1
|
Fiorelli D, Caruso V, Belardi R, Bernardini S, Nuccetelli M. Evaluation of autoantibody profile in healthy subjects after mRNA vaccination against COVID-19. Int Immunopharmacol 2023; 122:110592. [PMID: 37413933 DOI: 10.1016/j.intimp.2023.110592] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/16/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND SARS-CoV-2 severe acute respiratory syndrome has rapidly spread worldwide since 2019. All scientific and technological forces have concentrated towards the formulation of vaccines to contain the disease. In less than one year (December 2020) a first messenger RNA vaccine (Comirnaty, BioNTech/Pfizer) was authorized. However, the research community has wondered about possible side effects on the immune system, given the vaccines administration in phase 4. AIM This study aims to evaluate the mRNA vaccine impact on the development of possible positive autoantibody profile in healthcare workers without any previous underlying pathology, after first, second and booster dose of Pfizer vaccine, by determining: circulating immune complexes concentrations (CIC); anti-myeloperoxidase (MPO) and anti-proteinase 3 (PR3) autoantibodies, the presence of antinuclear antibodies (ANA) and subsequent second level tests (extractable nuclear antigen (ENA) screen, double-strand DNA, extractable nuclear antigen (ANA) profile). METHODS The subjects were divided according to anti-SARS-CoV-2 IgG RBD antibodies increasing concentrations in: Group I < 10 BAU/ml (N = 114); Group II > 1000 BAU/ml (N = 112); Group III > 2500 BAU/ml (N = 78). RESULTS Our data show no autoreactive response changes over time in healthy subjects after vaccination. In fact, evaluation of ANA, CIC, anti-MPO, anti-PR3 and the detection of specific autoantigens, did not display significant variations. CONCLUSIONS The results suggest the exclusion of a correlation between the administration of the vaccine and the possible onset of autoimmune disorders. Nevertheless, further investigations will be needed to test for any long-term side effects on an ever-growing population.
Collapse
Affiliation(s)
- Denise Fiorelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy.
| | - Vincenza Caruso
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Riccardo Belardi
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| |
Collapse
|
2
|
Zuo Y, Navaz S, Tsodikov A, Kmetova K, Kluge L, Ambati A, Hoy CK, Yalavarthi S, de Andrade D, Tektonidou MG, Sciascia S, Pengo V, Ruiz-Irastorza G, Michael Belmont H, Gerosa M, Fortin PR, de Jesus GR, Ware Branch D, Andreoli L, Rodriguez-Almaraz E, Petri M, Cervera R, Willis R, Karp DR, Li QZ, Cohen H, Bertolaccini ML, Erkan. D, Knight JS. Anti-Neutrophil Extracellular Trap Antibodies in Antiphospholipid Antibody-Positive Patients: Results From the Antiphospholipid Syndrome Alliance for Clinical Trials and InternatiOnal Networking Clinical Database and Repository. Arthritis Rheumatol 2023; 75:1407-1414. [PMID: 36862141 PMCID: PMC10758259 DOI: 10.1002/art.42489] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/18/2023] [Accepted: 02/28/2023] [Indexed: 03/03/2023]
Abstract
OBJECTIVE This study aimed to elucidate the presence, antigen specificities, and potential clinical associations of anti-neutrophil extracellular trap (anti-NET) antibodies in a multinational cohort of antiphospholipid (aPL) antibody-positive patients who did not have lupus. METHODS Anti-NET IgG/IgM levels were measured in serum samples from 389 aPL-positive patients; 308 patients met the classification criteria for antiphospholipid syndrome. Multivariate logistic regression with best variable model selection was used to determine clinical associations. For a subset of the patients (n = 214), we profiled autoantibodies using an autoantigen microarray platform. RESULTS We found elevated levels of anti-NET IgG and/or IgM in 45% of the aPL-positive patients. High anti-NET antibody levels are associated with more circulating myeloperoxidase (MPO)-DNA complexes, which are a biomarker of NETs. When considering clinical manifestations, positive anti-NET IgG was associated with lesions affecting the white matter of the brain, even after adjusting for demographic variables and aPL profiles. Anti-NET IgM tracked with complement consumption after controlling for aPL profiles; furthermore, patient serum samples containing high levels of anti-NET IgM efficiently deposited complement C3d on NETs. As determined by autoantigen microarray, positive testing for anti-NET IgG was significantly associated with several autoantibodies, including those recognizing citrullinated histones, heparan sulfate proteoglycan, laminin, MPO-DNA complexes, and nucleosomes. Anti-NET IgM positivity was associated with autoantibodies targeting single-stranded DNA, double-stranded DNA, and proliferating cell nuclear antigen. CONCLUSION These data reveal high levels of anti-NET antibodies in 45% of aPL-positive patients, where they potentially activate the complement cascade. While anti-NET IgM may especially recognize DNA in NETs, anti-NET IgG species appear to be more likely to target NET-associated protein antigens.
Collapse
Affiliation(s)
- Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sherwin Navaz
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alex Tsodikov
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Katarina Kmetova
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Lyndsay Kluge
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amala Ambati
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Claire K. Hoy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | | - D. Ware Branch
- University of Utah and Intermountain Healthcare, Salt Lake City, UT, USA
| | | | | | - Michelle Petri
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ricard Cervera
- Hospital Clinic de Barcelona, Barcelona, Catalonia, Spain
| | - Rohan Willis
- University of Texas Medical Branch, Galveston, TX, USA
| | - David R. Karp
- Division of Rheumatic Disease, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Quan-Zhen Li
- Department of Immunology, Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hannah Cohen
- Haemostasis Research Unit, Department of Haematology, University College London, London, UK
| | | | - Doruk Erkan.
- Barbara Volcker Center for Women and Rheumatic Disease, Hospital for Special Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Gong M, Dai L, Xie Z, Hong D, Li N, Fan X, Xie C. Serological and clinical associations of autoantibodies in Chinese patients with new-onset systemic lupus erythematosus. Sci Rep 2023; 13:10101. [PMID: 37344560 DOI: 10.1038/s41598-023-37100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
To study the clinical significance of autoantibodies in Chinese patients with new-onset systemic lupus erythematosus (SLE), we enrolled 526 new-onset patients who met the 1997 Updated American College of Rheumatology SLE Classification Criteria for a retrospective cohort study. Chi-square test and Wilcoxon rank-sum test were used to detect the relationship of autoantibodies with clinical manifestations and serological results respectively. Our results demonstrated that the positive rate of anti-ribosomal P protein (anti-P) antibody in female patients was higher than that in male patients (41.2% vs. 22%, P = 0.008). Patients with anti-SSB (43.95 ± 73.12 vs. 40.92 ± 75.75, P = 0.004; 63.93 ± 103.56 vs. 55.06 ± 120.84, P = 0.008 respectively) antibodies had higher levels of alanine aminotransferase (ALT) and aspartate transaminase (AST), whereas those with anti-P antibody (28.90 ± 25.70 vs. 50.08 ± 93.00, P = 0.014; 38.51 ± 48.19 vs. 69.95 ± 142.67, P = 0.047, respectively) had lower levels of them. Anti-dsDNA antibody (P = 0.021) was associated with pulmonary arterial hypertension (PAH). The patients with anti-Ro60 (P = 0.044), anti-P (P = 0.012) and anti-dsDNA (P = 0.013) antibodies were less likely to develop Interstitial lung disease. Anti-SmRNP antibody was correlated to lower prevalence of neuropsychiatric symptoms (P = 0.037), and patients with anti-centromere antibody (ACA) were more likely to develop serositis (P = 0.016).We identified five clusters of SLE-related autoantibodies, confirmed previously reported associations of autoantibodies, and discovered new associations.
Collapse
Affiliation(s)
- Muxue Gong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Li Dai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Zhuobei Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Dengxiao Hong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Ning Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Xiaoyun Fan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Changhao Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233003, China.
| |
Collapse
|
4
|
Abstract
Anti-histone antibodies (AHAs) make their appearance in a number of systemic autoimmune diseases including systemic lupus erythematosus (SLE) and drug-induced lupus erythematosus (DILE). Although being known for over 50 years, they are poorly studied and understood. There is emerging evidence for their use in predicting clinical features of SLE, diversifying their clinical use. AHAs, however, are probably less prevalent in DILE than once thought owing to a move away from older DILE drugs to modern biological agents which do not appear to elicit AHAs. This review examines the historical studies that have defined AHAs and looks at some of the recent work with these autoantibodies.
Collapse
Affiliation(s)
- Adrian Y S Lee
- Department of Immunology, Westmead Hospital, Westmead, Australia.,ICPMR, NSW Health Pathology, Westmead, Australia.,Westmead Clinical School, University of Sydney, Westmead, Australia
| |
Collapse
|
5
|
Elsayed SAR, Kamaly HM, Esmail MA. Co-positivity of anti-dsDNA, anti-nucleosome, and anti-smith autoantibodies as serological biomarkers for disease activity in systemic lupus erythematosus. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2022. [DOI: 10.1186/s43166-021-00110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Evaluation of disease activity in systemic lupus erythematosus (SLE) patients is important for modulating the therapeutic plan and decreasing organ damage. Autoantibodies are important serological biomarkers in SLE. We evaluated the effect of co-positivity of anti-dsDNA, anti-nucleosome, and anti-smith, autoantibodies on the SLEDAI score in SLE patients.
Results
Eighty adult SLE patients were included in this study. The correlations of the three autoantibodies with the SLEDAI score in addition to their sensitivity and specificity for the assessment of disease activity were analyzed. There was a highly significant difference between anti-dsDNA, anti-nucleosome, and anti-smith positive and negative groups as regards the SLEDAI score. Increased number of autoantibody positivity was associated with an increased mean rank of SLEDAI, and the three autoantibodies were positively correlated with each other and with the SLEDAI score. Roc curve analysis revealed that anti-smith has the highest sensitivity (90%) followed by anti-dsDNA and anti-nucleosome (85% for each). Moreover, anti-dsDNA had the highest specificity (88%) followed by anti-nucleosome (86%) then anti-smith (84%).
Conclusions
Anti-dsDNA, anti-nucleosome, and anti-smith autoantibodies have a positive correlation with the SLEDAI score, and they may be considered as good serological biomarkers for the assessment of disease activity in SLE patients.
Collapse
|
6
|
Circulating Free DNA and Its Emerging Role in Autoimmune Diseases. J Pers Med 2021; 11:jpm11020151. [PMID: 33672659 PMCID: PMC7924199 DOI: 10.3390/jpm11020151] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
Liquid biopsies can be used to analyse tissue-derived information, including cell-free DNA (cfDNA), circulating rare cells, and circulating extracellular vesicles in the blood or other bodily fluids, representing a new way to guide therapeutic decisions in cancer. Among the new challenges of liquid biopsy, we found clinical application in nontumour pathologies, including autoimmune diseases. Since the discovery of the presence of high levels of cfDNA in patients with systemic lupus erythaematosus (SLE) in the 1960s, cfDNA research in autoimmune diseases has mainly focused on the overall quantification of cfDNA and its association with disease activity. However, with technological advancements and the increasing understanding of the role of DNA sensing receptors in inflammation and autoimmunity, interest in cfDNA and autoimmune diseases has not expanded until recently. In this review, we provide an overview of the basic biology of cfDNA in the context of autoimmune diseases as a biomarker of disease activity, progression, and prediction of the treatment response. We discuss and integrate available information about these important aspects.
Collapse
|
7
|
Umare V, Pradhan V, Nath S, Rajadhyaksha A, Ghosh K, Nadkarni AH. Impact of functional IL-18 polymorphisms on genetic predisposition and diverse clinical manifestations of the disease in Indian SLE patients. Lupus 2019; 28:545-554. [PMID: 30857465 DOI: 10.1177/0961203319834677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Several studies have demonstrated associations between interleukin-18 polymorphisms and risk of systemic lupus erythematosus in different populations except one of Indian origin. We therefore investigated for the influence of interleukin-18 (-1297T/C, -607A/C, -137G/C; + 105A/C) polymorphisms on genetic susceptibility and clinical expression of the disease in Indian systemic lupus erythematosus patients. A total of 200 systemic lupus erythematosus patients and 201 controls were recruited. Genotyping of interleukin-18 polymorphisms were performed by polymerase chain reaction-restriction fragment length polymorphism. Serum interleukin-18 levels were measured by enzyme-linked immunosorbent assay. Interleukin-18 (-1297T/C; -137G/C) polymorphisms showed significant association with genetic susceptibility to the disease in our systemic lupus erythematosus cohort. Stratification analysis revealed -1297CC and -1297C associated with renal involvement (odds ratio = 3.4, correcting p value = 0.0207), (odds ratio = 2.0, correcting p value = 0.0054) respectively. Additionally, -1297C allele frequency was significantly increased in patients with anti-nucleosome antibody (odds ratio = 2.1, correcting p value = 0.0301). Haplotype analysis showed CC haplotype strongly associated with serositis (odds ratio = 9.1, correcting p values = 0.0009) and neurologic involvement (odds ratio = 9.3, correcting p value = 0.0018). We reported a 2.7-fold increase in serum interleukin-18 levels in patients (511.5 ± 242.3 pg/ml) compared to controls (189.4 ± 80.8 pg/ml) ( p < 0.0001). Furthermore, interleukin-18 levels were positively correlated with disease activity ( r = 0.548, p = 0.0001) and renal involvement in the patients with lupus nephritis ( r = 0.569, p < 0.0001). In summary, interleukin-18 polymorphisms elucidated in this study appear to confer genetic susceptibility to the disease and are associated with renal, serositis and neurologic involvement in Indian systemic lupus erythematosus patients.
Collapse
Affiliation(s)
- V Umare
- 1 National Institute of Immunohaematology, Indian Council of Medical Research, Mumbai, India
| | - V Pradhan
- 1 National Institute of Immunohaematology, Indian Council of Medical Research, Mumbai, India
| | - S Nath
- 2 Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States of America
| | - A Rajadhyaksha
- 3 Department of Rheumatology, King Edward Memorial Hospital, Mumbai, India
| | - K Ghosh
- 1 National Institute of Immunohaematology, Indian Council of Medical Research, Mumbai, India
| | - A H Nadkarni
- 1 National Institute of Immunohaematology, Indian Council of Medical Research, Mumbai, India
| |
Collapse
|
8
|
Phenotyping of autoreactive B cells with labeled nucleosomes in 56R transgenic mice. Sci Rep 2017; 7:13232. [PMID: 29038433 PMCID: PMC5643551 DOI: 10.1038/s41598-017-13422-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/21/2017] [Indexed: 01/22/2023] Open
Abstract
The phenotypic characterization of self-reactive B cells producing autoantibodies is one of the challenges to get further insight in the physiopathology of autoimmune diseases. We took advantage of our previously developed flow cytometry method, using labeled nucleosomes, prominent autoantigens in systemic lupus erythematosus, to analyze the phenotype of self-reactive B cells in the anti-DNA B6.56R mouse model. We showed that splenic anti-nucleosome B cells express mostly kappa light chains and harbor a marginal zone phenotype. Moreover, these autoreactive B cells fail to acquire a germinal center phenotype and are less abundant in the transitional T3 compartment. In conclusion, the direct detection of autoreactive B cells helped determine their phenotypic characteristics and provided a more direct insight into the B cell tolerance process in B6.56R mice. This method constitutes an interesting new tool to study the mechanisms of B cell tolerance breakdown in B6.56R mice crossed with autoimmune prone models.
Collapse
|
9
|
Identification of autoreactive B cells with labeled nucleosomes. Sci Rep 2017; 7:602. [PMID: 28377609 PMCID: PMC5428865 DOI: 10.1038/s41598-017-00664-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/28/2017] [Indexed: 12/15/2022] Open
Abstract
The pathogenesis of autoimmune diseases has not been completely elucidated yet, and only a few specific treatments have been developed so far. In autoimmune diseases mediated by pathogenic autoantibodies, such as systemic lupus erythematosus, the specific detection and analysis of autoreactive B cells is crucial for a better understanding of the physiopathology. Biological characterization of these cells may help to define new therapeutic targets. Very few techniques allowing the precise detection of autoreactive B cells have been described so far. Herein we propose a new flow cytometry technique for specific detection of anti-nucleosome B cells, which secrete autoantibodies in systemic lupus erythematosus, using labeled nucleosomes. We produced different fluorochrome-labeled nucleosomes, characterized them, and finally tested them in flow cytometry. Nucleosomes labeled via the cysteines present in H3 histone specifically bind to autoreactive B cells in the anti-DNA transgenic B6.56R mice model. The present work validates the use of fluorochrome-labeled nucleosomes via cysteines to identify anti-nucleosome B cells and offers new opportunities for the description of autoreactive B cell phenotype.
Collapse
|
10
|
Proteolytic inactivation of nuclear alarmin high-mobility group box 1 by complement protease C1s during apoptosis. Cell Death Discov 2016; 2:16069. [PMID: 27648302 PMCID: PMC5018544 DOI: 10.1038/cddiscovery.2016.69] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 08/02/2016] [Indexed: 02/08/2023] Open
Abstract
Effective clearance of apoptotic cells by phagocytes prevents the release of intracellular alarmins and manifestation of autoimmunity. This prompt efferocytosis is complemented by intracellular proteolytic degradation that occurs within the apoptotic cells and in the efferosome of the phagocytes. Although the role of extracellular proteases in apoptotic cells clearance is unknown, the strong association of congenital C1s deficiency with Systemic Lupus Erythematosus highlights the protective nature that this extracellular protease has against autoimmunity. The archetypical role of serine protease C1s as the catalytic arm of C1 complex (C1qC1r2C1s2) involve in the propagation of the classical complement pathway could not provide the biological basis for this association. However, a recent observation of the ability of C1 complex to cleave a spectrum of intracellular cryptic targets exposed during apoptosis provides a valuable insight to the underlying protective mechanism. High-mobility group box 1 (HMGB1), an intracellular alarmin that is capable of inducing the formation of antinuclear autoantibodies and causes lupus-like conditions in mice, is identified as a novel potential target by bioinformatics analysis. This is verified experimentally with C1s, both in its purified and physiological form as C1 complex, cleaving HMGB1 into defined fragments of 19 and 12 kDa. This cleavage diminishes HMGB1 ability to enhance lipopolysaccharide mediated pro-inflammatory cytokines production from monocytes, macrophages and dendritic cells. Further mass spectrometric analysis of the C1 complex treated apoptotic cellular proteins demonstrated additional C1s substrates and revealed the complementary role of C1s in apoptotic cells clearance through the proteolytic cleavage of intracellular alarmins and autoantigens. C1 complex may have evolved as, besides the bacteriolytic arm of antibodies in which it activates the complement cascade, a tissue renewal mechanism that reduces the immunogenicity of apoptotic tissue debris and decreases the likelihood of autoimmunity.
Collapse
|
11
|
Gatto M, Iaccarino L, Ghirardello A, Punzi L, Doria A. Clinical and pathologic considerations of the qualitative and quantitative aspects of lupus nephritogenic autoantibodies: A comprehensive review. J Autoimmun 2016; 69:1-11. [DOI: 10.1016/j.jaut.2016.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 12/11/2022]
|
12
|
Steiman AJ, Urowitz MB, Ibañez D, Li TT, Gladman DD, Wither J. Anti-dsDNA and Antichromatin Antibody Isotypes in Serologically Active Clinically Quiescent Systemic Lupus Erythematosus. J Rheumatol 2015; 42:810-6. [PMID: 25729033 DOI: 10.3899/jrheum.140796] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Serologically active clinically quiescent (SACQ) patients with systemic lupus erythematosus (SLE) are clinically quiescent despite serologic activity. Since studies suggest that antichromatin antibodies are more sensitive than anti-dsDNA antibodies in detecting active SLE, and that immunoglobulin (Ig) G, in particular complement-fixing subclasses, may be more pathogenic than IgM, we investigated the levels of anti-dsDNA and antichromatin isotypes in SACQ patients as compared to non-SACQ patients with SLE. METHODS Levels of IgM, IgA, IgG, and IgG1-4 antichromatin and anti-dsDNA were measured by ELISA. SACQ was defined as ≥ 2 years with the SLE Disease Activity Index 2000 (SLEDAI-2K) at 2 or 4 from serologic activity, during which patients could be taking antimalarials, but not corticosteroids or immunosuppressives. Unselected non-SACQ patients with SLE were used as comparators. SACQ patient serum samples were further stratified based on subsequent development of flare, defined as clinical SLEDAI-2K ≥ 1 and/or treatment initiation. Nonparametric statistics were used, and generalized estimating equations were applied to account for multiple samples in the same patient. RESULTS SACQ patients' complement-fixing antichromatin and anti-dsDNA IgG subclasses were significantly higher than those of non-SACQ patients. When the sample drawn latest in a SACQ period was analyzed, there was no difference between antichromatin or anti-dsDNA isotype or IgG subclass levels between patients who flared and those who remained SACQ, nor were consistent trends seen when samples were examined during SACQ and flare in the same patient. CONCLUSION The SACQ phenotype does not arise from a lack of pathogenic anti-dsDNA and/or antichromatin autoantibodies. Neither increases in antichromatin nor anti-dsDNA isotype or IgG subclass levels were predictive of or coincident with flare in SACQ patients.
Collapse
Affiliation(s)
- Amanda J Steiman
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Murray B Urowitz
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Dominique Ibañez
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Timothy T Li
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Dafna D Gladman
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Joan Wither
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network.
| |
Collapse
|
13
|
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by the production of antinuclear antibodies (ANA) in association with protean clinic manifestations. ANA can bind to nuclear molecules, most prominently DNA and histones in nucleosomes, to form complexes to promote pathogenesis. Because of the intrinsic immunological activity of the nuclear components, these complexes can amplify responses by interacting with diverse pattern recognition receptors and internal sensing systems. Among molecules associated with nucleosomal components, HMGB1, a non-histone protein, can emanate from activated and dying cells; HMGB1's immune activity is determined by post-translational modifications, redox state, and binding to other immune mediators. Although ANAs form complexes that deposit in the kidney or induce type 1 interferon, ANAs may also block immune activity. Together, these studies highlight the importance of complexes in the pathogenesis of lupus and their role as antigens, immunogens, and adjuvants.
Collapse
Affiliation(s)
- David S Pisetsky
- Medical Research Service, Durham Veterans Administration Medical Center , Durham, NC , USA and
| |
Collapse
|
14
|
El Bakry SA, El Din AB, El Dakrony AHM, Abaza NM, Abo-Shady RA, Mohamed NA, Nada OH. Anti-nucleosome antibodies: A potential surrogate marker for renal affection in lupus patients with insignificant proteinuria. EGYPTIAN RHEUMATOLOGIST 2014. [DOI: 10.1016/j.ejr.2013.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
15
|
Dwivedi N, Neeli I, Schall N, Wan H, Desiderio DM, Csernok E, Thompson PR, Dali H, Briand JP, Muller S, Radic M. Deimination of linker histones links neutrophil extracellular trap release with autoantibodies in systemic autoimmunity. FASEB J 2014; 28:2840-51. [PMID: 24671707 DOI: 10.1096/fj.13-247254] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autoantibodies to nuclear antigens arise in human autoimmune diseases, but a unifying pathogenetic mechanism remains elusive. Recently we reported that exposure of neutrophils to inflammatory conditions induces the citrullination of core histones by peptidylarginine deiminase 4 (PAD4) and that patients with autoimmune disorders produce autoantibodies that recognize such citrullinated histones. Here we identify histone H1 as an additional substrate of PAD4, localize H1 within neutrophil extracellular traps, and detect autoantibodies to citrullinated H1 in 6% of sera from patients with systemic lupus erythematosus and Sjögren's syndrome. No preference for deiminated H1 was observed in healthy control sera and sera from patients with scleroderma or rheumatoid arthritis. We map binding to the winged helix of H1 and determine that citrulline 53 represents a key determinant of the autoantibody epitope. In addition, we quantitate RNA for H1 histone subtypes in mature human neutrophils and identify citrulline residues by liquid chromatography and tandem mass spectrometry. Our results indicate that deimination of linker histones generates new autoantibody epitopes with enhanced potential for stimulating autoreactive human B cells.-Dwivedi, N., Neeli, I., Schall, N., Wan, H., Desiderio, D. M., Csernok, E., Thompson, P. R., Dali, H., Briand, J.-P., Muller, S., Radic, M. Deimination of linker histones links neutrophil extracellular trap release with autoantibodies in systemic autoimmunity.
Collapse
Affiliation(s)
- Nishant Dwivedi
- Department of Microbiology, Immunology, and Biochemistry, and
| | - Indira Neeli
- Department of Microbiology, Immunology, and Biochemistry, and
| | - Nicolas Schall
- Immunopathology and Therapeutic Chemistry, Laboratory of Excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| | - Haibao Wan
- Department of Neurology, Charles B. Stout Neuroscience Mass Spectrometry Laboratory, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dominic M Desiderio
- Department of Neurology, Charles B. Stout Neuroscience Mass Spectrometry Laboratory, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Elena Csernok
- Department of Rheumatology, University of Lübeck, Klinikum Bad Bramstedt, Bad Bramstedt, Germany; and
| | | | - Hayet Dali
- Immunopathology and Therapeutic Chemistry, Laboratory of Excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| | - Jean-Paul Briand
- Immunopathology and Therapeutic Chemistry, Laboratory of Excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| | - Sylviane Muller
- Immunopathology and Therapeutic Chemistry, Laboratory of Excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| | - Marko Radic
- Department of Microbiology, Immunology, and Biochemistry, and
| |
Collapse
|
16
|
Pisetsky DS. Immune activation by histones: plusses and minuses in inflammation. Eur J Immunol 2013; 43:3163-6. [PMID: 24165954 DOI: 10.1002/eji.201344175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 11/05/2022]
Abstract
Histones are highly cationic proteins that are essential components of the cell nucleus, interacting with DNA to form the nucleosome and regulating transcription. Histones, however, can transit from the cell nucleus during cell death and, once in an extracellular location, can serve as danger signals and activate immune cells. An article in this issue of the European Journal of Immunology [Eur. J. Immunol. 2013. 43: 3336-3342] reports that histones can activate monocyte-derived DCs via the NRLP3 inflammasome to induce the production of IL-1β. As such, histones, which can also stimulate TLRs, may drive events in the immunopathogenesis of a wide range of acute and chronic diseases marked by sterile inflammation. While the mechanism of this stimulation is not known, the positive charge of histones may provide a structural element to promote interaction with cells and activation of downstream signaling systems.
Collapse
Affiliation(s)
- David S Pisetsky
- Department of Medicine, Medical Research Service, Durham Veterans Affairs Medical Center, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
17
|
Mackern-Oberti JP, Llanos C, Carreño LJ, Riquelme SA, Jacobelli SH, Anegon I, Kalergis AM. Carbon monoxide exposure improves immune function in lupus-prone mice. Immunology 2013; 140:123-32. [PMID: 23691924 PMCID: PMC3809712 DOI: 10.1111/imm.12124] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/23/2013] [Accepted: 05/16/2013] [Indexed: 01/09/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple alterations affecting the normal function of immune cells, such as lymphocytes, dendritic cells (DCs) and monocytes. Although the understanding of autoimmunity has significantly increased, the breakthrough in effective therapies has been modest, making necessary the development of new therapeutic strategies. Here we propose that a new potential target for therapy is haem oxygenase-1 (HO-1), an enzyme that catalyses the degradation of the haem group into biliverdin, carbon monoxide (CO) and Fe(2+) . These products exhibit immunosuppressive and anti-inflammatory effects, which can contribute to improving tolerance during organ transplantation. Because HO-1 is highly expressed by immune cells involved in SLE pathogenesis, such as monocytes and DCs, we evaluated whether induction of HO-1 expression or the administration of CO could ameliorate disease in the FcγRIIb knockout (KO) mouse model for SLE. We found that CO administration decreased the expansion of CD11b(+) cells, prevented the decline of regulatory T cells and reduced anti-histone antibodies observed in untreated FcγRIIb KO mice. Furthermore, CO-treated animals and HO-1 induction showed less kidney damage compared with untreated mice. These data suggest that HO-1 modulation and CO administration can ameliorate autoimmunity and prevent the lupus symptoms shown by FcγRIIb KO mice, highlighting HO-1 as a potential new target for autoimmune therapy.
Collapse
MESH Headings
- Animals
- Autoimmunity/drug effects
- CD11b Antigen/metabolism
- Carbon Monoxide/administration & dosage
- Disease Models, Animal
- Enzyme Induction/drug effects
- Female
- Heme Oxygenase-1/biosynthesis
- Kidney/drug effects
- Kidney/enzymology
- Kidney/pathology
- Lupus Erythematosus, Systemic/enzymology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/therapy
- Male
- Membrane Proteins/biosynthesis
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Spleen/immunology
- Spleen/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Juan P Mackern-Oberti
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
18
|
Huang H, Chen HW, Evankovich J, Yan W, Rosborough BR, Nace GW, Ding Q, Loughran P, Beer-Stolz D, Billiar TR, Esmon CT, Tsung A. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. THE JOURNAL OF IMMUNOLOGY 2013; 191:2665-79. [PMID: 23904166 DOI: 10.4049/jimmunol.1202733] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cellular processes that drive sterile inflammatory injury after hepatic ischemia/reperfusion (I/R) injury are not completely understood. Activation of the inflammasome plays a key role in response to invading intracellular pathogens, but mounting evidence suggests that it also plays a role in inflammation driven by endogenous danger-associate molecular pattern molecules released after ischemic injury. The nucleotide-binding domain, leucine-rich repeat containing protein 3 (NLRP3) inflammasome is one such process, and the mechanism by which its activation results in damage and inflammatory responses following liver I/R is unknown. In this article, we report that both NLRP3 and its downstream target caspase-1 are activated during I/R and are essential for hepatic I/R injury, because both NLRP3 and caspase-1 knockout mice are protected from injury. Furthermore, inflammasome-mediated injury is dependent on caspase-1 expression in liver nonparenchymal cells. Although upstream signals that activate the inflammasome during ischemic injury are not well characterized, we show that endogenous extracellular histones activate the NLRP3 inflammasome during liver I/R through TLR9. This occurs through TLR9-dependent generation of reactive oxygen species. This mechanism is operant in resident liver Kupffer cells, which drive innate immune responses after I/R injury by recruiting additional cell types, including neutrophils and inflammatory monocytes. These novel findings illustrate a new mechanism by which extracellular histones and activation of NLRP3 inflammasome contribute to liver damage and the activation of innate immunity during sterile inflammation.
Collapse
Affiliation(s)
- Hai Huang
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Blebs produced by actin-myosin contraction during apoptosis release damage-associated molecular pattern proteins before secondary necrosis occurs. Cell Death Differ 2013; 20:1293-305. [PMID: 23787996 PMCID: PMC3770329 DOI: 10.1038/cdd.2013.69] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/13/2013] [Accepted: 05/19/2013] [Indexed: 11/26/2022] Open
Abstract
Apoptosis is a fundamental homeostatic mechanism essential for the normal growth, development and maintenance of every tissue and organ. Dying cells have been defined as apoptotic by distinguishing features, including cell contraction, nuclear fragmentation, blebbing, apoptotic body formation and maintenance of intact cellular membranes to prevent massive protein release and consequent inflammation. We now show that during early apoptosis limited membrane permeabilization occurs in blebs and apoptotic bodies, which allows release of proteins that may affect the proximal microenvironment before the catastrophic loss of membrane integrity during secondary necrosis. Blebbing, apoptotic body formation and protein release during early apoptosis are dependent on ROCK and myosin ATPase activity to drive actomyosin contraction. We identified 231 proteins released from actomyosin contraction-dependent blebs and apoptotic bodies by adapted SILAC (stable isotope labeling with amino acids in cell culture) combined with mass spectrometry analysis. The most enriched proteins released were the nucleosomal histones, which have previously been identified as damage-associated molecular pattern proteins (DAMPs) that can initiate sterile inflammatory responses. These results indicate that limited membrane permeabilization occurs in blebs and apoptotic bodies before secondary necrosis, leading to acute and localized release of immunomodulatory proteins during the early phase of active apoptotic membrane blebbing. Therefore, the shift from apoptosis to secondary necrosis is more graded than a simple binary switch, with the membrane permeabilization of apoptotic bodies and consequent limited release of DAMPs contributing to the transition between these states.
Collapse
|
20
|
The constant region affects antigen binding of antibodies to DNA by altering secondary structure. Mol Immunol 2013; 56:28-37. [PMID: 23665381 DOI: 10.1016/j.molimm.2013.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 11/23/2022]
Abstract
We previously demonstrated an important role of the constant region in the pathogenicity of anti-DNA antibodies. To determine the mechanisms by which the constant region affects autoantibody binding, a panel of isotype-switch variants (IgG1, IgG2a, IgG2b) was generated from the murine PL9-11 IgG3 autoantibody. The affinity of the PL9-11 antibody panel for histone was measured by surface plasmon resonance (SPR). Tryptophan fluorescence was used to determine wavelength shifts of the antibody panel upon binding to DNA and histone. Finally, circular dichroism spectroscopy was used to measure changes in secondary structure. SPR analysis revealed significant differences in histone binding affinity between members of the PL9-11 panel. The wavelength shifts of tryptophan fluorescence emission were found to be dependent on the antibody isotype, while circular dichroism analysis determined that changes in antibody secondary structure content differed between isotypes upon antigen binding. Thus, the antigen binding affinity is dependent on the particular constant region expressed. Moreover, the effects of antibody binding to antigen were also constant region dependent. Alteration of secondary structures influenced by constant regions may explain differences in fine specificity of anti-DNA antibodies between antibodies with similar variable regions, as well as cross-reactivity of anti-DNA antibodies with non-DNA antigens.
Collapse
|
21
|
|
22
|
Thabet Y, Cañas F, Ghedira I, Youinou P, Mageed RA, Renaudineau Y. Altered patterns of epigenetic changes in systemic lupus erythematosus and auto-antibody production: is there a link? J Autoimmun 2012; 39:154-60. [PMID: 22709855 DOI: 10.1016/j.jaut.2012.05.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 05/20/2012] [Indexed: 02/07/2023]
Abstract
The prominent feature of immunological defects in systemic lupus erythematosus (SLE) is the production of autoantibodies (auto-Abs) to nuclear antigens including DNA, histones and RNP. In addition, there is growing evidence that epigenetic changes play a key role in the pathogenesis of SLE. Autoreactive CD4(+) T cells and B cells in patients with SLE have evidence of altered patterns of DNA methylation as well as post-translational modifications of histones and ribonucleoproteins (RNP). A key question that has emerged from these two characteristic features of SLE is whether the two processes are linked. New data provide support for such a link. For example, there is evidence that hypomethylated DNA is immunogenic, that anti-histone auto-Abs in patients with SLE bind epigenetic-sensitive hot spots and that epigenetically-modified RNP-derived peptides can modulate lupus disease. All in all, the available evidence indicates that a better understanding of dysregulation in epigenetics in SLE may offer opportunities to develop new biomarkers and novel therapeutic strategies.
Collapse
Affiliation(s)
- Yosra Thabet
- EA2216 Immunology, Pathology and Immunotherapy, European University of Brittany, Brest, France
| | | | | | | | | | | |
Collapse
|
23
|
Seavey MM, Lu LD, Stump KL, Wallace NH, Ruggeri BA. Novel, orally active, proteasome inhibitor, delanzomib (CEP-18770), ameliorates disease symptoms and glomerulonephritis in two preclinical mouse models of SLE. Int Immunopharmacol 2012; 12:257-70. [DOI: 10.1016/j.intimp.2011.11.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/20/2011] [Accepted: 11/29/2011] [Indexed: 10/14/2022]
|
24
|
Sifuentes Giraldo WA, Ahijón Lana M, García Villanueva MJ, González García C, Vázquez Diaz M. Chilblain lupus induced by TNF-α antagonists: a case report and literature review. Clin Rheumatol 2011; 31:563-8. [DOI: 10.1007/s10067-011-1924-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 12/16/2011] [Indexed: 11/25/2022]
|
25
|
Sardeto GA, Simas LM, Skare TS, Nisihara RM, Utiyama SRR. Antinucleosome in systemic lupus erythematosus. A study in a Brazilian population. Clin Rheumatol 2011; 31:553-6. [DOI: 10.1007/s10067-011-1889-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 10/04/2011] [Accepted: 10/28/2011] [Indexed: 10/15/2022]
|
26
|
Hung WT, Chen YM, Lan JL, Chen HH, Chen YH, Chen DY, Hsieh CW, Wen MC. Antinucleosome antibodies as a potential biomarker for the evaluation of renal pathological activity in patients with proliferative lupus nephritis. Lupus 2011; 20:1404-1410. [DOI: 10.1177/0961203311417033] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The objective of this study is to evaluate the correlation between antinucleosome antibodies and renal pathological activity in patients with proliferative lupus nephritis (LN). We evaluated 36 patients with proliferative LN, 14 non-renal lupus patients and 10 healthy volunteers. Lupus activity was assessed using the British Isles Lupus Assessment Group 2004 (BILAG 2004) index, serum anti-double stranded DNA (anti-dsDNA) levels, serum complement levels and daily urinary protein levels. All 36 lupus nephritis patients received renal biopsy. Antinucleosome antibodies were detected by enzyme-linked immunosorbent assay (ELISA). Our results showed that levels of serum antinucleosome antibodies were significantly higher in LN patients (median 90.35 units/ml, interquartile range [IQR] 37.38–135.23) than in non-renal SLE patients (median 5.45 units/ml, IQR 2.6–28.93, p <0.05) and in healthy volunteers (median 3.35 units/ml, IQR 2.95–5.23, p <0.001). Serum levels of antinucleosome antibodies were positively correlated with BILAG index (Spearman’s r = 0.645, p <0.001) and serum anti-dsDNA antibody levels ( rs = 0.644, p <0.01), while serum levels of antinucleosome antibodies were negatively correlated with serum levels of C3 ( rs = -0.400, p <0.01) and C4 ( rs = -0.300, p <0.05). Serum levels of antinucleosome antibodies were positively correlated with the histological activity index of LN ( rs = 0.368, p <0.05). However, there was no significant correlation between serum levels of antinucleosome antibodies and the histological chronicity index. In conclusion, the serum level of antinucleosome antibodies is a potential biomarker for early recognition of renal involvement and evaluation of disease activity in SLE. Our preliminary results suggested that serum levels of antinucleosome antibodies might be a potential biomarker in evaluating pathological activity of LN.
Collapse
Affiliation(s)
- WT Hung
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
| | - YM Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
| | - JL Lan
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
- Institute of Biomedical Science, National Chung-Hsing University, Taiwan
| | - HH Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
| | - YH Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
| | - DY Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
- Institute of Biomedical Science, National Chung-Hsing University, Taiwan
- School of Medicine, Chung-Shan Medical University, Taiwan
| | - CW Hsieh
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taiwan
- School of Medicine, Chung-Shan Medical University, Taiwan
| | - MC Wen
- Department of Pathology, Taichung Veterans General Hospital, Taiwan
| |
Collapse
|
27
|
Huang H, Evankovich J, Yan W, Nace G, Zhang L, Ross M, Liao X, Billiar T, Xu J, Esmon CT, Tsung A. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice. Hepatology 2011; 54:999-1008. [PMID: 21721026 PMCID: PMC3213322 DOI: 10.1002/hep.24501] [Citation(s) in RCA: 281] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
UNLABELLED Sterile inflammatory insults are known to activate innate immunity and propagate organ damage through the recognition of extracellular damage-associated molecular pattern (DAMP) molecules. Although DAMPs such as endogenous DNA and nuclear high-mobility group box 1 have been shown to be critical in sterile inflammation, the role of nuclear histone proteins has not yet been investigated. We report that endogenous histones function as DAMPs after ischemic injury through the pattern recognition receptor Toll-like receptor (TLR) 9 to initiate inflammation. Using an in vivo model of hepatic ischemia/reperfusion (I/R) injury, we show that levels of circulating histones are significantly higher after I/R, and that histone neutralization significantly protects against injury. Injection of exogenous histones exacerbates I/R injury through cytotoxic effects mediated by TLR9 and MyD88. In addition, histone administration increases TLR9 activation, whereas neither TLR9 nor MyD88 mutant mice respond to exogenous histones. Furthermore, we demonstrate in vitro that extracellular histones enhance DNA-mediated TLR9 activation in immune cells through a direct interaction. CONCLUSION These novel findings reveal that histones represent a new class of DAMP molecules and serve as a crucial link between initial damage and activation of innate immunity during sterile inflammation.
Collapse
Affiliation(s)
- Hai Huang
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - John Evankovich
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Wei Yan
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Gary Nace
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Lemeng Zhang
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Mark Ross
- Department of Cell Biology, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Xinghua Liao
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| | - Jun Xu
- Department of Cardiovascular Biology Research Program, Oklahoma Medical FoundationOklahoma City, OK
| | - Charles T Esmon
- Department of Cardiovascular Biology Research Program, Oklahoma Medical FoundationOklahoma City, OK,Howard Hughes Medical Institute, Departments of Pathology and Biochemistry and Molecular Biology, University of Oklahoma Health Sciences CenterOklahoma City, OK
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh Medical CenterPittsburgh, PA
| |
Collapse
|
28
|
Hedberg A, Mortensen ES, Rekvig OP. Chromatin as a target antigen in human and murine lupus nephritis. Arthritis Res Ther 2011; 13:214. [PMID: 21542875 PMCID: PMC3132027 DOI: 10.1186/ar3281] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The present review focuses on pathogenic molecular and transcriptional events in patients with lupus nephritis. These factors are renal DNaseI, exposed chromatin fragments and the corresponding chromatin-reactive autoantibodies. Lupus nephritis is the most serious complication in human systemic lupus erythematosus, and is characterised by deposition of chromatin fragment-IgG complexes in the mesangial matrix and glomerular basement membranes. The latter deposition defines end-stage disease. This event is stringently linked to a renal-restricted shutdown of expression of the DNaseI gene, as determined by loss of DNaseI mRNA level and DNaseI enzyme activity. The major aim of the present review is to generate new therapeutic strategies based on new insight into the disease pathogenesis.
Collapse
Affiliation(s)
- Annica Hedberg
- Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Science, University of Tromsø, N-9037 Tromsø, Norway
| | | | | |
Collapse
|
29
|
Qureshi IA, Mehler MF. Emerging role of epigenetics in stroke: part 1: DNA methylation and chromatin modifications. ACTA ACUST UNITED AC 2010; 67:1316-22. [PMID: 21060009 DOI: 10.1001/archneurol.2010.275] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Epigenetic mechanisms refer to the complex and interrelated molecular processes that dynamically modulate gene expression and function within every cell in the body. These regulatory systems represent the long-sought-after molecular interfaces that mediate gene × environment interactions. Changes in the epigenome throughout life are responsible not only for controlling normal development, adult homeostasis, and aging but also for mediating responses to injury. Emerging evidence implicates a spectrum of epigenetic processes in the pathophysiology of stroke. In this review, we describe conventional epigenetic mechanisms (including DNA methylation, histone code modifications, nucleosome remodeling, and higher-order chromatin formation) and highlight the emerging roles each of these processes play in the pathobiology of stroke. We suggest that understanding these mechanisms may be important for discovering more sensitive and specific biomarkers for risk, onset, and progression of stroke. In addition, we highlight epigenetic approaches for stroke therapy, including the inhibition of DNA methyltransferase and histone deacetylase enzyme activities. These therapeutic approaches are still in their infancy, but preliminary results suggest that contemporary agents targeting these pathways can regulate the deployment of stress responses that modulate neural cell viability and promote brain repair and functional reorganization. Indeed, these agents even appear to orchestrate sophisticated cognitive functions, including learning and memory.
Collapse
Affiliation(s)
- Irfan A Qureshi
- Rosyln and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Institute for Brain Disorders and Neural Regeneration, and Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
30
|
Arnson Y, Shoenfeld Y, Alon E, Amital H. The Antiphospholipid Syndrome as a Neurological Disease. Semin Arthritis Rheum 2010; 40:97-108. [DOI: 10.1016/j.semarthrit.2009.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 04/08/2009] [Accepted: 05/03/2009] [Indexed: 02/06/2023]
|
31
|
Pemberton AD, Brown JK, Inglis NF. Proteomic identification of interactions between histones and plasma proteins: implications for cytoprotection. Proteomics 2010; 10:1484-93. [PMID: 20127695 DOI: 10.1002/pmic.200900818] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Extracellular histones released from cells during acute inflammation contribute to organ failure and death in a mouse model of sepsis, and histones are known to exert in vitro cytotoxicity in the absence of serum. Since addition of histones to serum and plasma is known to induce protein aggregation, we reasoned that plasma proteins may afford protection from cytotoxicity. We found that MODE-K mouse small intestinal epithelial cells were protected from histone-induced toxicity in the presence of 10% FCS. Therefore, the main aim of this study was to identify histone-interacting plasma proteins that might be involved in cytoprotection. The precipitate formed following addition of calf thymus histones to human EDTA plasma was characterised by shotgun proteomics, identifying a total of 36 protein subunits, including complement components, coagulation factors, protease inhibitors and apolipoproteins. The highly sulphated glycosaminoglycan heparin inhibited histone-induced plasma protein aggregation. Moreover, histones bound to heparin agarose were capable of pulling down plasma proteins from solution, indicating their effective cross-linking properties. It was particularly notable that inter-alpha-trypsin inhibitor was prominent among the histone-precipitated proteins, since it contains a chondroitin sulphate glycan chain, and suggests a potential role for this protein in histone sequestration during acute inflammation in vivo.
Collapse
Affiliation(s)
- Alan D Pemberton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Veterinary Centre, Roslin, Midlothian, UK.
| | | | | |
Collapse
|
32
|
Mahler M, Fritzler MJ. Epitope specificity and significance in systemic autoimmune diseases. Ann N Y Acad Sci 2010; 1183:267-87. [DOI: 10.1111/j.1749-6632.2009.05127.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Kiss E, Lakos G, Szegedi G, Poor G, Szodoray P. Anti-nuscleosome antibody, a reliable indicator for lupus nephritis. Autoimmunity 2010; 42:393-8. [PMID: 19811255 DOI: 10.1080/08916930903002446] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Systemic lupus erythematosus (SLE) is characterized by the presence of antinuclear antibodies (ANA). ANAs are aimed against native DNA, as well as histone proteins and small nuclear ribonucleoproteins, also could be targeted against the entire chromatin, consisted of nucleosomes. Herein, we assessed the frequency and the concentration of anti-nucleosome antibodies (aNS) in a group of 107 Hungarian patients with SLE. Moreover, correlations between these autoantibody concentrations and disease activity, as well as organ manifestations, mainly renal symptoms were evaluated. The occurrence of positive aNS, anti-dsDNA and anti-histone (aH) antibodies were 39.2, 28.0 and 47.6%, respectively. All the three autoantibodies were represented with significantly higher titers in patients with lupus nephritis. A positive correlation was found between the concentration of these autoantibodies and disease activity. The long disease duration (mean 8.5 years) and consequent low disease activity (mean disease activity index: 3.28) could be the reason for the relatively low occurrence of ANAs amongst patients. Besides anti-dsDNA, the assessment of aNS and aH antibodies could be useful in the diagnosis and follow-up of SLE. The authors discuss the possible role of aNS autoantibodies in the pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Emese Kiss
- National Institute of Rheumatology and Physiotherapy, H-1525, Budapest, Hungary
| | | | | | | | | |
Collapse
|
34
|
Dhir V, Singh AP, Aggarwal A, Naik S, Misra R. Increased T-lymphocyte apoptosis in lupus correlates with disease activity and may be responsible for reduced T-cell frequency: a cross-sectional and longitudinal study. Lupus 2009; 18:785-91. [PMID: 19578102 DOI: 10.1177/0961203309103152] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Apoptosis of lymphocytes is increased in patients with lupus. This may be pathogenic leading to increased load of autoantigens or may be a bystander effect of immune activation. A major unresolved issue is whether apoptosis is related to disease activity. Also its association with lymphocyte frequencies, anti-nucleosomal antibodies and serum IL 10 levels needs to be explored further. The aims of this study are to measure T- and B-lymphocyte apoptosis in patients with lupus and look at the effect of disease activity in a cross-sectional and longitudinal design and to determine frequency of T and B cells, level of anti-nucleosomal antibodies and serum IL 10 and assess their relationship with apoptosis. This study included 41 patients with SLE and 20 controls. A cutoff value of 4 in systemic lupus erythematosus disease activity index (SLEDAI) was used to separate active from inactive SLE. The frequency and degree of apoptosis of T and B lymphocyte were enumerated by flow cytometry using peripheral blood mononuclear cells (PBMCs) stained with CD3/CD19 and annexin V/PI. The data for T/B cell frequency are represented as % of these cells in the PBMC population, whereas percentage of apoptotic cells is out of total T or B cells. Serum anti-nucleosomal antibodies and IL 10 were assayed using ELISA. A repeat assessment of these parameters was carried out in 11 active patients when they became inactive. We found higher T-lymphocyte apoptosis in patients with SLE versus controls (14.8 +/- 9.2, 7.2 +/- 3.0; P < 0.05) and a lower frequency of T cells (72.7 +/- 12.6, 79.9 +/- 5.8; P < 0.05). T-lymphocyte apoptosis was higher in patients with active disease compared with inactive (18.5 +/- 11.3, 11.6 +/- 5.4; P = 0.05). Further, T-lymphocyte apoptosis directly correlated with SLEDAI (r = 0.37, P < 0.05) and inversely with T-cell frequency (r = -0.29, P < 0.05). Anti-nucleosomal antibodies correlated with SLEDAI but not apoptosis. On longitudinal follow-up, a decline in T-cell apoptosis was seen in patients with SLE, however this was not statistically significant. We confirmed a higher degree of apoptosis in T-lymphocytes in patients with SLE and found a direct correlation of T-cell apoptosis with disease activity. Patients had reduced T-cell frequency, which inversely correlated with T-cell apoptosis and may suggest a cause-effect relationship.
Collapse
Affiliation(s)
- V Dhir
- Department of Immunology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | | | | | | | | |
Collapse
|
35
|
Faurschou M, Penkowa M, Andersen CB, Starklint H, Jacobsen S. Renal cell apoptosis in human lupus nephritis: a histological study. Lupus 2009; 18:994-9. [DOI: 10.1177/0961203309106175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nuclear autoantigens from apoptotic cells are believed to drive the immunological response in systemic lupus erythematosus (SLE). Conflicting data exist as to the possible renal origin of apoptotic cells in SLE patients with nephritis. We assessed the level of renal cell apoptosis in kidney biopsies from 35 patients with lupus nephritis by means of terminal deoxynucleotidyl-transferase (TdT)-mediated deoxyuridine triphosphate (dUTP)-digoxigenin nick end labeling (TUNEL). Five samples of normal kidney tissue served as control specimens. We did not observe apoptotic glomerular cells in any of the control or nephritis biopsies. Scarce apoptotic tubular cells were seen in 13 of 35 (37%) of the nephritis specimens and in two of five (40%) of the control sections. Within the SLE cohort, patients with TUNEL-positive tubular cells in their renal biopsies had significantly higher activity index scores for tubulointerstitial mononuclear cell infiltration than patients without apoptotic tubular cells in their biopsies ( P = 0.01). Furthermore, the level of tubular cell apoptosis displayed a statistically significant, positive correlation with the activity index score for mononuclear cell infiltration (rs = 0.472, P = 0.004) but not with scores for other activity or chronicity index components. These observations indicate that the degree of tubular cell apoptosis correlates with the severity of tubulointerstitial inflammation in SLE-associated nephritis. However, our findings do not suggest that apoptotic renal cells constitute a quantitatively important source of auto-antibody-inducing nuclear auto-antigens in human lupus nephritis.
Collapse
Affiliation(s)
- M Faurschou
- Department of Rheumatology, The National University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - M Penkowa
- Section of Neuroprotection, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - CB Andersen
- Department of Pathology, The National University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - H Starklint
- Department of Pathology, Vejle Hospital, Vejle, Denmark
| | - S Jacobsen
- Department of Rheumatology, The National University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|