Division of Nephrology, Dialysis and Transplantation, Istituto G. Gaslini, Largo G. Gaslini 5, 16147 Genoa, Italy (D.B.); (A.M.)
| | - Maria Ludovica Degl’Innocenti
- Division of Nephrology, Dialysis and Transplantation, Istituto G. Gaslini, Largo G. Gaslini 5, 16147 Genoa, Italy (D.B.); (A.M.)
| | - Alberto Magnasco
- Division of Nephrology, Dialysis and Transplantation, Istituto G. Gaslini, Largo G. Gaslini 5, 16147 Genoa, Italy (D.B.); (A.M.)
| | - Francesca Lugani
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS IstitutoGianninaGaslini, 16147 Genoa, Italy; (F.L.); (M.B.)
| | - Marco Prunotto
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland;
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, Scientific Institute for Research and Health Care, IRCCS IstitutoGianninaGaslini, 16147 Genoa, Italy; (F.L.); (M.B.)
- Fondazione per le MalattieRenalinel Bambino, 16100 Genoa, Italy
| |
Collapse
|
40
|
Nossent J, Raymond W, Ognjenovic M, Kang A, Chakera A, Wong D. The importance of tubuloreticular inclusions in lupus nephritis. Pathology 2019; 51:727-732. [PMID: 31668566 DOI: 10.1016/j.pathol.2019.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/23/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022]
Abstract
Tubuloreticular inclusions (TRI) are distinctive cytoplasmic structures of unknown origin that typically associate with autoimmune and viral diseases. We investigated the clinical and prognostic relevance of TRI detection in patients with lupus nephritis (LN). We conducted a single centre study of patients (n=84) with biopsy evidence of LN. Clinical variables included demographics, SLEDAI score, and autoantibody profiling; while histological evaluation included TRI presence, International Society of Nephrology/Renal Pathology Society (ISN/RPS) classification with NIH activity and chronicity indices, immunofluorescence, and other EM findings. Patients with and without TRI were compared by non-parametric statistical methods and survival analysis for the endpoints of death and renal failure. TRI were detected in 37 patients (44%) that were younger (28.4 vs 34.3 years, p=0.02) and more often from Asian background (37.8% vs 19.1%, p=0.04) compared to patients without TRI. SLEDAI score (11 vs 12 units, p=0.36) and amount of proteinuria (370 vs 340 mg/mmol, p=0.71) were similar in both groups; however, TRI positive patients had increased frequency of anti-SSB antibodies (16% vs 2%, p=0.02), 'full house' immune complex deposition (85% vs 58%, p=0.04) and subendothelial electron dense deposits (83% vs 65%, p=0.07), but were less often anti-dsDNA Ab positive (62% vs 85%, p=0.02). Patient and renal survival were not influenced by TRI status. TRI were observed in nearly half of all LN patients and TRI positive patients more often carried anti-SSB antibodies. However, TRI had little bearing on disease presentation or outcome in LN.
Collapse
Affiliation(s)
- Johannes Nossent
- School of Medicine, Faculty of Health Science, University of Western Australia, Crawley, WA, Australia; Department of Rheumatology, Sir Charles Gairdner Hospital, Perth, WA, Australia.
| | - Warren Raymond
- School of Medicine, Faculty of Health Science, University of Western Australia, Crawley, WA, Australia
| | - Milica Ognjenovic
- School of Medicine, Faculty of Health Science, University of Western Australia, Crawley, WA, Australia
| | - Alexandra Kang
- PathWest Laboratory Medicine, Anatomical Pathology, QEII Medical Centre, Perth, WA, Australia
| | - Aron Chakera
- School of Medicine, Faculty of Health Science, University of Western Australia, Crawley, WA, Australia; Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Daniel Wong
- PathWest Laboratory Medicine, Anatomical Pathology, QEII Medical Centre, Perth, WA, Australia
| |
Collapse
|
41
|
Ahn SS, Jung SM, Yoo J, Lee SW, Song JJ, Park YB. Anti-Smith antibody is associated with disease activity in patients with new-onset systemic lupus erythematosus. Rheumatol Int 2019; 39:1937-1944. [PMID: 31552434 DOI: 10.1007/s00296-019-04445-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/11/2019] [Indexed: 10/26/2022]
Abstract
Although anti-Smith (Sm) antibody is a highly specific antibody for systemic lupus erythematosus (SLE), the significance of anti-Sm antibody in patients with SLE is unclear. This study aimed to evaluate the association between anti-Sm antibodies and disease activity in patients with new-onset SLE. We included patients who were tested for anti-Sm antibodies at SLE diagnosis and within 12 months after diagnosis. SLE disease activity index (SLEDAI) was obtained at the time of the anti-Sm antibody test. The baseline disease activity was compared between patients with and without anti-Sm antibodies. The longitudinal association between disease activity and anti-Sm antibodies was also evaluated in total patients and in those with anti-Sm antibodies. Among 92 patients who were tested for anti-Sm antibodies at SLE diagnosis, 67 and another 67 patients were followed up for the presence of anti-Sm antibodies at 6 and 12 months, respectively. Although the baseline SLEDAI was comparable in patients with and without anti-Sm antibodies, the serum level of anti-Sm antibody was significantly correlated with SLEDAI (P = 0.003). At 12 months, anti-Sm antibody positivity was associated with higher SLEDAI and anti-dsDNA titer (P = 0.002, both). In addition, the changes in anti-Sm antibody titer over 12 months were correlated with the alterations in SLEDAI (P = 0.029).Anti-Sm antibody was associated with the baseline disease activity and the alteration of disease activity in patients with new-onset SLE. Monitoring of anti-Sm antibody titer may help assess the disease activity in SLE.
Collapse
Affiliation(s)
- Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Juyoung Yoo
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
42
|
Wang X, Xia Y. Anti-double Stranded DNA Antibodies: Origin, Pathogenicity, and Targeted Therapies. Front Immunol 2019; 10:1667. [PMID: 31379858 PMCID: PMC6650533 DOI: 10.3389/fimmu.2019.01667] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 07/03/2019] [Indexed: 01/02/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by high-titer serological autoantibodies, including antibodies that bind to double-stranded DNA (dsDNA). The origin, specificity, and pathogenicity of anti-dsDNA antibodies have been studied from a wider perspective. These autoantibodies have been suggested to contribute to multiple end-organ injuries, especially to lupus nephritis, in patients with SLE. Moreover, serum levels of anti-DNA antibodies fluctuate with disease activity in patients with SLE. By directly binding to self-antigens or indirectly forming immune complexes, anti-dsDNA antibodies can accumulate in the glomerular and tubular basement membrane. These autoantibodies can also trigger the complement cascade, penetrate into living cells, modulate gene expression, and even induce profibrotic phenotypes of renal cells. In addition, the expression of suppressor of cytokine signaling 1 is reduced by anti-DNA antibodies simultaneously with upregulation of profibrotic genes. Anti-dsDNA antibodies may even participate in the pathogenesis of SLE by catalyzing hydrolysis of certain DNA molecules or peptides in cells. Recently, anti-dsDNA antibodies have been explored in greater depth as a therapeutic target in the management of SLE. A substantial amount of data indicates that blockade of pathogenic anti-dsDNA antibodies can prevent or even reverse organ damage in murine models of SLE. This review focuses on the recent research advances regarding the origin, specificity, classification, and pathogenicity of anti-dsDNA antibodies and highlights the emerging therapies associated with them.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
43
|
Rekvig OP. The dsDNA, Anti-dsDNA Antibody, and Lupus Nephritis: What We Agree on, What Must Be Done, and What the Best Strategy Forward Could Be. Front Immunol 2019; 10:1104. [PMID: 31156647 PMCID: PMC6529578 DOI: 10.3389/fimmu.2019.01104] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/30/2019] [Indexed: 12/19/2022] Open
Abstract
This study aims to understand what lupus nephritis is, its origin, clinical context, and its pathogenesis. Truly, we encounter many conceptual and immanent tribulations in our attempts to search for the pathogenesis of this disease—and how to explain its assumed link to SLE. Central in the present landscape stay a short history of the early studies that substantiated the structures of isolated or chromatin-assembled mammalian dsDNA, and its assumed, highly controversial role in induction of anti-dsDNA antibodies. Arguments discussed here may provoke the view that anti-dsDNA antibodies are not what we think they are, as they may be antibodies operational in quite different biological contexts, although they bind dsDNA by chance. This may not mean that these antibodies are not pathogenic but they do not inform how they are so. This theoretical study centers the content around the origin and impact of extra-cellular DNA, and if dsDNA has an effect on the adaptive immune system. The pathogenic potential of chromatin-anti-dsDNA antibody interactions is limited to incite lupus nephritis and dermatitis which may be linked in a common pathogenic process. These are major criteria in SLE classification systems but are not shared with other defined manifestations in SLE, which may mean that they are their own disease entities, and not integrated in SLE. Today, the models thought to explain lupus nephritis are divergent and inconsistent. We miss a comprehensive perspective to try the different models against each other. To do this, we need to take all elements of the syndrome SLE into account. This can only be achieved by concentrating on the interactions between autoimmunity, immunopathology, deviant cell death and necrotic chromatin in context of elements of system science. System science provides a framework where data generated by experts can be compared, and tested against each other. This approach open for consensus on central elements making up “lupus nephritis” to separate what we agree on and how to understand the basis for conflicting models. This has not been done yet in a systematic context.
Collapse
Affiliation(s)
- Ole Petter Rekvig
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
44
|
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease whose pathogenesis can be conceptualized by a model based on a central role for immune complexes (ICs) between antinuclear antibodies and nucleic acids. According to this model, ICs can promote pathogenesis by two main mechanisms: deposition in the tissue to incite local inflammation and interaction with cells of the innate immune system to stimulate the production of cytokines, most prominently type 1 interferon. The latter stimulation results from the uptake of DNA and RNA in the form of ICs into cells and subsequent signaling by internal nucleic acid sensors for DNA and RNA. These sensors are likely important for the response to intracellular infection, although they may also be triggered during cell stress or injury by DNA or RNA aberrantly present in the cytoplasm. For IC formation, a source of extracellular DNA and RNA is essential. The current model of SLE posits that cell death is the origin of the nucleic acids in the ICs and that impairment of clearance mechanisms increases the amount of nuclear material in the extracellular space. This model of SLE is important since it points to new approaches to therapy; agents targeting interferon or the interferon receptor are examples of therapeutic approaches derived from this model. Future studies will explore novel biomarkers to monitor the operation of these mechanisms and to elucidate other steps in pathogenesis that can be targeted for therapy.
Collapse
Affiliation(s)
- David S Pisetsky
- Department of Medicine and Immunology, Duke University Medical Center and Medical Research Service, VA Medical Center, Durham, NC, USA
| |
Collapse
|
45
|
Pasha HF, Tantawy EA, Youssef MA. Osteopontin and interleukin-17A genes polymorphisms in Egyptian systemic lupus erythematosus patients: A relation to disease activity and severity. Gene 2019; 702:107-113. [PMID: 30898714 DOI: 10.1016/j.gene.2019.02.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022]
Abstract
Osteopontin (OPN) is involved in the regulation of the immune response and is accused in the pathogenesis of several autoimmune diseases including systemic lupus erythematosus (SLE). An obvious link between OPN and T cells, particularly T helper 17 cells is reported, where OPN produced by dendritic cells supports interleukin-17 (IL-17) expression, contributing to pathology of autoimmune disorders. The aim of the study was to investigate the association of genotypes and alleles frequencies of OPN 9250 (rs1126616) and IL-17A 197 (rs2275913) genes polymorphisms with their serum levels, susceptibility, disease activity and severity in Egyptian SLE patients. A total of 80 SLE patients and 80 healthy subjects were enrolled. The PCR-RFLP technique was used to detect OPN 9250 C/T and IL-17A 197 G/A genes polymorphisms. Serum OPN and IL- 17 levels were measured by the enzyme-linked immunosorbent assay. OPN TT genotype and T allele were significantly detected in SLE patients more than controls (P = 0.003, P < 0.001 respectively). IL-17A AA genotype showed non-significant higher frequency in SLE patients than in their controls (P = 0.07). While only the A allele of IL-17A polymorphism was significantly elevated in patients (P = 0.048). There was statistical significant association between OPN CT and TT genotypes and both renal and mucocutaneous manifestations. Also IL-17A AG and AA genotypes was significantly associated with renal, mucocutaneous in addition to the hematological manifestations. Serum OPN levels were significantly increased with TT genotype while serum IL-17 levels were significantly increased with AA genotype. Disease activity and severity scores were significantly elevated with both OPN TT and IL-17A AA genotypes. In conclusion, OPN 9250 C/T and IL-17A 197 G/A genes polymorphisms and their serum levels seemed to have a role in pathogenesis of SLE.
Collapse
Affiliation(s)
- Heba F Pasha
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Enas A Tantawy
- Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Manal A Youssef
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
46
|
Abstract
Connective tissue diseases (CTDs), also known as systemic autoimmune diseases, involve a variety of autoantibodies against cellular components. An important factor regarding these autoantibodies is that each antibody is exclusively related to a certain clinical feature of the disease type, which may prove useful in clinical practice. Thus far, more than 100 types of autoantibodies have been found in CTDs, and most of their target antigens have been identified. Many of these autoantigens are enzymes or regulators involved in important cellular functions, such as gene replication, transcription, repair/recombination, RNA processing, and protein synthesis, as well as proteins that form complexes with RNA and DNA. This article reviews the autoantibodies for each CTD, along with an assessment of their clinical significance, and provides suggestions regarding their utilization for clinical practice.
Collapse
Affiliation(s)
- Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
47
|
Rauch J, Salem D, Subang R, Kuwana M, Levine JS. β2-Glycoprotein I-Reactive T Cells in Autoimmune Disease. Front Immunol 2018; 9:2836. [PMID: 30619248 PMCID: PMC6295551 DOI: 10.3389/fimmu.2018.02836] [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: 05/17/2018] [Accepted: 11/16/2018] [Indexed: 01/15/2023] Open
Abstract
Anti-phospholipid syndrome (APS) and systemic lupus erythematosus (SLE) are autoimmune diseases characterized by autoantibody production and autoantibody-related pathology. Anti-phospholipid antibodies (aPL) are found in all patients with APS and in 20-30% of individuals with SLE. aPL recognize a number of autoantigens, but the primary target in both APS and SLE is β2-glycoprotein I (β2GPI). The production of IgG aPL in APS and SLE, as well as the association of aPL with certain MHC class II molecules, has led to investigation of the role of β2GPI-reactive T helper (Th). β2GPI-reactive CD4 Th cells have been associated with the presence of aPL and/or APS in both primary APS and secondary APS associated with SLE, as well as in SLE patients and healthy controls lacking aPL. CD4 T cells reactive with β2GPI have also been associated with atherosclerosis and found within atherosclerotic plaques. In most cases, the epitopes targeted by autoreactive β2GPI-reactive CD4 T cells in APS and SLE appear to arise as a consequence of antigenic processing of β2GPI that is structurally different from the soluble native form. This may arise from molecular interactions (e.g., with phospholipids), post-translational modification (e.g., oxidation or glycation), genetic alteration (e.g., β2GPI variants), or molecular mimicry (e.g., microbiota). A number of T cell epitopes have been characterized, particularly in Domain V, the lipid-binding domain of β2GPI. Possible sources of negatively charged lipid that bind β2GPI include oxidized LDL, activated platelets, microbiota (e.g., gut commensals), and dying (e.g., apoptotic) cells. Apoptotic cells not only bind β2GPI, but also express multiple other cellular autoantigens targeted in both APS and SLE. Dying cells that have bound β2GPI thus provide a rich source of autoantigens that can be recognized by B cells across a wide range of autoantigen specificities. β2GPI-reactive T cells could potentially provide T cell help to autoantigen-specific B cells that have taken up and processed apoptotic (or other dying) cells, and subsequently present β2GPI on their surface in the context of major histocompatibility complex (MHC) class II molecules. Here, we review the literature on β2GPI-reactive T cells, and highlight findings supporting the hypothesis that these T cells drive autoantibody production in both APS and SLE.
Collapse
Affiliation(s)
- Joyce Rauch
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - David Salem
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Rebecca Subang
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Jerrold S Levine
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago and Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
48
|
Srdic-Rajic T, Kohler H, Jurisic V, Metlas R. Antibody Epitope Specificity for dsDNA Phosphate Backbone Is an Intrinsic Property of the Heavy Chain Variable Germline Gene Segment Used. Front Immunol 2018; 9:2378. [PMID: 30405605 PMCID: PMC6200867 DOI: 10.3389/fimmu.2018.02378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/25/2018] [Indexed: 11/29/2022] Open
Abstract
Analysis of protein sequences by the informational spectrum method (ISM) enables characterization of their specificity according to encoded information represented with defined frequency (F). Our previous data showed that F(0.367) is characteristic for variable heavy chain (VH) domains (a combination of variable (V), diversity (D) and joining (J) gene segments) of the anti-phosphocholine (PC) T15 antibodies and mostly dependent on the CDR2 region, a site for PC phosphate group binding. Because the T15 dsDNA-reactive U4 mutant also encodes F(0.367), we hypothesized that the same frequency may also be characteristic for anti-DNA antibodies. Data obtained from an analysis of 60 spontaneously produced anti-DNA antibody VH domain sequences supported our hypothesis only for antibodies, which use V gene segment in germline configuration, such as S57(VH31), MRL-DNA22, and VH11, members of the VH1 (J558) and VH7 (S107) gene families. The important finding is that out of seven V gene segments used by spontaneous anti-DNA antibodies, F(0.367) is only expressed by the germline configuration of these three V gene segments. The data suggest that antibody specificity for the phosphate group moiety delineated as F(0.367) is the intrinsic property of the V germline gene segments used, whereas paratope/epitope interaction with antigens bearing this epitope, such as PC or dsDNA, requires corresponding antibody VH conformation that is susceptible to somatic mutation(s).
Collapse
Affiliation(s)
- Tatjana Srdic-Rajic
- Department of Experimental Pharmacology, National Cancer Research Center, Belgrade, Serbia
| | - Heinz Kohler
- Department of Microbiology and Immunology, University of Kentucky, Lexington, KY, United States
| | - Vladimir Jurisic
- Faculties of Medicinal Science, University of Kragujevac, Kragujevac, Serbia
| | - Radmila Metlas
- Vinča Institute of Nuclear Science, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
49
|
Wang JJ, Colella AD, Beroukas D, Chataway TK, Gordon TP. Precipitating anti-dsDNA peptide repertoires in lupus. Clin Exp Immunol 2018; 194:273-282. [PMID: 30086185 DOI: 10.1111/cei.13197] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2018] [Indexed: 12/15/2022] Open
Abstract
Anti-double-stranded (ds)DNA autoantibodies are prototypical serological markers of systemic lupus erythematosus (SLE), but little is known about their immunoglobulin variable (IgV) region composition at the level of the secreted (serum) proteome. Here, we use a novel proteomic workflow based on de novo mass spectrometric sequencing of anti-dsDNA precipitins to analyse IgV subfamily expression and mutational signatures of high-affinity, precipitating anti-dsDNA responses. Serum anti-dsDNA proteomes were oligoclonal with shared (public) expression of immunoglobulin (Ig)G heavy chain variable region (IGHV) and kappa chain variable region (IGKV) subfamilies. IgV peptide maps from eight subjects showed extensive public and random (private) amino acid replacement mutations with prominent arginine substitutions across heavy (H)- and light (L)-chains. Shared sets of L-chain complementarity determining region 3 (CDR3) peptides specified by arginine substitutions were sequenced from the dominantly expressed IGKV3-20 subfamily, with changes in expression levels of a clonal L-chain CDR3 peptide by quantitative multiple reaction monitoring (MRM) paralleling the rise and fall of anti-dsDNA levels by Farr radioimmunoassays (RIA). The heavily mutated IgV peptide signatures of precipitating anti-dsDNA autoantibody proteomes reflect the strong selective forces that shape humoral anti-dsDNA responses in germinal centres. Direct sequencing of agarose gel precipitins using microlitre volumes of stored sera streamlines the antibody sequencing workflow and is generalizable to other precipitating serum antibodies.
Collapse
Affiliation(s)
- J J Wang
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - A D Colella
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - D Beroukas
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - T K Chataway
- Flinders Proteomics Facility, Flinders University, Bedford Park, SA, Australia
| | - T P Gordon
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| |
Collapse
|
50
|
Soni C, Reizis B. DNA as a self-antigen: nature and regulation. Curr Opin Immunol 2018; 55:31-37. [PMID: 30261321 DOI: 10.1016/j.coi.2018.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/11/2018] [Indexed: 12/12/2022]
Abstract
High-affinity antibodies to double-stranded DNA are a hallmark of systemic lupus erythematosus (SLE) and are thought to contribute to disease flares and tissue inflammation such as nephritis. Notwithstanding their clinical importance, major questions remain about the development and regulation of these pathogenic anti-DNA responses. These include the mechanisms that prevent anti-DNA responses in healthy subjects, despite the constant generation of self-DNA and the abundance of DNA-reactive B cells; the nature and physical form of antigenic DNA in SLE; the regulation of DNA availability as an antigen; and potential therapeutic strategies targeting the pathogenic DNA in SLE. This review summarizes current progress in these directions, focusing on the role of secreted DNases in the regulation of antigenic extracellular DNA.
Collapse
Affiliation(s)
- Chetna Soni
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|