1
|
Stockfelt M, Teng YKO, Vital EM. Opportunities and limitations of B cell depletion approaches in SLE. Nat Rev Rheumatol 2025; 21:111-126. [PMID: 39815102 DOI: 10.1038/s41584-024-01210-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2024] [Indexed: 01/18/2025]
Abstract
B cell depletion with rituximab, a chimeric monoclonal antibody that selectively targets B cells by binding CD20, has been used off label in severe and resistant systemic lupus erythematosus (SLE) for over two decades. Several biological mechanisms limit the efficacy of rituximab, including immunological reactions towards the chimeric molecule, increased numbers of residual B cells, including plasmablasts and plasma cells, and a post-treatment surge in B cell-activating factor (BAFF) levels. Consequently, rituximab induces remission in only a proportion of patients, and safety issues limit its use. However, the use of rituximab has established the value of B cell depletion strategies in SLE and has guided the development of several improved B cell depletion therapies for SLE. These include enhanced monoclonal antibodies, modalities that redirect the specificity of patient T cells using chimeric antigen receptor T cells or bispecific T cell engagers, and combination treatment that simultaneously inhibits the BAFF pathway. In this Review, we consider evidence gathered from over two decades of using rituximab in SLE and examine how B cell depletion therapies could be further optimized to achieve immunological and clinical efficacy. In addition, we discuss the prospects of B cell depletion strategies for personalized treatment in SLE based on genetic research and studies in pre-symptomatic individuals.
Collapse
Affiliation(s)
- Marit Stockfelt
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Y K Onno Teng
- Center of Expertise for Lupus, Vasculitis and Complement-mediated Systemic disease (LuVaCs), Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
2
|
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
|
3
|
Fava A, Wagner CA, Guthridge CJ, Kheir J, Macwana S, DeJager W, Gross T, Izmirly P, Belmont HM, Diamond B, Davidson A, Utz PJ, Weisman MH, Magder LS, Accelerating Medicines Partnership (AMP) RA/SLE Network, Guthridge JM, Petri M, Buyon J, James JA. Association of Autoantibody Concentrations and Trajectories With Lupus Nephritis Histologic Features and Treatment Response. Arthritis Rheumatol 2024; 76:1611-1622. [PMID: 38962936 PMCID: PMC11521769 DOI: 10.1002/art.42941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/22/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE Autoantibodies are a hallmark of lupus nephritis (LN), but their association with LN classes and treatment response are not adequately known. In this study, we quantified circulating autoantibodies in the Accelerating Medicines Partnership LN longitudinal cohort to identify serological biomarkers of LN histologic classification and treatment response and how these biomarkers change over time based on treatment response. METHODS Peripheral blood samples were collected from 279 patients with systemic lupus erythematosus undergoing diagnostic kidney biopsy based on proteinuria. Of these, 268 were diagnosed with LN. Thirteen autoantibody specificities were measured by bead-based assays (Bio-Rad Bioplex 2200) and anti-C1q by enzyme-linked immunosorbent assay at the time of biopsy (baseline) and at 3, 6, and 12 months after biopsy. Clinical response was determined at 12 months. RESULTS Proliferative LN (International Society of Nephrology/Renal Pathology Society class III/IV±V, n = 160) was associated with higher concentrations of anti-C1q, anti-chromatin, anti-double-stranded DNA (dsDNA), and anti-ribosomal P autoantibodies compared to nonproliferative LN (classes I/II/V/VI, n = 108). Anti-C1q and-dsDNA were independently associated with proliferative LN. In proliferative LN, higher baseline anti-C1q levels predicted complete response (area under the curve [AUC] 0.72; P = 0.002) better than baseline proteinuria (AUC 0.59; P = 0.21). Furthermore, all autoantibody levels except for anti-La/SSB decreased over 12 months in patients with proliferative, but not membranous, LN with a complete response. CONCLUSION Baseline levels of anti-C1q and anti-dsDNA may serve as noninvasive biomarkers of proliferative LN, and anti-C1q may predict complete response at the time of kidney biopsy. In addition, tracking autoantibodies over time may provide further insights into treatment response and pathogenic mechanisms in patients with proliferative LN.
Collapse
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Catriona A. Wagner
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Carla J. Guthridge
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Joseph Kheir
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Susan Macwana
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Wade DeJager
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Tim Gross
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Peter Izmirly
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York, NY
| | | | - Betty Diamond
- Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases Research, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
- Donald and Barbara Zucker School of Medicine, Northwell Health, Hempstead, NY
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael H Weisman
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Laurence S. Magder
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD
| | | | - Joel M. Guthridge
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Michelle Petri
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Jill Buyon
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
4
|
Strizzi CT, Ambrogio M, Zanoni F, Bonerba B, Bracaccia ME, Grandaliano G, Pesce F. Epitope Spreading in Immune-Mediated Glomerulonephritis: The Expanding Target. Int J Mol Sci 2024; 25:11096. [PMID: 39456878 PMCID: PMC11507388 DOI: 10.3390/ijms252011096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Epitope spreading is a critical mechanism driving the progression of autoimmune glomerulonephritis. This phenomenon, where immune responses broaden from a single epitope to encompass additional targets, contributes to the complexity and severity of diseases such as membranous nephropathy (MN), lupus nephritis (LN), and ANCA-associated vasculitis (AAV). In MN, intramolecular spreading within the phospholipase A2 receptor correlates with a worse prognosis, while LN exemplifies both intra- and intermolecular spreading, exacerbating renal involvement. Similarly, ANCA reactivity in AAV highlights the destructive potential of epitope diversification. Understanding these immunological cascades reveals therapeutic opportunities-targeting early epitope spreading could curb disease progression. Despite promising insights, the clinical utility of epitope spreading as a prognostic tool remains debated. This review provides a complete overview of the current evidence, exploring the dual-edged nature of epitope spreading, the intricate immune mechanisms behind it, and its therapeutic implications. By elucidating these dynamics, we aim to pave the way for more precise, targeted interventions in autoimmune glomerular diseases.
Collapse
Affiliation(s)
- Camillo Tancredi Strizzi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.A.); (B.B.); (G.G.)
- Nephrology, Dialysis and Transplantation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Martina Ambrogio
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.A.); (B.B.); (G.G.)
- Nephrology, Dialysis and Transplantation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesca Zanoni
- Department of Nephrology, Dialysis, and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Bibiana Bonerba
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.A.); (B.B.); (G.G.)
- Nephrology, Dialysis and Transplantation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Elena Bracaccia
- Division of Renal Medicine, Ospedale Isola Tiberina-Gemelli Isola, 00186 Rome, Italy;
| | - Giuseppe Grandaliano
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.A.); (B.B.); (G.G.)
- Nephrology, Dialysis and Transplantation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Pesce
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.A.); (B.B.); (G.G.)
- Division of Renal Medicine, Ospedale Isola Tiberina-Gemelli Isola, 00186 Rome, Italy;
| |
Collapse
|
5
|
Saavedra PHV, Trzeciak AJ, Lipshutz A, Daman AW, O'Neal AJ, Liu ZL, Wang Z, Romero-Pichardo JE, Rojas WS, Zago G, van den Brink MRM, Josefowicz SZ, Lucas CD, Anderson CJ, Rudensky AY, Perry JSA. Broad-spectrum antibiotics disrupt homeostatic efferocytosis. Nat Metab 2024; 6:1682-1694. [PMID: 39122784 PMCID: PMC7616532 DOI: 10.1038/s42255-024-01107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024]
Abstract
The clearance of apoptotic cells, termed efferocytosis, is essential for tissue homeostasis and prevention of autoimmunity1. Although past studies have elucidated local molecular signals that regulate homeostatic efferocytosis in a tissue2,3, whether signals arising distally also regulate homeostatic efferocytosis remains elusive. Here, we show that large peritoneal macrophage (LPM) display impairs efferocytosis in broad-spectrum antibiotics (ABX)-treated, vancomycin-treated and germ-free mice in vivo, all of which have a depleted gut microbiota. Mechanistically, the microbiota-derived short-chain fatty acid butyrate directly boosts efferocytosis efficiency and capacity in mouse and human macrophages, and rescues ABX-induced LPM efferocytosis defects in vivo. Bulk messenger RNA sequencing of butyrate-treated macrophages in vitro and single-cell messenger RNA sequencing of LPMs isolated from ABX-treated and butyrate-rescued mice reveals regulation of efferocytosis-supportive transcriptional programmes. Specifically, we find that the efferocytosis receptor T cell immunoglobulin and mucin domain containing 4 (TIM-4, Timd4) is downregulated in LPMs of ABX-treated mice but rescued by oral butyrate. We show that TIM-4 is required for the butyrate-induced enhancement of LPM efferocytosis capacity and that LPM efferocytosis is impaired beyond withdrawal of ABX. ABX-treated mice exhibit significantly worse disease in a mouse model of lupus. Our results demonstrate that homeostatic efferocytosis relies on distal metabolic signals and suggest that defective homeostatic efferocytosis may explain the link between ABX use and inflammatory disease4-7.
Collapse
Affiliation(s)
- Pedro H V Saavedra
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Biology, Northeastern University, Boston, MA, USA.
| | - Alissa J Trzeciak
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allie Lipshutz
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Anya J O'Neal
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zong-Lin Liu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaoquan Wang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Jesús E Romero-Pichardo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Waleska Saitz Rojas
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giulia Zago
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Christopher D Lucas
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
- Institute for Regeneration and Repair, Edinburgh BioQuarter, UK
| | | | - Alexander Y Rudensky
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin S A Perry
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Ding Y, Zhou Y, Zhao J, Wu C, Zhang S, Jiang N, Qian J, Zhang L, Li J, Xu D, Leng X, Wang Q, Tian X, Li M, Zeng X. The additional role of anti-nucleosome antibodies in the prediction of renal damage in systemic lupus erythematosus based on CSTAR (XXV). Lupus 2024; 33:986-997. [PMID: 38853349 DOI: 10.1177/09612033241260231] [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: 06/11/2024]
Abstract
OBJECTIVES The predominant determinant of an unfavorable prognosis among Systemic Lupus Erythematosus (SLE) patients resides in the irreversible organ damage. This prospective cohort study aimed to identify the additional value of anti-nucleosome antibodies on organ damage accumulation in SLE patients. METHODS Based on the Chinese SLE Treatment and Research group (CSTAR) registry, demographic characteristics, autoantibodies profiles, and clinical manifestations were collected at baseline. Follow-up data were collected by reviewing clinical records. RESULTS Of 2481 SLE patients with full follow-up data, 663 (26.7%) were anti-nucleosome antibodies positive and 1668 (68.0%) were anti-dsDNA antibodies positive. 764 (30.8%) patients developed new organ damage during a mean follow-up of 4.31 ± 2.60 years. At baseline, patients with positive anti-nucleosome antibodies have a higher rate of lupus nephritis (50.7% vs 36.2%, p < .001). According to the multivariable Cox regression analysis, both anti-nucleosome (HR = 1.30, 95% CI, 1.09-1.54, p < .001) and anti-dsDNA antibodies (HR=1.68, 95% CI, 1.38-2.05, p < .001) were associated with organ damage accumulation. Anti-nucleosome (HR = 2.51, 95% CI, 1.81-3.46, p < .001) and anti-dsDNA antibodies (HR = 1.69, 95% CI, 1.39-2.06, p < .001) were independent predictors for renal damage. Furthermore, the combination of the two antibodies can provide more accurate information about renal damage in overall SLE patients (HR = 3.19, 95% CI, 2.49-4.10, p < .001) and patients with lupus nephritis at baseline (HR = 2.86, 95% CI, 2.29-3.57, p < .001). CONCLUSION Besides anti-dsDNA antibodies, anti-nucleosome antibodies can also provide information about organ damage accrual during follow-up. The ability of co-positivity of anti-nucleosome and anti-dsDNA antibodies in predicting renal damage may lead to additional benefits in the follow-up of these patients.
Collapse
Affiliation(s)
- Yufang Ding
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yangzhong Zhou
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Chanyuan Wu
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Nan Jiang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Junyan Qian
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Li Zhang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jing Li
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Dong Xu
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaomei Leng
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Qian Wang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xinping Tian
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
7
|
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
|
8
|
Chai F, Peng H, Qin L, Liu C, Zeng Y, Wang R, Xu G, Wang R, Wei G, Huang H, Lan Y, Chen W, Wang C. MicroRNA miR-181d-5p regulates the MAPK signaling pathway by targeting mitogen-activated protein kinase 8 (MAPK8) to improve lupus nephritis. Gene 2024; 893:147961. [PMID: 37931853 DOI: 10.1016/j.gene.2023.147961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is a common immune disease. The microRNA (miR)-181d-5p is a potential target for treating kidney injury. However, the therapeutic role of miR-181d-5p in LN has not been investigated. This study aimed to investigate the role of miR-181d-5p in targeting mitogen-activated protein kinase 8 (MAPK8) and stimulating the MAPK signaling pathway in LN. METHODS RT-qPCR was performed to identify the variations in miR-181d-5p expression in peripheral blood mononuclear cells (PBMCs) obtained from 42 LN patients, 30 healthy individuals, 6 MRL/lpr mice and 6 C57BL/6 mice. Western blot was used to detect the effect of miR-181d-5p on the MAPK signaling pathway in THP-1 cells and MRL/lpr mice. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect the effect of miR-181d-5p on antinuclear antibodies and inflammatory factors. A dual-luciferase reporter assay was used to verify whether miR-181d-5p directly targets MAPK8. Flow cytometry was performed to evaluate apoptosis rates in transfected THP-1 cells. RESULTS miR-181d-5p expression was downregulated in PBMCs of LN patients (P < 0.01) and MRL/lpr mice (P < 0.05). A dual luciferase reporter assay demonstrated that miR-181d-5p inhibits MAPK8 (P < 0.01). Overexpression of miR-181d-5p inhibited the phosphorylation of p38 (P < 0.001) and p44/42 (P < 0.01). Moreover, miR-181d-5p decreased the apoptosis rate of THP-1 cells (P < 0.001), and reduced the secretion of IL-6 (P < 0.01) and TNF-α (P < 0.01). Furthermore, overexpression of miR-181d-5p decreased anti-dsDNA antibody (P < 0.05), anti-Sm antibody (P < 0.01), and fibrosis levels in MRL/lpr mice. CONCLUSION Upregulation of miR-181d-5p showed anti-inflammatory and anti-apoptotic effects on THP-1 cells in vitro and kidney injury in vivo. These effects were achieved by miR-181d-5p targeting MAPK8 to inhibit phosphorylation of p38 and p44/42. These results may offer new insights for improving therapeutic strategies against lupus nephritis.
Collapse
Affiliation(s)
- Fu Chai
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Huixin Peng
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Linxiu Qin
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Chunhong Liu
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yonglong Zeng
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Rong Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guidan Xu
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Rongqi Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guijiang Wei
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Huayi Huang
- Roswell Park Comprehensive Cancer Center, Surgical Oncology, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Yan Lan
- Department of Dermatology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Wencheng Chen
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.
| | - Chunfang Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.
| |
Collapse
|
9
|
Alduraibi FK, Tsokos GC. Lupus Nephritis Biomarkers: A Critical Review. Int J Mol Sci 2024; 25:805. [PMID: 38255879 PMCID: PMC10815779 DOI: 10.3390/ijms25020805] [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: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Lupus nephritis (LN), a major complication in individuals diagnosed with systemic lupus erythematosus, substantially increases morbidity and mortality. Despite marked improvements in the survival of patients with severe LN over the past 50 years, complete clinical remission after immunosuppressive therapy is achieved in only half of the patients. Therefore, timely detection of LN is vital for initiating prompt therapeutic interventions and improving patient outcomes. Biomarkers have emerged as valuable tools for LN detection and monitoring; however, the complex role of these biomarkers in LN pathogenesis remains unclear. Renal biopsy remains the gold standard for the identification of the histological phenotypes of LN and guides disease management. However, the molecular pathophysiology of specific renal lesions remains poorly understood. In this review, we provide a critical, up-to-date overview of the latest developments in the field of LN biomarkers.
Collapse
Affiliation(s)
- Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - George C. Tsokos
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
| |
Collapse
|
10
|
Chen L, Yuan M, Tan Y, Zhao M. Serum IgE anti-dsDNA autoantibodies in patients with proliferative lupus nephritis are associated with tubulointerstitial inflammation. Ren Fail 2023; 45:2273981. [PMID: 38059453 PMCID: PMC11001354 DOI: 10.1080/0886022x.2023.2273981] [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: 03/24/2023] [Accepted: 10/17/2023] [Indexed: 12/08/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the overproduction of multiple autoantibodies. Lupus nephritis (LN), the most common cause of morbidity and mortality, requires early detection. However, only a limited number of serum biomarkers have been associated with the disease activity of LN. Serum IgE anti-dsDNA autoantibodies are prevalent in patients with SLE and may be associated with the pathogenesis of LN. In this study, serum samples from 88 patients with biopsy-proven proliferative LN were collected along with complete clinical and pathological data to investigate the clinical and pathological associations of anti-dsDNA IgE autoantibodies using ELISA. This study found that the prevalence of IgE anti-dsDNA autoantibodies in patients with proliferative LN was 38.6% (34/88). Patients with anti-dsDNA IgE autoantibodies were more prone to acute kidney injury (17/34 vs. 14/54; p = .025). Levels of anti-dsDNA IgE autoantibodies were associated with interstitial inflammation (r = 0.962, p = .017). Therefore, anti-dsDNA IgE autoantibody levels are associated with tubulointerstitial inflammation in patients with proliferative LN.
Collapse
Affiliation(s)
- Leran Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Mo Yuan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| |
Collapse
|
11
|
Choi SE, Park DJ, Kang JH, Lee SS. Significance of co-positivity for anti-dsDNA, -nucleosome, and -histone antibodies in patients with lupus nephritis. Ann Med 2023; 55:1009-1017. [PMID: 36896834 PMCID: PMC10795605 DOI: 10.1080/07853890.2023.2187076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
OBJECTIVE The aim of this study was to define the clinical, histopathologic, and prognostic features associated with simultaneous positivity for anti-dsDNA, -nucleosome, and -histone antibodies (3-pos) in Korean patients with biopsy-proven lupus nephritis (LN). METHODS The 102 patients included in the study had undergone kidney biopsy prior to the start of induction treatment, were treated with immunosuppressives, and followed-up for >12 months. RESULTS In total, 44 (43.1%) of the 102 LN patients were 3-pos. Patients with 3-pos had a higher SLEDAI-2K score (p = .002), lower lymphocyte count (p = .004), and higher rates of proteinuria > 3.5 g/24 h (p = .039) and positivity for urinary sediments (p = .005) at the time of renal biopsy than non-3-pos patients. 3-pos patients had a more proliferative form of LN (p = .045) in the renal histopathologic findings, and as co-positivity gradually increased from 0 to 3, the total activity score in the renal biopsy findings increased significantly (p = .033). In addition, 3-pos patients had a more rapid eGFR decline than non-3-pos patients after a follow-up of 83.2 months (p = .016). CONCLUSIONS Our findings suggest that 3-pos is related to severe LN and that 3-pos patients are more likely to experience a rapid decline of renal function than non-3-pos patients.KEY MESSAGEPatients with co-positivity for anti-dsDNA, -nucleosome, and -histone antibodies (3-pos) had higher disease activity and a worse renal histopathology than those without co-positivity.3-pos patients had a more rapid decline of renal function than non-3-pos patients.
Collapse
Affiliation(s)
- Sung-Eun Choi
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School & Hospital, Gwangju, Republic of Korea
| | - Dong-Jin Park
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School & Hospital, Gwangju, Republic of Korea
| | - Ji-Hyoun Kang
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School & Hospital, Gwangju, Republic of Korea
| | - Shin-Seok Lee
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School & Hospital, Gwangju, Republic of Korea
| |
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
|
Gatto M, Depascale R, Stefanski AL, Schrezenmeier E, Dörner T. Translational implications of newly characterized pathogenic pathways in systemic lupus erythematosus. Best Pract Res Clin Rheumatol 2023; 37:101864. [PMID: 37625930 DOI: 10.1016/j.berh.2023.101864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023]
Abstract
Improved characterization of relevant pathogenic pathways in systemic lupus erythematosus (SLE) has been further delineated over the last decades. This led to the development of targeted treatments including belimumab and anifrolumab, which recently became available in clinics. Therapeutic targets in SLE encompass interferon (IFN) signaling, B-T costimulation including immune checkpoints, and increasing modalities of B lineage targeting, such as chimeric antigen receptor (CAR) T cells directed against CD19 or sequential anti-B cell targeting. Patient profiling based on characterization of underlying molecular abnormalities, often performed through comprehensive omics analyses, has recently been shown to better predict patients' treatment responses and also holds promise to unravel key molecular mechanisms driving SLE. SLE carries two key signatures, namely the IFN and B lineage/plasma cell signatures. Recent advances in SLE treatments clearly indicate that targeting innate and adaptive immunity is successful in such a complex autoimmune disease. Although those signatures may interact at the molecular level and provide the basis for the first selective treatments in SLE, it remains to be clarified whether these distinct treatments show different treatment responses among certain patient subsets. In fact, notwithstanding the remarkable amount of novel clues for innovative SLE treatment, harmonization of big data within tailored treatment strategies will be instrumental to better understand and treat this challenging autoimmune disorder. This review will provide an overview of recent improvements in SLE pathogenesis, related insights by analyses of big data and machine learning as well as technical improvements in conducting clinical trials with the ultimate goal that translational research results in improved patient outcomes.
Collapse
Affiliation(s)
- Mariele Gatto
- Unit of Rheumatology, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Roberto Depascale
- Unit of Rheumatology, Department of Medicine, University of Padova, Padova, Italy
| | - Ana Luisa Stefanski
- Deutsches Rheumaforschungszentrum Berlin, a Leibniz Institute, Berlin, Germany
| | - Eva Schrezenmeier
- Deutsches Rheumaforschungszentrum Berlin, a Leibniz Institute, Berlin, Germany; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Dörner
- Deutsches Rheumaforschungszentrum Berlin, a Leibniz Institute, Berlin, Germany; Department of Rheumatology and Clinical Immunology - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
14
|
Hsu TC, Yang YH, Wang LC, Lee JH, Yu HH, Lin YT, Hu YC, Chiang BL. Risk factors for subsequent lupus nephritis in patients with juvenile-onset systemic lupus erythematosus: a retrospective cohort study. Pediatr Rheumatol Online J 2023; 21:28. [PMID: 36964531 PMCID: PMC10039593 DOI: 10.1186/s12969-023-00806-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/08/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND Lupus nephritis (LN) is a crucial organ involvement in systemic lupus erythematosus (SLE). Patients with LN have higher morbidity and mortality rates than those without. Among all patients with LN, 20-40% had delayed onset, but the data for patients with juvenile-onset SLE (jSLE), who have a higher percentage of LN than patients with adult-onset SLE (aSLE), were limited. This study aimed to determine the risk factors for subsequent LN in patients with jSLE. METHODS A retrospective cohort study was conducted between 2008 and 2018 in a single tertiary medical centre. Patients with diagnosed jSLE were reviewed. We investigated those without LN at diagnosis and whether they developed LN afterward. The primary outcome was the development of subsequent LN. Clinical manifestations at diagnosis, serial laboratory data, and treatments were reviewed during follow-up periods. RESULTS Among the 48 patients with jSLE without initial LN, 20 developed subsequent LN later (Group 1), whereas 28 remained free of LN (Group 2). There was no difference in the percentage of initial manifestations except for more discoid rashes in Group 2 patients. In the Cox regression model, elevated average anti-double-stranded DNA (dsDNA) antibody, low average serum complements, and high average erythrocyte sedimentation rate (ESR) levels during follow-up were predictors of subsequent LN. After adjusting for these factors in multivariable analyses, only high average anti-dsDNA antibody and high average ESR levels remained predictive of subsequent LN. For every 100 IU/ml increase in anti-dsDNA antibody, the risk for subsequent LN in jSLE increases by 1.29 times (hazard ratio = 1.29, 95% confidence interval 1.055-1.573). CONCLUSION Persistently high anti-dsDNA antibody and ESR levels during the follow-up period were risk factors for subsequent LN in patients with jSLE.
Collapse
Affiliation(s)
- Tzu-Chuan Hsu
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
- Department of Paediatrics, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yao-Hsu Yang
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
- Department of Paediatrics, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Li-Chieh Wang
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
| | - Jyh-Hong Lee
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
| | - Hsin-Hui Yu
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
| | - Yu-Tsan Lin
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
| | - Ya-Chiao Hu
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Bor-Luen Chiang
- Department of Paediatrics, National Taiwan University Hospital, No. 8 Chung-Shan South Road, Taipei, 100, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Muacevic A, Adler JR, Al Zahrani RA, Kari JA. Lupus Nephritis in an Adolescent Girl With Hyper-Immunoglobulin E. Cureus 2023; 15:e34332. [PMID: 36865980 PMCID: PMC9972904 DOI: 10.7759/cureus.34332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2023] [Indexed: 01/30/2023] Open
Abstract
We report the case of an adolescent girl with frequent hospital admissions for severe eczematous skin rashes with recurrent epistaxis and chest infections. Investigations revealed persistent severely elevated serum total immunoglobulin E (IgE) levels but normal levels of other immunoglobulins, suggesting hyper-IgE syndrome. The first skin biopsy revealed superficial dermatophytic dermatitis (tinea corpora). Another biopsy performed after six months revealed a prominent basement membrane with dermal mucin, suggesting an underlying autoimmune disease. Her condition was complicated by proteinuria, hematuria, hypertension, and edema. A kidney biopsy revealed class IV lupus nephritis, according to the International Society of Nephrology/Renal Pathology Society (ISN/RPS). Based on the American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) criteria, she was diagnosed with systemic lupus erythematosus (SLE). She was first administered with intravenous pulse methylprednisolone (600 mg/m2) for three consecutive days, followed by oral prednisolone (40 mg/m2) daily, mycophenolate mofetil tablets (600 mg/m2/dose) twice daily, hydroxychloroquine (200 mg) once daily, and three classes of antihypertensive medications. She maintained normal renal functions with no lupus morbidity for 24 months, then rapidly progressed to end-stage kidney disease, and was then started on three to four sessions of regular hemodialysis per week. Hyper-IgE is known to be a marker of immune dysregulation as it facilitates the generation of immune complexes (ICs) that mediate lupus nephritis and juvenile SLE. Regardless of the different factors that are impacting the production of IgE, the present case illustrated that juvenile patients with SLE may have increased IgE levels, indicating that higher IgE levels might have a role in lupus pathogenesis and prognosis. The mechanisms regarding the increased levels of IgE in subjects with lupus need further investigation. Further studies are thus required to assess the incidence, prognosis, and possible new specific management for hyper-IgE in juvenile SLE.
Collapse
|
16
|
Karasawa T, Sato R, Imaizumi T, Hashimoto S, Fujita M, Aizawa T, Tsugawa K, Kawaguchi S, Seya K, Terui K, Tanaka H. Glomerular endothelial expression of type I IFN-stimulated gene, DExD/H-Box helicase 60 via toll-like receptor 3 signaling: possible involvement in the pathogenesis of lupus nephritis. Ren Fail 2022; 44:137-145. [PMID: 35392757 PMCID: PMC9004514 DOI: 10.1080/0886022x.2022.2027249] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/03/2022] [Accepted: 01/03/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Sustained type I interferon (IFN) activation via Toll-like receptor (TLR) 3, 7 and 9 signaling has been reported to play a pivotal role in the development of lupus nephritis (LN). Although type I IFN activation has been shown to induce interferon-stimulated genes (ISGs) expression in systemic lupus erythematosus, the implication of ISGs expression in intrinsic glomerular cells remains largely unknown. METHODS We treated cultured human glomerular endothelial cells (GECs) with polyinosinic-polycytidylic acid (poly IC), R848, and CpG (TLR3, TLR7, and TLR9 agonists, respectively) and analyzed the expression of DExD/H-Box Helicase 60 (DDX60), a representative ISG, using quantitative reverse transcription-polymerase chain reaction and western blotting. Additionally, RNA interference against IFN-β or DDX60 was performed. Furthermore, cleavage of caspase 9 and poly (ADP-ribose) polymerase (PARP), markers of cells undergoing apoptosis, was examined using western blotting. We conducted an immunofluorescence study to examine endothelial DDX60 expression in biopsy specimens from patients with LN. RESULTS We observed that endothelial expression of DDX60 was induced by poly IC but not by R848 or CpG, and RNA interference against IFN-β inhibited poly IC-induced DDX60 expression. DDX60 knockdown induced cleavage of caspase 9 and PARP. Intense endothelial DDX60 expression was observed in biopsy specimens from patients with diffuse proliferative LN. CONCLUSION Glomerular endothelial DDX60 expression may prevent apoptosis, which is involved in the pathogenesis of LN. Modulating the upregulation of the regional innate immune system via TLR3 signaling may be a promising treatment target for LN.
Collapse
Affiliation(s)
- Takao Karasawa
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Riko Sato
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shun Hashimoto
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Masashi Fujita
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Tomomi Aizawa
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Koji Tsugawa
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Shogo Kawaguchi
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kazuhiko Seya
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kiminori Terui
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Hiroshi Tanaka
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
- Department of School Health Science, Hirosaki University Faculty of Education, Hirosaki, Japan
| |
Collapse
|
17
|
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
|
18
|
Yu C, Li P, Dang X, Zhang X, Mao Y, Chen X. Lupus nephritis: new progress in diagnosis and treatment. J Autoimmun 2022; 132:102871. [PMID: 35999111 DOI: 10.1016/j.jaut.2022.102871] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multifactorial autoimmune disease that affects many organs, including the kidney. Lupus nephritis (LN) is a common manifestation characterized by heterogeneous clinical and histopathological findings, and often associates with poor prognosis. The diagnosis and treatment of LN is challenging, depending largely on renal biopsy, and there is no reliable non-invasive LN biomarker. Up to now, the complete remission rate of LN is only 20%∼30% after receiving six months of standard treatment, which is far from satisfactory. Moreover, adverse reactions to immunosuppressants, especially glucocorticoids, further compromise the prognosis of LN. Biological reagents targetting autoimmune responses and inflammatory pathways, bring hope to the treatment of intractable lupus. The European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) and KDIGO (Kidney Disease: Improving Global Outcomes) have been working on and launched the recommendations for the management of LN. In this review, we update our knowledge in the pathogenesis, diagnosis, and management of LN and prospect for the future potential targets in the management of LN.
Collapse
Affiliation(s)
- Chen Yu
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xin Dang
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yonghui Mao
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
19
|
Lou H, Ling GS, Cao X. Autoantibodies in systemic lupus erythematosus: From immunopathology to therapeutic target. J Autoimmun 2022; 132:102861. [PMID: 35872103 DOI: 10.1016/j.jaut.2022.102861] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 11/26/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ inflammatory damage and wide spectrum of autoantibodies. The autoantibodies, especially anti-dsDNA and anti-Sm autoantibodies are highly specific to SLE, and participate in the immune complex formation and inflammatory damage on multiple end-organs such as kidney, skin, and central nervous system (CNS). However, the underlying mechanisms of autoantibody-induced tissue damage and systemic inflammation are still not fully understood. Single cell analysis of autoreactive B cells and monoclonal antibody screening from patients with active SLE has improved our understanding on the origin of autoreactive B cells and the antigen targets of the pathogenic autoantibodies. B cell depletion therapies have been widely studied in the clinics, but the development of more specific therapies against the pathogenic B cell subset and autoantibodies with improved efficacy and safety still remain a big challenge. A more comprehensive autoantibody profiling combined with functional characterization of autoantibodies in diseases development will shed new insights on the etiology and pathogenesis of SLE and guide a specific treatment to individual SLE patients.
Collapse
Affiliation(s)
- Hantao Lou
- Ludwig Institute of Cancer Research, University of Oxford, Oxford, OX3 7DR, UK; Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| | - Guang Sheng Ling
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xuetao Cao
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK; Nankai-Oxford International Advanced Institute, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
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
|
Reed JH. Transforming mutations in the development of pathogenic B cell clones and autoantibodies. Immunol Rev 2022; 307:101-115. [PMID: 35001403 DOI: 10.1111/imr.13064] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 12/16/2022]
Abstract
Autoimmune diseases are characterized by serum autoantibodies, some of which are pathogenic, causing severe manifestations and organ injury. However, autoantibodies of the same antigenic reactivity are also present in the serum of asymptomatic people years before they develop any clinical signs of autoimmunity. Autoantibodies can arise during multiple stages of B cell development, and various genetic and environmental factors drive their production. However, what drives the development of pathogenic autoantibodies is poorly understood. Advances in single-cell technology have enabled the deep analysis of rare B cell clones producing pathogenic autoantibodies responsible for vasculitis in patients with primary Sjögren's syndrome complicated by mixed cryoglobulinaemia. These findings demonstrated a cascade of genetic events involving stereotypic immunoglobulin V(D)J recombination and transforming somatic mutations in lymphoma genes and V(D)J regions that disrupted antibody quality control mechanisms and decreased autoantibody solubility. Most studies consider V(D)J mutations that enhance autoantibody affinity to drive pathology; however, V(D)J mutations that increase autoantibody propensity to form insoluble complexes could be a major contributor to autoantibody pathogenicity. Defining the molecular characteristics of pathogenic autoantibodies and failed tolerance checkpoints driving their formation will improve prognostication, enabling early treatment to prevent escalating organ damage and B cell malignancy.
Collapse
Affiliation(s)
- Joanne H Reed
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Santos JE, Vicente R, Malvar B, Santos I, Coimbra M, Amoedo M, Pires C. Lupus-like nephritis with positive anti-neutrophil cytoplasmic antibodies and negative antinuclear antibodies. J Bras Nefrol 2022; 44:121-125. [PMID: 33107901 PMCID: PMC8943873 DOI: 10.1590/2175-8239-jbn-2020-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022] Open
Abstract
Antineutrophil cytoplasmic antibodies (ANCAs) are associated with small vessel vasculitis but their prevalence is not rare in other immune diseases. In lupus nephritis (LN), their pathological role and clinical relevance have been the target of controversial views. We present a case of acute kidney injury and nephrotic syndrome in a young woman with diffuse global proliferative and membranous nephritis on her kidney biopsy, showing a full-house immunofluorescence pattern, very allusive of class IV + V LN, but lacking associated clinical criteria and laboratory findings to support the diagnosis of systemic lupus erythematosus (SLE). Furthermore, the patient presented with high titers of ANCA, steadily decreasing alongside the renal function and proteinuria improvements, with mycophenolate mofetil (MMF) and steroid treatment. The authors believe this is a case of lupus-like nephritis, in which ANCAs are immunological markers, although they are not directly involved in the pathogenesis.
Collapse
Affiliation(s)
| | - Rita Vicente
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| | - Beatriz Malvar
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| | - Iolanda Santos
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| | - Miguel Coimbra
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| | - Manuel Amoedo
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| | - Carlos Pires
- Hospital Espírito Santo de Évora, Department of Nephrology, Évora, Portugal
| |
Collapse
|
23
|
Dahlqvist J, Fulco CP, Ray JP, Liechti T, de Boer CG, Lieb DJ, Eisenhaure TM, Engreitz JM, Roederer M, Hacohen N. Systematic identification of genomic elements that regulate FCGR2A expression and harbor variants linked with autoimmune disease. Hum Mol Genet 2021; 31:1946-1961. [PMID: 34970970 PMCID: PMC9239749 DOI: 10.1093/hmg/ddab372] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND FCGR2A binds antibody-antigen complexes to regulate the abundance of circulating and deposited complexes along with downstream immune and autoimmune responses. Although the abundance of FCRG2A may be critical in immune-mediated diseases, little is known about whether its surface expression is regulated through cis genomic elements and non-coding variants. In the current study, we aimed to characterize the regulation of FCGR2A expression, the impact of genetic variation and its association with autoimmune disease. METHODS We applied CRISPR-based interference and editing to scrutinize 1.7 Mb of open chromatin surrounding the FCGR2A gene to identify regulatory elements. Relevant transcription factors (TFs) binding to these regions were defined through public databases. Genetic variants affecting regulation were identified using luciferase reporter assays and were verified in a cohort of 1996 genotyped healthy individuals using flow cytometry. RESULTS We identified a complex proximal region and five distal enhancers regulating FCGR2A. The proximal region split into subregions upstream and downstream of the transcription start site, was enriched in binding of inflammation-regulated TFs, and harbored a variant associated with FCGR2A expression in primary myeloid cells. One distal enhancer region was occupied by CCCTC-binding factor (CTCF) whose binding site was disrupted by a rare genetic variant, altering gene expression. CONCLUSIONS The FCGR2A gene is regulated by multiple proximal and distal genomic regions, with links to autoimmune disease. These findings may open up novel therapeutic avenues where fine-tuning of FCGR2A levels may constitute a part of treatment strategies for immune-mediated diseases.
Collapse
Affiliation(s)
- Johanna Dahlqvist
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA,Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Charles P Fulco
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA,Bristol Myers Squibb, Cambridge, MA 02142, USA
| | - John P Ray
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA,Systems Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Thomas Liechti
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20814, USA
| | - Carl G de Boer
- Klarman Cell Observatory, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA,School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - David J Lieb
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Thomas M Eisenhaure
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Jesse M Engreitz
- Center for Cell Circuits, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA,BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital, Stanford University School of Medicine, Stanford, CA, USA
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20814, USA
| | - Nir Hacohen
- To whom correspondence should be addressed at: The Broad Institute of MIT and Harvard University, 415 Main Street, Cambridge, MA 02142, USA. Tel: +1 6177147234, Fax: +1 6177148956;
| |
Collapse
|
24
|
Ponticelli C, Moroni G, Fornoni A. Lupus Membranous Nephropathy. GLOMERULAR DISEASES 2021; 1:10-20. [PMID: 36751488 PMCID: PMC9677716 DOI: 10.1159/000512278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/09/2020] [Indexed: 11/19/2022]
Abstract
Background Lupus membranous nephropathy (LMN) is a rare disease, usually associated with nephrotic syndrome. Methods We reviewed the literature by searching for the following terms on Pubmed.gov: lupus nephritis, membranous nephropathy (MN), lupus membranous nephropathy, nephrotic syndrome, and Class V lupus nephritis. Results The histology of LMN at light microscopy is similar to that of primary MN. Cases of MN associated with focal or diffuse proliferation are not considered LMN by the International Society of Nephrology/Renal Pathology Society classification. Immunofluorescence study of LMN shows deposits of all immunoglobulins and complement. Tubulo-reticular structures, extraglomerular deposits, subepithelial, and scanty subendothelial deposits can be seen on electron microscopy. Phospholipase A2 receptor deposits are usually but not necessarily absent in LMN. The pathogenesis is still not completely understood. The inflammatory milieu of lupus may favor the development of autoantigens and intraglomerular assembly of immune complexes. These are more often associated with mesangial or endocapillary hypercellular lesions. Alternatively, autoantibodies may bind autoantigens in the glomerular subepithelium, triggering a signaling cascade leading to LMN. A central role in the development of podocyte injury and proteinuria is played by the components of complement C5b-C9. CKD progression in LMN is slow but may be accelerated by the frequency of renal flares. Persistent nephrotic syndrome and/or the frequent use of corticosteroids may lead to a series of life-threatening complications. Discussion Treatment of arterial hypertension, dyslipidemia, and diabetes are of paramount importance. Besides specific therapies of these complications, hydroxychloroquine and vitamin D supplementation are recommended. Immunosuppression should be limited to patients with nephrotic proteinuria. The most frequently used drugs are corticosteroids, calcineurin inhibitors, cyclophosphamide, mycophenolate, and rituximab, alone or combined. Early detection and treatment of renal flares is of paramount importance to prevent CKD progression.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Nephrology, IRCCS Ospedale Maggiore Policlinico (retired), Milan, Italy,*Claudio Ponticelli, Via Ampere 126, IT–20131 Milan (Italy),
| | - Gabriella Moroni
- Nephrology Unit Fondazione IRCCS Ca' Granda Ospedale Maggiore Milano, Milan, Italy
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension and Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miami, Florida, USA
| |
Collapse
|
25
|
Singh D, Oudit O, Hajtovic S, Sarbaugh D, Salis R, Adebowale T, James J, Spatz LA. Antibodies to an Epstein Barr Virus protein that cross-react with dsDNA have pathogenic potential. Mol Immunol 2021; 132:41-52. [PMID: 33545624 DOI: 10.1016/j.molimm.2021.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/07/2021] [Accepted: 01/17/2021] [Indexed: 01/02/2023]
Abstract
Pathogens such as the Epstein Barr virus (EBV) have long been implicated in the etiology of systemic lupus erythematosus (SLE). The Epstein Barr virus nuclear antigen I (EBNA-1) has been shown to play a role in the development of anti-nuclear antibodies characteristic of SLE. One mechanism by which EBV may play a role in SLE is molecular mimicry. We previously generated two monoclonal antibodies (mAbs) to EBNA-1 and demonstrated that they cross-react with double-stranded DNA (dsDNA). In the present study, we demonstrate that these mAbs have pathogenic potential. We show that they can bind to isolated rat glomeruli and that binding can be greatly diminished by pretreatment of glomeruli with DNase I, suggesting that these mAbs bind dsDNA in the kidney. We also demonstrate that these antibodies can deposit in the kidney when injected into mice and can induce proteinuria and elicit histopathological alterations consistent with glomerulonephritis. Finally, we show that these antibodies can cross-react with laminin and collagen IV in the extracellular matrix suggesting that direct binding to the glomerular basement membrane or mesangial matrix may also contribute to the antibody deposition in the kidney. In summary, our results indicate that EBNA-1 can elicit antibodies that cross-react with dsDNA, that can deposit in the kidney, and induce kidney damage. These results are significant because they support the role of a viral protein in SLE and lupus nephritis.
Collapse
Affiliation(s)
- Divya Singh
- The Molecular, Cellular, and Biomedical Sciences Department, The CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Omar Oudit
- The Department of Chemistry & Biochemistry, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Sabastian Hajtovic
- The CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Dylan Sarbaugh
- The Department of Biology, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Rafatu Salis
- The Department of Biology, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Temitayo Adebowale
- The Department of Biology, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Justin James
- The CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA
| | - Linda A Spatz
- The Molecular, Cellular, and Biomedical Sciences Department, The CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY, 10031, USA.
| |
Collapse
|
26
|
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
|
27
|
Lou H, Wojciak-Stothard B, Ruseva MM, Cook HT, Kelleher P, Pickering MC, Mongkolsapaya J, Screaton GR, Xu XN. Autoantibody-dependent amplification of inflammation in SLE. Cell Death Dis 2020; 11:729. [PMID: 32908129 PMCID: PMC7481301 DOI: 10.1038/s41419-020-02928-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Anti-double stranded DNA antibodies (anti-dsDNA) are a hallmark of SLE but their role in disease pathogenesis is not fully resolved. Anti-dsDNA in serum are highly heterogeneous therefore in this study, we aimed to dissect the functional specificities of anti-dsDNA using a panel of human monoclonal antibodies (humAbs) generated from patients with active lupus nephritis. A total of 46 ANA reactive humAbs were isolated and divided into four broad classes based on their reactivity to histones, DNA and Crithidia. Functional analysis indicated that one subclass of antibodies bound strongly to decondensed DNA areas in neutrophil extracellular traps (NETs) and protected NETs from nuclease digestion, similar to the sera from active SLE patients. In addition, these anti-dsDNA antibodies could stimulate type I interferon responses in mononuclear phagocytic cells, or NF-kB activity in endothelial cells, by uptake of NETs-anti-NETs immune complexes and subsequently trigging inflammatory responses in an Fc-gamma receptor (Fcg-R)-dependant manner. Together our data suggest that only a subset of anti-dsDNA antibodies is capable to amplify inflammatory responses by deposit in the nephritic kidney in vivo, protecting NETs digestion as well as uptake of NETs immune complexes into Fcg-R-expressing cells in vitro.
Collapse
Affiliation(s)
- Hantao Lou
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK.
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK.
| | - Beata Wojciak-Stothard
- Department of Medicine, Centre for Pharmacology and Therapeutics, Imperial College London, London, W12 0HS, UK
| | - Marieta M Ruseva
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - H Terence Cook
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - Peter Kelleher
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK
- Department of Infection and Immunity, Charing Cross Hospital North West London Pathology, London, W6 8RF, UK
| | - Matthew C Pickering
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - Juthathip Mongkolsapaya
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
- Division of Medical Sciences, John Radcliffe Hospital, Oxford University, Oxford, OX3 9DU, UK
| | - Gavin R Screaton
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
- Division of Medical Sciences, John Radcliffe Hospital, Oxford University, Oxford, OX3 9DU, UK
| | - Xiao-Ning Xu
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK.
| |
Collapse
|
28
|
Dorraji SE, Kanapathippillai P, Hovd AMK, Stenersrød MR, Horvei KD, Ursvik A, Figenschau SL, Thiyagarajan D, Fenton CG, Pedersen HL, Fenton KA. Kidney Tertiary Lymphoid Structures in Lupus Nephritis Develop into Large Interconnected Networks and Resemble Lymph Nodes in Gene Signature. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2203-2225. [PMID: 32818496 DOI: 10.1016/j.ajpath.2020.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
Immune aggregates organized as tertiary lymphoid structures (TLS) are observed within the kidneys of patients with systemic lupus erythematosus and lupus nephritis (LN). Renal TLS was characterized in lupus-prone New Zealand black × New Zealand white F1 mice analyzing cell composition and vessel formation. RNA sequencing was performed on transcriptomes isolated from lymph nodes, macrodissected TLS from kidneys, and total kidneys of mice at different disease stages by using a personal genome machine and RNA sequencing. Formation of TLS was found in anti-double-stranded DNA antibody-positive mice, and the structures were organized as interconnected large networks with distinct T/B cell zones with adjacent dendritic cells, macrophages, plasma cells, high endothelial venules, supporting follicular dendritic cells network, and functional germinal centers. Comparison of gene profiles of whole kidney, renal TLS, and lymph nodes revealed a similar gene signature of TLS and lymph nodes. The up-regulated genes within the kidneys of lupus-prone mice during LN development reflected TLS formation, whereas the down-regulated genes were involved in metabolic processes of the kidney cells. A comparison with human LN gene expression revealed similar up-regulated genes as observed during the development of murine LN and TLS. In conclusion, kidney TLS have a similar cell composition, structure, and gene signature as lymph nodes and therefore may function as a kidney-specific type of lymph node.
Collapse
Affiliation(s)
- Seyed Esmaeil Dorraji
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Premasany Kanapathippillai
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Aud-Malin Karlsson Hovd
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Mikael Ryan Stenersrød
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Kjersti Daae Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Anita Ursvik
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Stine Linn Figenschau
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Christopher Graham Fenton
- Genomic Support Center, Department of Clinical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromso, Norway
| | - Hege Lynum Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Kristin Andreassen Fenton
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
29
|
Abstract
Antibody-mediated rejection continues to hinder long-term survival of solid organ allografts. Natural antibodies (Nabs) with polyreactive and autoreactive properties have recently emerged as potential contributors to antibody-mediated graft rejection. This review discusses Nabs, their functions in health and disease, their significance in rejection following kidney, heart, and lung transplantation, and their implication in serum reactivity to key antigens associated with rejection. Finally, potential effector mechanisms of Nabs in the context of transplantation are explored.
Collapse
|
30
|
Gatto M, Radu CM, Luisetto R, Ghirardello A, Bonsembiante F, Trez D, Valentino S, Bottazzi B, Simioni P, Cavicchioli L, Doria A. Immunization with Pentraxin3 prevents transition from subclinical to clinical lupus nephritis in lupus-prone mice: Insights from renal ultrastructural findings. J Autoimmun 2020; 111:102443. [PMID: 32265078 DOI: 10.1016/j.jaut.2020.102443] [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: 12/11/2019] [Revised: 03/15/2020] [Accepted: 03/22/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Pentraxin3 (PTX3) is an emerging player in lupus nephritis (LN). Anti-PTX3 antibodies showed to delay LN occurrence in vivo. AIM To evaluate renal changes following immunization with PTX3 in a murine model of LN. MATERIALS AND METHODS Twenty-two lupus-prone New Zealand Black/White (NZB/W)F1 mice were divided into two groups (n = 11) and subcutaneously injected with human recombinant (hr)PTX3 100 μg or phosphate buffer saline (PBS) 200 μl, three times 3 weeks apart, starting before development of proteinuria. Five mice from each group were scheduled for sacrifice at week 22 and 6 from each group at week 29. Renal lesions included electron-dense deposits (EDD), glomerular deposition of IgG, complement and PTX3 as markers of renal inflammation. They were evaluated by immunofluorescence (IF), confocal and immunoelectron microscopy (IEM). Validated semiquantitative scores were used when available to score renal lesions. Chi-squared test with Fisher exact test was used for comparison. RESULTS Nineteen out of 22 mice were sacrificed as scheduled. Only hrPTX3-immunized mice developed anti-PTX3 antibodies. Compared to PBS-injected mice, they displayed a dramatic decrease in glomerular deposits of IgG, C1q and PTX3, as well as in the amount of EDD (p = 0.006) and podocyte effacement (p = 0.043). Importantly, PTX3 was pinpointed inside the EDD and co-localized with nuclear material. CONCLUSIONS Immunization with PTX3 prevented progression from the preclinical to the clinical stage of LN, inciting anti-PTX3 antibodies and preventing renal PTX3 deposition. PTX3 is a novel component of EDD, submitting it as one initiating autoantigen in LN and as potential target for early treatment.
Collapse
Affiliation(s)
- Mariele Gatto
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Claudia M Radu
- Department of Women's and Children's Health, University of Padova, Padova, Italy; Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anna Ghirardello
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Federico Bonsembiante
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Padova, Italy; Department of Animal Medicine, Production and Health University of Padova, Italy
| | - Davide Trez
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Padova, Italy
| | | | | | - Paolo Simioni
- Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Laura Cavicchioli
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Padova, Italy
| | - Andrea Doria
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, Padova, Italy.
| |
Collapse
|
31
|
Chen A, Lee K, Guan T, He JC, Schlondorff D. Role of CD8+ T cells in crescentic glomerulonephritis. Nephrol Dial Transplant 2020; 35:564-572. [PMID: 30879039 PMCID: PMC7139212 DOI: 10.1093/ndt/gfz043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/09/2019] [Indexed: 01/20/2023] Open
Abstract
Crescentic glomerulonephritis (cGN) comprises three main types according to the pathogenesis and immunofluorescence patterns: anti-glomerular basement membrane antibody cGN, vasculitis-associated cGN and post-infectious immune complex cGN. In this brief review of the immune-pathogenesis of cGN, the focus is mainly on the role of CD8+ T cells in the progression of cGN. Under control conditions, Bowman's capsule (BC) provides a protected immunological niche by preventing access of cytotoxic CD8+ T cells to Bowman's space and thereby podocytes. Even in experimental nephrotoxic nephritis, leukocytes accumulate around the glomeruli, but remain outside of BC, as long as the latter remains intact. However, when and where breaches in BC occur, the inflammatory cells can gain access to and destroy podocytes, thus converting cGN into rapidly progressive glomerulonephritis (RPGN). These conclusions also apply to human cGN, where biopsies show that loss of BC integrity is associated with RPGN and progression to end-stage kidney disease. We propose a two-hit hypothesis for the role of cytotoxic CD8+ T cells in the progression of cGN. The initial insult occurs in response to the immune complex formation or deposition, resulting in local capillary and podocyte injury (first hit). The injured podocytes release neo-epitopes, eventually causing T-cell activation and migration to the glomerulus. Upon generation of breaches in BC, macrophages and CD8+ T cells can now gain access to the glomerular space and destroy neo-epitope expressing podocytes (second hit), resulting in RPGN. While further investigation will be required to test this hypothesis, future therapeutic trials should consider targeting of CD8+ T cells in the therapy of progressive cGN.
Collapse
Affiliation(s)
- Anqun Chen
- Division of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian province, China
| | - Kyung Lee
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tianjun Guan
- Division of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian province, China
| | - John Cijiang He
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Renal Section, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Detlef Schlondorff
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Yang F, He Y, Zhai Z, Sun E. Programmed Cell Death Pathways in the Pathogenesis of Systemic Lupus Erythematosus. J Immunol Res 2019; 2019:3638562. [PMID: 31871956 PMCID: PMC6913273 DOI: 10.1155/2019/3638562] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/04/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by excessive inflammatory and immune responses and tissue damage. Increasing evidence has demonstrated the important role of programmed cell death in SLE pathogenesis. When apoptosis encounters with defective clearance, accumulated apoptotic cells lead to secondary necrosis. Different forms of lytic cell death, including secondary necrosis after apoptosis, NETosis, necroptosis, and pyroptosis, contribute to the release of damage-associated molecular patterns (DAMPs) and autoantigens, resulting in triggering immunity and tissue damage in SLE. However, the role of autophagy in SLE pathogenesis is in dispute. This review briefly discusses different forms of programmed cell death pathways and lay particular emphasis on inflammatory cell death pathways such as NETosis, pyroptosis, and necroptosis and their roles in the inflammatory and immune responses in SLE.
Collapse
Affiliation(s)
- Fangyuan Yang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
- Department of Rheumatology and Immunology, Shunde Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Sakhi H, Moktefi A, Bouachi K, Audard V, Hénique C, Remy P, Ollero M, El Karoui K. Podocyte Injury in Lupus Nephritis. J Clin Med 2019; 8:jcm8091340. [PMID: 31470591 PMCID: PMC6780135 DOI: 10.3390/jcm8091340] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by a broad spectrum of renal lesions. In lupus glomerulonephritis, histological classifications are based on immune-complex (IC) deposits and hypercellularity lesions (mesangial and/or endocapillary) in the glomeruli. However, there is compelling evidence to suggest that glomerular epithelial cells, and podocytes in particular, are also involved in glomerular injury in patients with SLE. Podocytes now appear to be not only subject to collateral damage due to glomerular capillary lesions secondary to IC and inflammatory processes, but they are also a potential direct target in lupus nephritis. Improvements in our understanding of podocyte injury could improve the classification of lupus glomerulonephritis. Indeed, podocyte injury may be prominent in two major presentations: lupus podocytopathy and glomerular crescent formation, in which glomerular parietal epithelial cells play also a key role. We review here the contribution of podocyte impairment to different presentations of lupus nephritis, focusing on the podocyte signaling pathways involved in these lesions.
Collapse
Affiliation(s)
- Hamza Sakhi
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Anissa Moktefi
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Pathology, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Khedidja Bouachi
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Vincent Audard
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Carole Hénique
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Philippe Remy
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Mario Ollero
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Khalil El Karoui
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France.
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France.
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France.
| |
Collapse
|
34
|
Mazón-Cabrera R, Vandormael P, Somers V. Antigenic Targets of Patient and Maternal Autoantibodies in Autism Spectrum Disorder. Front Immunol 2019; 10:1474. [PMID: 31379804 PMCID: PMC6659315 DOI: 10.3389/fimmu.2019.01474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose behavioral symptoms become apparent in early childhood. The underlying pathophysiological mechanisms are only partially understood and the clinical manifestations are heterogeneous in nature, which poses a major challenge for diagnosis, prognosis and intervention. In the last years, an important role of a dysregulated immune system in ASD has emerged, but the mechanisms connecting this to a disruption of brain development are still largely unknown. Although ASD is not considered as a typical autoimmune disease, self-reactive antibodies or autoantibodies against a wide variety of targets have been found in a subset of ASD patients. In addition, autoantibodies reactive to fetal brain proteins have also been described in the prenatal stage of neurodevelopment, where they can be transferred from the mother to the fetus by transplacental transport. In this review, we give an extensive overview of the antibodies described in ASD according to their target antigens, their different origins, and timing of exposure during neurodevelopment.
Collapse
Affiliation(s)
| | | | - Veerle Somers
- Biomedical Research Institute, Faculty of Medicine and Life Science, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
35
|
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
|
36
|
Bonegio RG, Lin JD, Beaudette-Zlatanova B, York MR, Menn-Josephy H, Yasuda K. Lupus-Associated Immune Complexes Activate Human Neutrophils in an FcγRIIA-Dependent but TLR-Independent Response. THE JOURNAL OF IMMUNOLOGY 2019; 202:675-683. [PMID: 30610165 DOI: 10.4049/jimmunol.1800300] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the presence of autoantibodies against nucleic acids and nucleoproteins. Anti-dsDNA Abs are considered a hallmark of SLE, and previous studies have indicated that nucleic acid-containing immune complexes (ICs) induce B cell and dendritic cell activation in a TLR-dependent process. How ICs containing nucleic acids affect neutrophil function has not been well investigated. In this study, we report that nucleic acid-containing ICs derived from the sera of SLE patients induce human and mouse neutrophil activation through TLR-independent mechanisms. Soluble ICs containing Sm/RNP, an RNA Ag, activate human neutrophils to produce reactive oxygen species (ROS) and IL-8. In contrast, ICs containing DNA have to be immobilized to efficiently activate neutrophils. We found that deleting TLR7 or TLR9, the receptors for RNA and DNA, had no effect on mouse neutrophil activation induced by RNA-containing and immobilized DNA-containing ICs. Binding of ICs are mediated through FcγRIIA and FcγRIIIB. However, neutrophil activation induced by RNA- and DNA-containing ICs requires FcγRIIA, as blocking FcγRIIA inhibited ROS release from neutrophils. RNA-containing ICs induce calcium flux, whereas TLR7/8 ligand R848 do not. Surprisingly, chloroquine inhibits calcium flux induced by RNA-containing ICs, suggesting that this lesser known function of chloroquine is involved in the neutrophil activation induced by ICs. These data indicate the SLE-derived ICs activate neutrophils to release ROS and chemokines in an FcγRIIA-dependent and TLR7- and TLR9-independent manner that likely contributes to local tissue inflammation and damage.
Collapse
Affiliation(s)
- Ramon G Bonegio
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118.,Renal Section, VA Boston Healthcare System, Boston, MA 02130; and
| | - Jessica D Lin
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | | | - Michael R York
- Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | - Hanni Menn-Josephy
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | - Kei Yasuda
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118;
| |
Collapse
|
37
|
Gatto M, Luisetto R, Ghirardello A, Cavicchioli L, Codolo G, Biasiolo A, Maggioni G, Saccon F, Beggio M, Cappon A, Venturini R, Pontisso P, Doria A. SERPINB3 Delays Glomerulonephritis and Attenuates the Lupus-Like Disease in Lupus Murine Models by Inducing a More Tolerogenic Immune Phenotype. Front Immunol 2018; 9:2081. [PMID: 30254646 PMCID: PMC6141748 DOI: 10.3389/fimmu.2018.02081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022] Open
Abstract
Objective: To explore the effects of SERPINB3 administration in murine lupus models with a focus on lupus-like nephritis. Methods: 40 NZB/W F1 mice were subdivided into 4 groups and intraperitoneally injected with recombinant SERPINB3 (7.5 μg/0.1 mL or 15 μg/0.1 mL) or PBS (0.1 mL) before (group 1 and 2) or after (group 3 and 4) the development of proteinuria (≥100 mg/dl). Two additional mice groups were provided by including 20 MRL/lpr mice which were prophylactically injected with SERPINB3 (10 mice, group 5) or PBS (10 mice, group 6). Time of occurrence and levels of anti-dsDNA and anti-C1q antibodies, proteinuria and serum creatinine, overall- and proteinuria-free survival were assessed in mice followed up to natural death. Histological analysis was performed in kidneys of both lupus models. The Th17:Treg cell ratio was assessed by flow-cytometry in splenocytes of treated and untreated MRL/lpr mice. Statistical analysis was performed using non parametric tests and Kaplan-Meier curves, when indicated. Results: Autoantibody levels and proteinuria were significantly decreased and time of occurrence significantly delayed in SERPINB3-treated mice vs. controls. In agreement with these findings, proteinuria-free and overall survival were significantly improved in SERPINB3-treated groups vs. controls. Histological analysis demonstrated a lower prevalence of severe tubular lesions in kidneys of group 5 vs. group 6. SERPINB3-treated mice showed an overall trend toward a reduced prevalence of severe lesions in both strains. Th17:Treg ratio was significantly decreased in splenocytes of MRL/lpr mice treated with SERPINB3, compared to untreated control mice. Conclusions: SERPINB3 significantly improves disease course and delays the onset of severe glomerulonephritis in lupus-prone mice, possibly inducing a more tolerogenic immune phenotype.
Collapse
Affiliation(s)
- Mariele Gatto
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anna Ghirardello
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Laura Cavicchioli
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Gaia Codolo
- Department of Biology, University of Padova, Padova, Italy
| | - Alessandra Biasiolo
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Maggioni
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesca Saccon
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marianna Beggio
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Cappon
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberta Venturini
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
| | - Patrizia Pontisso
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
38
|
Wang J, Cao P, Qi YY, Chen XP, Ma L, Deng RR, Zhang LL, Zhao Y. The relationship between cell apoptosis dysfunction and FEN1 E160D mutation in lupus nephritis patients. Autoimmunity 2018; 50:476-480. [PMID: 29239254 DOI: 10.1080/08916934.2017.1402302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study aims to evaluate the role of FEN1 E160D mutation in lupus nephritis (LN) patients with cell apoptosis dysfunction. METHODS (1) Cell apoptosis was detected from 50 paraffin samples obtained from renal biopsies of patients with Class IV LN by TUNEL method and the relationship of the systemic lupus erythematosus disease activity index 2000 (SLEDAI 2000) and renal tissue cell apoptotic index (AI) was discussed. (2) FEN1 gene 61563142-61563342 containing E160D were analysed by extracting genomic DNA from peripheral blood collected from the above 50 LN patients and 25 patients with nephrectomy caused by renal trauma. The difference between these two groups was statistically significant. RESULTS Cell apoptosis was detected in all patients with LN, and correlation analysis results revealed a positive relationship between SLEDAI 2000 and AI (r = 0.39, p = .032). The FEN1 gene 61563142-61563342 fragment had site mutations at C/- (+61563189), A/T (+61563198), A/- (+61563204), G/T (+61563303), and T/C (+61563304). However, no statistical significance was found between LN patients detected with cell apoptosis and healthy individuals. CONCLUSIONS This study revealed that cell apoptosis dysfunction plays a key role in the pathogenesis of LN, even though the difference in FEN1 gene 61563142-61563342 between LN patients and healthy individuals was not statistically significant. Larger sample size studies or genome-wide association studies are needed.
Collapse
Affiliation(s)
- Jing Wang
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Ping Cao
- b Thinks People's Hospital , Gan Su , PR China
| | - Yuan-Yuan Qi
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Xue-Ping Chen
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Li Ma
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Rong-Rong Deng
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Li-Li Zhang
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| | - Yu Zhao
- a Department of Nephropathy , Lanzhou University Second Hospital , Lanzhou , PR China
| |
Collapse
|
39
|
Charles N, Chemouny JM, Daugas E. Basophil involvement in lupus nephritis: a basis for innovation in daily care. Nephrol Dial Transplant 2018; 34:750-756. [DOI: 10.1093/ndt/gfy245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Nicolas Charles
- Centre de Recherche sur l’Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d’Excellence Inflamex, DHU FIRE, Paris, France
| | - Jonathan M Chemouny
- Service de Néphrologie, Hôpital Bichat, AP-HP, Université Paris Diderot, DHU FIRE, INSERM U1149, Paris, France
- Institut Micalis, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Eric Daugas
- Centre de Recherche sur l’Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d’Excellence Inflamex, DHU FIRE, Paris, France
- Service de Néphrologie, Hôpital Bichat, AP-HP, Université Paris Diderot, DHU FIRE, INSERM U1149, Paris, France
| |
Collapse
|
40
|
Pedersen HL, Horvei KD, Thiyagarajan D, Norby GE, Seredkina N, Moroni G, Eilertsen GØ, Holdaas H, Strøm EH, Bakland G, Meroni PL, Rekvig OP. Lupus nephritis: low urinary DNase I levels reflect loss of renal DNase I and may be utilized as a biomarker of disease progression. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2018; 4:193-203. [PMID: 29624903 PMCID: PMC6065113 DOI: 10.1002/cjp2.99] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 11/21/2022]
Abstract
Renal DNase I is lost in advanced stages of lupus nephritis. Here, we determined if loss of renal DNase I reflects a concurrent loss of urinary DNase I, and whether absence of urinary DNase I predicts disease progression. Mouse and human DNase I protein and DNase I endonuclease activity levels were determined by western blot, gel, and radial activity assays at different stages of the murine and human forms of the disease. Cellular localization of DNase I was analyzed by immunohistochemistry, immunofluorescence, confocal microscopy, and immunoelectron microscopy. We further compared DNase I levels in human native and transplanted kidneys to determine if the disease depended on autologous renal genes, or whether the nephritic process proceeded also in transplanted kidneys. The data indicate that reduced renal DNase I expression level relates to serious progression of lupus nephritis in murine, human native, and transplanted kidneys. Notably, silencing of renal DNase I correlated with loss of DNase I endonuclease activity in the urine samples. Thus, urinary DNase I levels may therefore be used as a marker of lupus nephritis disease progression and reduce the need for renal biopsies.
Collapse
Affiliation(s)
- Hege L Pedersen
- Department of Medical Biology, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Kjersti D Horvei
- Department of Medical Biology, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Dhivya Thiyagarajan
- Department of Medical Biology, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Gudrun E Norby
- Department of Rheumatology, OUH-Oslo University Hospital, Oslo, Norway
| | - Natalya Seredkina
- Department of Medical Biology, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Gabriella Moroni
- Nephrological Unit, Fondazione Ca' Granda IRCCS, Ospedale Maggiore, Milano, Italy
| | - Gro Ø Eilertsen
- Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Hallvard Holdaas
- Department of Medicine, OUH-Oslo University Hospital, Oslo, Norway
| | - Erik H Strøm
- Department of Pathology, OUH-Oslo University Hospital, Oslo, Norway
| | - Gunnstein Bakland
- Department of Rheumatology, University Hospital of North Norway, Tromsø, Norway
| | - Pier-Luigi Meroni
- Department of Clinical Sciences and Community Health, University of Milan and IRCCS, Istituto Auxologico Italiano, Milan, Italy
| | - Ole P Rekvig
- Department of Medical Biology, UiT-the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
41
|
Ferreira I, Croca S, Raimondo MG, Matharu M, Miller S, Giles I, Isenberg D, Ioannou Y, Hanly JG, Urowitz MB, Anderson N, Aranow C, Askanase A, Bae SC, Bernatsky S, Bruce IN, Buyon J, Clarke AE, Dooley MA, Fortin P, Ginzler E, Gladman D, Gordon C, Inanc M, Jacobsen S, Kalunian K, Kamen D, Khamashta M, Lim S, Manzi S, Merrill J, Nived O, Peschken C, Petri M, Ramsey-Goldman R, Ruiz-Irastorza G, Sanchez-Guerrero J, Steinson K, Sturfelt GK, van Vollenhoven R, Wallace DJ, Zoma A, Rahman A. Nitrated nucleosome levels and neuropsychiatric events in systemic lupus erythematosus; a multi-center retrospective case-control study. Arthritis Res Ther 2017; 19:287. [PMID: 29273092 PMCID: PMC5741886 DOI: 10.1186/s13075-017-1495-6] [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: 01/06/2017] [Accepted: 12/08/2017] [Indexed: 11/20/2022] Open
Abstract
Background In patients with systemic lupus erythematosus (SLE) there is no serological test that will reliably distinguish neuropsychiatric (NP) events due to active SLE from those due to other causes. Previously we showed that serum levels of nitrated nucleosomes (NN) were elevated in a small number of patients with NPSLE. Here we measured serum NN in samples from a larger population of patients with SLE and NP events to see whether elevated serum NN could be a marker for NPSLE. Methods We obtained serum samples from patients in the Systemic Lupus International Collaborative Clinics (SLICC) inception cohort. This included 216 patients with NP events and two matched controls with SLE but no NP events for each of these patients. For the NP patients we tested samples taken before, during and after the NP event. Results Twenty-six patients had events attributed to SLE according to the most stringent SLICC attribution rule. In these patients there was no association between onset of event and elevated serum NN. In 190 patients in whom events were not attributed to SLE by the SLICC rules, median serum NN was elevated at the onset of event (P = 0.006). The predominant clinical features in this group of 190 patients were headache, mood disorders and anxiety. Conclusions Serum NN levels rise at the time of an NP event in a proportion of patients with SLE. Further studies are needed to determine the value of serum NN as a biomarker for NPSLE.
Collapse
Affiliation(s)
- Isabel Ferreira
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK
| | - Sara Croca
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK
| | - Maria Gabriella Raimondo
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK
| | - Manjit Matharu
- Headache Group, Institute of Neurology and The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Sarah Miller
- Headache Group, Institute of Neurology and The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Ian Giles
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK
| | - David Isenberg
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK.,Arthritis Research UK Centre for Adolescent Rheumatology, UCL/UCLH/Great Ormond Street Hospital, London, UK
| | - Yiannis Ioannou
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK.,Arthritis Research UK Centre for Adolescent Rheumatology, UCL/UCLH/Great Ormond Street Hospital, London, UK
| | - John G Hanly
- Division of Rheumatology, Dalhousie University and Queen Elizabeth II Health Sciences Center, Halifax, NS, Canada
| | - Murray B Urowitz
- Lupus Program, Centre for Prognosis Studies in The Rheumatic Disease and Krembil Research Institute, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Nicole Anderson
- Lupus Program, Centre for Prognosis Studies in The Rheumatic Disease and Krembil Research Institute, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Cynthia Aranow
- Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Anca Askanase
- Rheumatology, Columbia University, New York, NY, USA
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Sasha Bernatsky
- Divisions of Clinical Epidemiology and Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| | - Ian N Bruce
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, The University of Manchester, and NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Jill Buyon
- New York School of Medicine, New York, NY, USA
| | - Ann E Clarke
- Division of Rheumatology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mary Anne Dooley
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Paul Fortin
- Centre Hospitalier de l'Université Laval (CHUL), Québec, QC, Canada
| | - Ellen Ginzler
- Downstate Medical Center Rheumatology, Brooklyn, New York, NY, USA
| | - Dafna Gladman
- Lupus Program, Centre for Prognosis Studies in The Rheumatic Disease and Krembil Research Institute, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Murat Inanc
- Department of Internal Medicine, Istanbul University, Istanbul, Turkey
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Centre For Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | | | - Diane Kamen
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Sam Lim
- Department of Medicine, Emory University, Atlanta, GA, USA
| | - Susan Manzi
- Allegheny Health Network, Pittsburgh, PA, USA
| | - Joan Merrill
- Clinical Pharmacology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ola Nived
- Department of Rheumatology, Lund University, Lund, Sweden
| | - Christine Peschken
- Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Michelle Petri
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Guillermo Ruiz-Irastorza
- Autoimmune Diseases Research Unit, Department of Internal Medicine, BioCruces Health Research Institute. Hospital Universitario Cruces, University of the Basque Country, Barakaldo, Bizkaia, Spain
| | - Jorge Sanchez-Guerrero
- Mount Sinai Hospital and University Health Network, University of Toronto, Toronto, ON, Canada
| | - Kristjan Steinson
- Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Ronald van Vollenhoven
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Sweden
| | - Daniel J Wallace
- Cedars-Sinai Medical Center/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Asad Zoma
- Department of Rheumatology Hairmyres Hospital, East Kilbride, Scotland, UK
| | - Anisur Rahman
- Centre for Rheumatology Research, University College London, Fourth Floor Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
42
|
Kong W, Fu J, Liu N, Jiao C, Guo G, Luan J, Wang H, Yao L, Wang L, Yamamoto M, Pi J, Zhou H. Nrf2 deficiency promotes the progression from acute tubular damage to chronic renal fibrosis following unilateral ureteral obstruction. Nephrol Dial Transplant 2017; 33:771-783. [PMID: 29126308 DOI: 10.1093/ndt/gfx299] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/04/2017] [Indexed: 01/24/2023] Open
Affiliation(s)
- Weiwei Kong
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Nan Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Congcong Jiao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Junjun Luan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Huihui Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Lining Wang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University of Medicine, Sendai, Japan
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Hua Zhou
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
43
|
Yung S, Chan TM. Molecular and Immunological Basis of Tubulo-Interstitial Injury in Lupus Nephritis: a Comprehensive Review. Clin Rev Allergy Immunol 2017; 52:149-163. [PMID: 26961386 DOI: 10.1007/s12016-016-8533-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lupus nephritis is an important cause of kidney failure in patients of Asian, African, or Hispanic descent. Its etiology and pathogenesis are multifactorial and remain to be elucidated. Accumulating evidence suggests that anti-double-stranded DNA (dsDNA) antibodies play a critical role in the pathogenesis, through its direct binding to cross-reactive antigens on resident renal cells or indirect binding through chromatin material to extracellular matrix components, resulting in complement activation, cell activation and proliferation, and induction of inflammatory and fibrotic processes. While tubulo-interstitial damage portends poor long-term renal prognosis, the mechanisms leading to tubulo-interstitial injury in lupus nephritis has received relatively less attention to date. Immune deposition along the tubular basement membrane is often observed in lupus nephritis and correlates with tubulo-interstitial infiltration of immune cells and interstitial fibrosis. Anti-dsDNA antibodies bind to resident renal cells, including proximal renal tubular epithelial cells, and contribute to renal inflammation and fibrosis. There is emerging evidence that epigenetic influence such as DNA methylation, histone modification, and microRNAs (miRs) also contribute to kidney fibrosis. Overexpression of miR-150 is observed in renal biopsies from patients with lupus nephritis and correlates with kidney fibrosis and chronicity score. Mycophenolate mofetil (MMF) is an established and effective standard-of-care therapy for patients with lupus nephritis. Accumulating data suggest that in addition to its immunosuppressive actions on lymphocyte proliferation, mycophenolic acid (MPA), the active metabolite of MMF, can exert a direct effect on nonimmune cells. Mediators of inflammation and fibrosis induced by anti-dsDNA antibodies in cultured proximal renal tubular epithelial cells are ameliorated by the addition of MPA, suggesting that in addition to its immunosuppressive actions, MPA may also have a beneficial effect in improving tubulo-interstitial inflammation and fibrosis through its direct action on proximal renal tubular epithelial cells.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Tak Mao Chan
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
44
|
Wu S, Wang J, Li F. Dysregulation of miRNA-146a contributes to the development of lupus nephritis via targeting of TRAF6. Per Med 2017; 14:131-139. [PMID: 29754557 DOI: 10.2217/pme-2016-0065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Aim: The objective of this study was to identify the association between genotypes of miR-146a rs2910164 and expression of TRAF6 as well as the risk of lupus nephritis (LN). Results: A total of 567 systemic lupus erythematosus patients both with and without LN were included in the study. The luciferase activity of cells that carried miR-146a mimics was much lower than control and the miR-146a mRNA expression with the GG SNP was significantly overexpressed compared with that in GC and CC groups. Expressions of TRAF6 mRNA and protein with GG were markedly lower than those in GC and CC groups. Mesangial cells treated with miR-146a inhibitors displayed higher expression of TRAF6 mRNA and protein compared with scramble control, miR-146a mimics and TRAF6 siRNA groups. Conclusion: Rs2910164 is associated with the risk of LN and could function as a therapeutic target of the disease.
Collapse
Affiliation(s)
- Shupeng Wu
- Department of Rheumatism & Immunology, Tai'an Central Hospital, Taian, China
| | - Jing Wang
- Department of Geriatric Diseases, Tai'an Central Hospital, Taian, China
| | - Fang Li
- Department of Rheumatism & Immunology, Tai'an Central Hospital, Taian, China
| |
Collapse
|
45
|
Ward F, Bargman JM. Membranous Lupus Nephritis: The Same, But Different. Am J Kidney Dis 2016; 68:954-966. [DOI: 10.1053/j.ajkd.2016.07.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/06/2016] [Indexed: 01/07/2023]
|
46
|
Kolkhir P, Pogorelov D, Olisova O, Maurer M. Comorbidity and pathogenic links of chronic spontaneous urticaria and systemic lupus erythematosus--a systematic review. Clin Exp Allergy 2016; 46:275-87. [PMID: 26545308 DOI: 10.1111/cea.12673] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chronic spontaneous urticaria (CSU) is a common mast cell-driven disease characterized by the development of wheals (hives), angioedema (AE), or both for > 6 weeks. It is thought that autoimmunity is a common cause of CSU, which is often associated with autoimmune thyroiditis, whereas the link to other autoimmune disorders such as systemic lupus erythematosus (SLE) has not been carefully explored. Here, we systematically reviewed the existing literature for information on the prevalence of CSU in SLE (and vice versa) and we examined the possible clinical and pathogenetic relationship between CSU and SLE. The prevalence of CSU and CSU-like rash in SLE was investigated by 42 independent studies and comorbidity in adult patients reportedly ranged from 0% to 21.9% and 0.4% to 27.5%, respectively (urticarial vasculitis: 0-20%). In children with SLE, CSU was reported in 0-1.2% and CSU-like rash in 4.5-12% (urticarial vasculitis: 0-2.2%). In contrast, little information is available on the prevalence of SLE in patients with CSU, and more studies are needed to determine the rate of comorbidity. Recent insights on IgG- and IgE-mediated autoreactivity suggest similarities in the pathogenesis of CSU and SLE linking inflammation and autoimmunity with the activation of the complement and coagulation system. Future studies of patients with either or both conditions could help to better define common pathomechanisms in CSU and SLE and to develop novel targeted treatment options for patients with CSU and SLE.
Collapse
Affiliation(s)
- P Kolkhir
- Department of Dermatology and Venereology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - D Pogorelov
- Department of Dermatology and Venereology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - O Olisova
- Department of Dermatology and Venereology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - M Maurer
- Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
47
|
Rekvig OP, Thiyagarajan D, Pedersen HL, Horvei KD, Seredkina N. Future Perspectives on Pathogenesis of Lupus Nephritis: Facts, Problems, and Potential Causal Therapy Modalities. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2772-2782. [PMID: 27664472 DOI: 10.1016/j.ajpath.2016.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/30/2016] [Indexed: 12/22/2022]
Abstract
Divergent incommensurable models have been developed to explain the pathogenesis of lupus nephritis. Most contemporary models favor a central role for anti-chromatin antibodies. How they exert their pathogenic effect has, however, endorsed conflicts that at least for now preclude insight into definitive pathogenic pathways. The following paradigms are contemporarily in conflict with each other: i) the impact of anti-double-stranded DNA (dsDNA) antibodies that cross-react with inherent renal antigens, ii) the impact of anti-dsDNA antibodies targeting exposed chromatin in glomeruli, and iii) the impact of relative antibody avidity for dsDNA, chromatin fragments, or cross-reacting antigens. Aside from these three themes, the pathogenic role of T cells in lupus nephritis is not clear. These different models should be tested through a collaboration between scientists belonging to the different paradigms. If it turns out that there are different pathogenic pathways in lupus nephritis, the emerging pathogenic mechanism(s) may be encountered with new individual causal therapy modalities. Today, therapy is still unspecific and far from interfering with the cause(s) of the disorder. This review attempts to describe what we know about processes that may cause lupus nephritis and how such basic processes may be affected if we can specifically interrupt them. Secondary inflammatory mechanisms, cytokine signatures, activation of complement, and other contributors to inflammation will not be discussed herein; rather, the events that trigger these factors will be discussed.
Collapse
Affiliation(s)
- Ole P Rekvig
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway; Department of Radiology, University Hospital of North Norway, Tromsø, Norway; Norwegian Center for Molecular Medicine, University of Oslo, Oslo, Norway.
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Hege L Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Kjersti D Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Natalya Seredkina
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
48
|
Pedersen HL, Horvei KD, Thiyagarajan D, Seredkina N, Rekvig OP. Murine and Human Lupus Nephritis: Pathogenic Mechanisms and Theoretical Strategies for Therapy. Semin Nephrol 2016; 35:427-38. [PMID: 26573545 DOI: 10.1016/j.semnephrol.2015.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lupus nephritis is one of the most serious manifestations of systemic lupus erythematosus, and represents one of the criteria implemented to classify systemic lupus erythematosus. Although studied for decades, no consensus has been reached related to the basic cellular, molecular, and immunologic mechanism(s) responsible for lupus nephritis. No causal treatments have been developed; therapy is approached mainly with nonspecific immunosuppressive medications. More detailed insight into disease mechanisms therefore is indispensable to develop new therapeutic strategies. In this review, contemporary knowledge on the pathogenic mechanisms of lupus nephritis is discussed based on recent data in murine and human lupus nephritis. Specific focus is given to the effect of anti-double-stranded DNA/antinucleosome antibodies in the kidneys and whether they bind exposed chromatin fragments in glomeruli or whether they bind inherent glomerular structures by cross-recognition. Overall, the data presented here favor the exposed chromatin model because we did not find any indication to substantiate the anti-double-stranded DNA antibody cross-reacting model. At the end of this review we present data on why chromatin fragments are expressed in the glomeruli of patients with lupus nephritis, and discuss how this knowledge can be used to direct the development of future therapies.
Collapse
Affiliation(s)
- Hege Lynum Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | - Kjersti Daae Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Natalya Seredkina
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Ole Petter Rekvig
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway; Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
49
|
Goilav B, Putterman C. The Role of Anti-DNA Antibodies in the Development of Lupus Nephritis: A Complementary, or Alternative, Viewpoint? Semin Nephrol 2016; 35:439-43. [PMID: 26573546 DOI: 10.1016/j.semnephrol.2015.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Kidney disease, or lupus nephritis, is the organ involvement that is most closely associated with specific autoantibodies in patients with SLE. The concept of anti-DNA antibodies being instrumental in the pathogenesis of lupus nephritis emerged ~50 years ago, and has been a topic of debate ever since. This article focuses on the description of the renal sub-cellular targets of nephritogenic autoantibodies and offers a counter-point opinion to the article by Pedersen et al. In addition, we provide an overview of some of the mechanisms by which anti-DNA antibodies bind to their renal targets and the pathogenic relevance to clinical nephritis.
Collapse
Affiliation(s)
- Beatrice Goilav
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY
| | - Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
50
|
Mistry P, Kaplan MJ. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol 2016; 185:59-73. [PMID: 27519955 DOI: 10.1016/j.clim.2016.08.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
Abstract
Nephritis is one of the most severe complications of systemic lupus erythematosus (SLE). One key characteristic of lupus nephritis (LN) is the deposition of immune complexes containing nucleic acids and/or proteins binding to nucleic acids and autoantibodies recognizing these molecules. A variety of cell death processes are implicated in the generation and externalization of modified nuclear autoantigens and in the development of LN. Among these processes, apoptosis, primary and secondary necrosis, NETosis, necroptosis, pyroptosis, and autophagy have been proposed to play roles in tissue damage and immune dysregulation. Cell death occurs in healthy individuals during conditions of homeostasis yet autoimmunity does not develop, at least in part, because of rapid clearance of dying cells. In SLE, accelerated cell death combined with a clearance deficiency may lead to the accumulation and externalization of nuclear autoantigens and to autoantibody production. In addition, specific types of cell death may modify autoantigens and alter their immunogenicity. These modified molecules may then become novel targets of the immune system and promote autoimmune responses in predisposed hosts. In this review, we examine various cell death pathways and discuss how enhanced cell death, impaired clearance, and post-translational modifications of proteins could contribute to the development of lupus nephritis.
Collapse
Affiliation(s)
- Pragnesh Mistry
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|