1
|
McGovern DP, Jones RB, Jayne DRW, Smith RM. The Expanding Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Armamentarium. Drugs 2025; 85:325-341. [PMID: 39969779 DOI: 10.1007/s40265-024-02143-z] [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: 12/15/2024] [Indexed: 02/20/2025]
Abstract
The complex pathophysiology of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) is reflected in the heterogeneity of the presenting clinical syndromes caused by these diseases but also provides a variety of conceivable molecular and cellular targets that can be therapeutically manipulated. The last decade has seen an expansion of established and potential therapies for treating AAV, some of which target the dysfunctional autoreactive immune response and others aim to ameliorate the downstream consequences of local vascular inflammation and necrosis. The success and widespread adoption of the anti-CD20 monoclonal antibody, rituximab, as an agent to both induce and maintain remission, has heralded a change in the standard-of-care management of AAV, replacing the "old guard" combination of cyclophosphamide and high-dose corticosteroids established in the 1970s. The development and approval of avacopan, a first-in-class small-molecule antagonist to the main receptor for the complement anaphylatoxin C5a, has the potential to reduce the corticosteroid burden experienced by patients with AAV and may also improve outcomes for those with AAV kidney disease. It marks the culmination of almost 20 years of international collaboration, from understanding the pathological role of complement in basic murine models of AAV through to a phase III clinical trial, and emphasises the importance of following promising translational discoveries through to drug development and clinical deployment. This article summarises how recent progress in our understanding of the basic pathophysiology of AAV has resulted in the development of new and effective treatments and, reciprocally, how studying the impact of these treatments in patients has advanced our understanding of dysfunctional immunobiology in disease.
Collapse
Affiliation(s)
- Dominic P McGovern
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK.
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Rachel B Jones
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David R W Jayne
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rona M Smith
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
2
|
Li M, Hao X, Shi D, Cheng S, Zhong Z, Cai L, Jiang M, Ding L, Ding L, Wang C, Yu X. Identification of susceptibility loci and relevant cell type for IgA nephropathy in Han Chinese by integrative genome-wide analysis. Front Med 2024; 18:862-877. [PMID: 39343836 DOI: 10.1007/s11684-024-1086-2] [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: 11/22/2023] [Accepted: 05/17/2024] [Indexed: 10/01/2024]
Abstract
Although many susceptibility loci for IgA nephropathy (IgAN) have been identified, they only account for 11.0% of the overall IgAN variance. We performed a large genome-wide meta-analysis of IgAN in Han Chinese with 3616 cases and 10 417 controls to identify additional genetic loci of IgAN. Considering that inflammatory bowel disease (IBD) and asthma might share an etiology of dysregulated mucosal immunity with IgAN, we performed cross-trait integrative analysis by leveraging functional annotations of relevant cell type and the pleiotropic information from IBD and asthma. Among 8 669 456 imputed variants, we identified a novel locus at 4p14 containing the long noncoding RNA LOC101060498. Cell type enrichment analysis based on annotations suggested that PMA-I-stimulated CD4+CD25-IL17+ Th17 cell was the most relevant cell type for IgAN, which highlights the essential role of Th17 pathway in the pathogenesis of IgAN. Furthermore, we identified six more novel loci associated with IgAN, which included three loci showing pleiotropic effects with IBD or asthma (2q35/PNKD, 6q25.2/SCAF8, and 22q11.21/UBE2L3) and three loci specific to IgAN (14q32.32/TRAF3, 16q22.2/TXNL4B, and 21q21.3/LINC00113) in the pleiotropic analysis. Our findings support the involvement of mucosal immunity, especially T cell immune response and IL-17 signal pathway, in the development of IgAN and shed light on further investigation of IgAN.
Collapse
Affiliation(s)
- Ming Li
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dianchun Shi
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China
| | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhong Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Nephrology (Sun Yat-sen University), and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Lu Cai
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Nephrology (Sun Yat-sen University), and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Minghui Jiang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin Ding
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lanbo Ding
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xueqing Yu
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Williams CEC, Lamond M, Marro J, Chetwynd AJ, Oni L. A narrative review of potential drug treatments for nephritis in children with IgA vasculitis (HSP). Clin Rheumatol 2023; 42:3189-3200. [PMID: 37755547 PMCID: PMC10640478 DOI: 10.1007/s10067-023-06781-8] [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: 06/19/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023]
Abstract
Immunoglobulin A (IgA) vasculitis (IgAV, also known as Henoch-Schoenlein purpura, HSP) is the most common vasculitis of childhood. It usually presents with a simple, self-limiting disease course; however, a small subset of patients may develop kidney involvement (IgAV-N) which occurs 4-12 weeks after disease onset and is the biggest contributor to long-term morbidity. Treatment currently targets patients with established kidney involvement; however; there is a desire to work towards early prevention of inflammation during the window of opportunity between disease presentation and onset of significant nephritis. There are no clinical trials evaluating drugs which may prevent or halt the progression of nephritis in children with IgAV apart from the early use of corticosteroids which have no benefit. This article summarises the latest scientific evidence and clinical trials that support potential therapeutic targets for IgAV-N that are currently being developed based on the evolving understanding of the pathophysiology of IgAV-N. These span the mucosal immunity, B-cell and T-cell modulation, RAAS inhibition, and regulation of complement pathways, amongst others. Novel drugs that may be considered for use in early nephritis include TRF-budesonide; B-cell inhibiting agents including belimumab, telitacicept, blisibimod, VIS649, and BION-1301; B-cell depleting agents such as rituximab, ofatumumab, and bortezomib; sparsentan; angiotensin converting enzyme inhibitors (ACE-Is); and complement pathway inhibitors including avacopan, iptacopan, and narsoplimab. Further clinical trials, as well as pre-clinical scientific studies, are needed to identify mechanistic pathways as there may be an opportunity to prevent nephritis in this condition. Key Points • Kidney involvement is the main cause of long-term morbidity and mortality in IgA vasculitis despite the current treatment recommendations. • The evolving understanding of the pathophysiology of IgA vasculitis is allowing exploration of novel treatment options which target underlying immune pathways. • Novel treatments currently being trialled in IgA nephropathy may have benefit in IgA vasculitis due to the similarities in the underlying pathophysiology, such as TRF-budesonide, B-cell modulators, and complement inhibitors. • Further studies, including clinical trials of novel drugs, are urgently needed to improve the long-term outcomes for children with IgA vasculitis nephritis.
Collapse
Affiliation(s)
- Chloe E C Williams
- Royal Liverpool and Broadgreen University Hospital Trusts, Liverpool, UK
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Megan Lamond
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Julien Marro
- School of Medicine, University of Liverpool, Liverpool, UK
| | - Andrew J Chetwynd
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Louise Oni
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Paediatric Nephrology, Institute in the Park Building, University of Liverpool, Alder Hey Children's NHS Foundation Trust Hospital, Eaton Road, Liverpool, L12 2AP, UK.
| |
Collapse
|
4
|
Ostendorf L, Burns M, Wagner DL, Enghard P, Amann K, Mei H, Eckardt KU, Seelow E, Schreiber A. Daratumumab for the treatment of refractory ANCA-associated vasculitis. RMD Open 2023; 9:rmdopen-2022-002742. [PMID: 36627149 PMCID: PMC9835944 DOI: 10.1136/rmdopen-2022-002742] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Objective Treatment-refractory antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a life-threatening condition without evidence-based treatment options. One emerging treatment option for several antibody-mediated autoimmune diseases is the anti-CD38 antibody daratumumab, which depletes autoantibody-secreting plasma cells.Methods We treated two patients with severe life-threatening AAV with renal and pulmonary manifestation despite induction therapy with rituximab and cyclophosphamide with four to eight doses of 1800 mg daratumumab. We followed clinical and immunological responses.Results The first patient with myeloperoxidase-ANCA-positive microscopic polyangiitis had resolution of pneumonitis and pleuritis and stabilisation of kidney function after daratumumab. The second patient with proteinase 3-ANCA-positive granulomatosis with polyangiitis, diffuse alveolar haemorrhage necessitating extracorporeal membrane oxygenation (ECMO) and acute kidney failure, requiring kidney replacement therapy, was weaned off ECMO, mechanical ventilation and dialysis and discharged home after daratumumab. Clinical improvement was paralleled by a strong reduction in serum ANCA levels as well as total IgG, indicating depletion of plasma cells. Apart from the depletion of CD38+ natural killer cells, blood leucocyte levels were not notably influenced by daratumumab. Only mild adverse events, such as hypogammaglobulinaemia and an upper respiratory tract infection occurred.Conclusion Daratumumab was safe and effective in inducing remission in two patients with severe treatment-refractory AAV, warranting prospective clinical trials to establish safety and efficacy.
Collapse
Affiliation(s)
- Lennard Ostendorf
- Department of Nephrology and Medical Intensive Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany .,Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Marie Burns
- Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Dimitrios Laurin Wagner
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany,Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany,Institute of Transfusion Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Henrik Mei
- Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Evelyn Seelow
- Department of Nephrology and Medical Intensive Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adrian Schreiber
- Department of Nephrology and Medical Intensive Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
Molnár A, Studinger P, Ledó N. Diagnostic and Therapeutic Approach in ANCA-Associated Glomerulonephritis: A Review on Management Strategies. Front Med (Lausanne) 2022; 9:884188. [PMID: 35721093 PMCID: PMC9205443 DOI: 10.3389/fmed.2022.884188] [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: 02/25/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis is a destructive small vessel vasculitis affecting multiple organs. Renal involvement often leads to end-stage renal disease and increases mortality. Prompt diagnosis and initiation of adequate immunosuppressive therapy are critical for the best patient and kidney outcomes. However, considerable heterogeneity in symptoms and severity across the patients frequently hinder the diagnosis and management. The objective of this review is to emphasize the heterogeneity of the ANCA-associated vasculitis, facilitate the recognition and give guidance to the therapeutical possibilities. We present epidemiologic and risk factors, pathogenesis, and provide comprehensive clinical features of the disease. This article also focuses on the currently available therapeutic options and emerging cellular and molecular targets for the management of systemic and especially renal disease. We conducted extensive literature research published on PubMed and Google Scholar. We systematically reviewed, analyzed, and assembled databases, covering a broad spectrum of aspects of the disease. We compared and summarized the recommendations of two recent guidelines on ANCA-associated vasculitis. The incidence of ANCA-associated vasculitis, hence glomerulonephritis shows a steady increase. Familiarity with the presenting symptoms and laboratory abnormalities are necessary for rapid diagnosis. Early initiation of treatment is the key aspect for favorable patient and renal outcomes. A better understanding of the pathogenesis constantly leads to more targeted and therefore more efficient and less toxic treatment.
Collapse
Affiliation(s)
| | | | - Nóra Ledó
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Ahmadian E, Khatibi SMH, Vahed SZ, Ardalan M. Novel treatment options in rituximab-resistant membranous nephropathy patients. Int Immunopharmacol 2022; 107:108635. [DOI: 10.1016/j.intimp.2022.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/28/2022]
|
7
|
Canales-Herrerias P, Crickx E, Broketa M, Sokal A, Chenon G, Azzaoui I, Vandenberghe A, Perima A, Iannascoli B, Richard-Le Goff O, Castrillon C, Mottet G, Sterlin D, Robbins A, Michel M, England P, Millot GA, Eyer K, Baudry J, Mahevas M, Bruhns P. High-affinity autoreactive plasma cells disseminate through multiple organs in patients with immune thrombocytopenic purpura. J Clin Invest 2022; 132:153580. [PMID: 35503254 PMCID: PMC9197514 DOI: 10.1172/jci153580] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
The major therapeutic goal for immune thrombocytopenic purpura (ITP) is to restore normal platelet counts using drugs to promote platelet production or by interfering with mechanisms responsible for platelet destruction. Eighty percent of patients with ITP possess anti–integrin αIIbβ3 IgG autoantibodies that cause platelet opsonization and phagocytosis. The spleen is considered the primary site of autoantibody production by autoreactive B cells and platelet destruction. The immediate failure in approximately 50% of patients to recover a normal platelet count after anti-CD20 rituximab-mediated B cell depletion and splenectomy suggests that autoreactive, rituximab-resistant, IgG-secreting B cells (IgG-SCs) reside in other anatomical compartments. We analyzed more than 3,300 single IgG-SCs from spleen, bone marrow, and/or blood of 27 patients with ITP, revealing high interindividual variability in affinity for αIIbβ3, with variations over 3 logs. IgG-SC dissemination and range of affinities were, however, similar for each patient. Longitudinal analysis of autoreactive IgG-SCs upon treatment with the anti-CD38 mAb daratumumab demonstrated variable outcomes, from complete remission to failure with persistence of high-affinity anti–αIIbβ3 IgG-SCs in the bone marrow. This study demonstrates the existence and dissemination of high-affinity autoreactive plasma cells in multiple anatomical compartments of patients with ITP that may cause the failure of current therapies.
Collapse
Affiliation(s)
| | - Etienne Crickx
- INSERM U1151/CNRS UMS8253, Institut Necker-Enfants Malades, Paris, France
| | - Matteo Broketa
- Department of Immunology, Institut Pasteur, Paris, France
| | - Aurélien Sokal
- INSERM U1151/CNRS UMS8253, Institut Necker-Enfants Malades, Paris, France
| | - Guilhem Chenon
- Laboratoire Colloïdes et Matériaux Divisés (LCMD), ESPCI, Paris, France
| | - Imane Azzaoui
- Service de Médecine Interne, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, Paris, France
| | - Alexis Vandenberghe
- Service de Médecine Interne, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, Paris, France
| | - Angga Perima
- Department of Immunology, Institut Pasteur, Paris, France
| | | | | | | | | | | | - Ailsa Robbins
- Department of Internal Medicine, Infectious Diseases, and Clinical Immunolo, Robert Debré Hospital, Reims University Hospitals, Reims, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Créteil, France
| | - Patrick England
- Plateforme de Biophysique Moléculaire, Institut Pasteur, Paris, France
| | - Gael A Millot
- Hub Bioinformatique et Biostatistique, Institut Pasteur, Paris, France
| | - Klaus Eyer
- Laboratory for Functional Immune Repertoire Analysis, ETH Zurich, Zurich, Switzerland
| | - Jean Baudry
- Laboratoire Colloïdes et Matériaux Divisés (LCMD), ESPCI, Paris, France
| | - Matthieu Mahevas
- INSERM U1151/CNRS UMS8253, Institut Necker-Enfants Malades, Paris, France
| | | |
Collapse
|
8
|
B cells in autoimmune hepatitis: bystanders or central players? Semin Immunopathol 2022; 44:411-427. [PMID: 35488094 PMCID: PMC9256567 DOI: 10.1007/s00281-022-00937-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
B cells are central for the adaptive immune system to mount successful immune responses not only as antibody producers but also as regulators of cellular immunity. These multifaceted features are also reflected in autoimmunity where autoreactive B cells can fuel disease by production of cytotoxic autoantibodies, presentation of autoantigens to autoreactive T cells, and secretion of cytokines and chemokines that either promote detrimental immune activation or impair regulatory T and B cells. The role of B cells and autoantibodies in autoimmune hepatitis (AIH) have been controversially discussed, with typical autoantibodies and hypergammaglobulinemia indicating a key role, while strong HLA class II association suggests T cells as key players. In this review, we summarize current knowledge on B cells in AIH and how different B cell subpopulations may drive AIH progression beyond autoantibodies. We also discuss recent findings of B cell-directed therapies in AIH.
Collapse
|
9
|
Merino-Vico A, van Hamburg JP, Tas SW. B Lineage Cells in ANCA-Associated Vasculitis. Int J Mol Sci 2021; 23:387. [PMID: 35008813 PMCID: PMC8745114 DOI: 10.3390/ijms23010387] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease that affects small sized blood vessels and can lead to serious complications in the lungs and kidneys. The prominent presence of ANCA autoantibodies in this disease implicates B cells in its pathogenesis, as these are the precursors of the ANCA-producing plasma cells (PCs). Further evidence supporting the potential role of B lineage cells in vasculitis are the increased B cell cytokine levels and the dysregulated B cell populations in patients. Confirmation of the contribution of B cells to pathology arose from the beneficial effect of anti-CD20 therapy (i.e., rituximab) in AAV patients. These anti-CD20 antibodies deplete circulating B cells, which results in amelioration of disease. However, not all patients respond completely, and this treatment does not target PCs, which can maintain ANCA production. Hence, it is important to develop more specific therapies for AAV patients. Intracellular signalling pathways may be potential therapeutic targets as they can show (disease-specific) alterations in certain B lineage cells, including pathogenic B cells, and contribute to differentiation and survival of PCs. Preliminary data on the inhibition of certain signalling molecules downstream of receptors specific for B lineage cells show promising therapeutic effects. In this narrative review, B cell specific receptors and their downstream signalling molecules that may contribute to pathology in AAV are discussed, including the potential to therapeutically target these pathways.
Collapse
Affiliation(s)
- Ana Merino-Vico
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sander W. Tas
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
10
|
Villacorta J, Martinez-Valenzuela L, Martin-Capon I, Bordignon-Draibe J. Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: Toward an Individualized Approach. Nephron Clin Pract 2021; 146:121-137. [PMID: 34915506 DOI: 10.1159/000520727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/29/2021] [Indexed: 11/19/2022] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), characterized by the presence of autoantibodies to neutrophil cytoplasmic antigens, proteinase 3 (PR3), and myeloperoxidase (MPO), typically involves small blood vessels of the respiratory tract and kidneys. It includes distinct clinical syndromes: microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), and eosinophilic GPA. GPA is commonly associated with PR3-ANCA, while MPA is associated with MPO-ANCA. AAVs have a complex pathogenesis, influenced by genetics and environmental factors. There is evidence for a loss of tolerance to neutrophil proteins, which leads to ANCA-mediated neutrophil activation and injury, with effector T cells and activation of the alternative pathway of the complement also involved. Advances in immunosuppressive treatment have drastically reduced mortality of AAV in the past decades, opting for a more individualized approach. Careful assessment of ANCA specificity, disease activity, organ damage, and quality of life allows for a tailored immunosuppressive therapy. Contemporary AAV treatment is characterized by regimens that minimize the cumulative exposure to glucocorticoids and cyclophosphamide, and novel approaches including complement blockage and immunosuppressant combinations might be the standard of care in the future. In this review, we examine the pathogenesis, clinical approach, and evidence-based treatment options for the management of AAV patients.
Collapse
Affiliation(s)
- Javier Villacorta
- Nephrology Department, Ramón y Cajal University Hospital, IRYCIS, Alcala University, Madrid, Spain
| | - Laura Martinez-Valenzuela
- Nephrology Department, Bellvitge University Hospital, IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, Barcelona, Spain
| | - Irene Martin-Capon
- Nephrology Department, Ramón y Cajal University Hospital, IRYCIS, Alcala University, Madrid, Spain
| | - Juliana Bordignon-Draibe
- Nephrology Department, Bellvitge University Hospital, IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
11
|
Hu J, Wu W, Yu M, Xia Z, Gao C. MicroRNA-340-5p inhibits endothelial apoptosis, inflammatory response, and pro-coagulation by targeting KDM4C in anti-neutrophil cytoplasmic antibody (ANCA)-mediated glomerulonephritis through activation of B cells. Autoimmunity 2021; 54:343-352. [PMID: 34121556 DOI: 10.1080/08916934.2021.1937609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis, a class of systemic autoimmune diseases, results in damage of various critical organs including kidneys, lungs, eyes, and nervous system. MicroRNA-340-5p was confirmed to be downregulated in autoimmune pathogenesis. However, the role of miR-340-5p remains unknown in ANCA-induced glomerulonephritis (GN). The current study aimed to explore the role of miR-340-5p in ANCA-induced GN. The animal models of ANCA-induced GN was established by experimental autoimmune vasculitis (EAV) operation. The primary glomerular endothelial cells (PGEnCs) were treated with anti-myeloperoxidase (anti-MPO) to mimic cell injury in vitro. The renal function was analysed by measuring serum creatinine, blood urea nitrogen, urine blood, urine protein and urine leukocytes. The levels of RNA and proteins were examined by RT-qPCR and western blot analysis, respectively. The binding capacity between miR-340-5p and KDM4C was detected by luciferase reporter assay. Cell apoptosis was analysed by flow cytometry in vitro. Cell viability was determined by CCK-8 assay. The cleaved caspase-3 activity was analysed by immunofluorescent assay. Cell inflammation was measured by western blot. Cell procoagulant activity was assessed by FXa generation assay. The histological changes of renal tissues were assessed by Haematoxylin and eosin (H&E) staining assay. The correlation between miR-340-5p and KDM4C level (or content of TNF-α and IL-6) was analysed by Pearson correlation analysis. The injection of anti-MPO IgG induced a significant elevation of Serum creatinine and blood urea nitrogen in serum, as well as urine blood, urine protein and urine leukocytes. Importantly, KDM4C was downregulated in model group. In mechanism, we identified that miR-340-5p bound with KDM4C 3'untranslated region (UTR), negatively regulated KDM4C in endothelial cells and negatively correlated with KDM4C in serum of GN rats. In function, we found that miR-340-5p promoted B cell activation and proliferation by downregulating KDM4C. The in vitro assays showed that the decrease of cell viability induced by anti-MPO was reversed by miR-340-5p overexpression, and further reduced by KDM4C overexpression. Inversely, the suppressive effects of miR-340-5p mimics on cell apoptosis, cleaved caspase-3 activity, inflammatory response and pro-coagulation were countervailed by KDM4C overexpression in anti-MPO-treated cells. The in vivo assays validated that miR-340-5p overexpression mitigated the impairment of renal function, and histological changes induced by anti-MPO IgG injection in model group. Finally, we found the negative correlation between miR-340-5p and TNF-α (or IL-6) content in serum of GN rats. In conclusion, we found that miR-340-5p inhibited endothelial apoptosis and inflammatory response by targeting KDM4C in ANCA-mediated GN through activation of B cells, implying a potential novel insight for treatment of ANCA-mediated GN.
Collapse
Affiliation(s)
- Jian Hu
- Department of Pediatrics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Wu
- Department of Pediatrics, Longgang District Central Hospital of Shenzhen, Shenzhen, China
| | - Min Yu
- Department of Pediatrics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Zhengkun Xia
- Department of Pediatrics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Chunlin Gao
- Department of Pediatrics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Du W, Han M, Zhu X, Xiao F, Huang E, Che N, Tang X, Zou H, Jiang Q, Lu L. The Multiple Roles of B Cells in the Pathogenesis of Sjögren's Syndrome. Front Immunol 2021; 12:684999. [PMID: 34168653 PMCID: PMC8217880 DOI: 10.3389/fimmu.2021.684999] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Primary Sjögren’s syndrome (pSS) is a chronic autoimmune disease characterized by lymphocytic infiltration and tissue destruction of exocrine glands such as salivary glands. Although the formation of ectopic lymphoid tissue in exocrine glands and overproduction of autoantibodies by autoreactive B cells highlight the critical involvement of B cells in disease development, the precise roles of various B cell subsets in pSS pathogenesis remain partially understood. Current studies have identified several novel B cell subsets with multiple functions in pSS, among which autoreactive age-associated B cells, and plasma cells with augmented autoantibody production contribute to the disease progression. In addition, tissue-resident Fc Receptor-Like 4 (FcRL4)+ B cell subset with enhanced pro-inflammatory cytokine production serves as a key driver in pSS patients with mucosa-associated lymphoid tissue (MALT)-lymphomas. Recently, regulatory B (Breg) cells with impaired immunosuppressive functions are found negatively correlated with T follicular helper (Tfh) cells in pSS patients. Further studies have revealed a pivotal role of Breg cells in constraining Tfh response in autoimmune pathogenesis. This review provides an overview of recent advances in the identification of pathogenic B cell subsets and Breg cells, as well as new development of B-cell targeted therapies in pSS patients.
Collapse
Affiliation(s)
- Wenhan Du
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Man Han
- Division of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital and Fudan University, Shanghai, China
| | - Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Enyu Huang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Nan Che
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Xiaopo Tang
- Division of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital and Fudan University, Shanghai, China
| | - Quan Jiang
- Division of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.,Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
13
|
Zhu S, Hou S, Lu Y, Sheng W, Cui Z, Dong T, Feng H, Wan Q. USP36-Mediated Deubiquitination of DOCK4 Contributes to the Diabetic Renal Tubular Epithelial Cell Injury via Wnt/β-Catenin Signaling Pathway. Front Cell Dev Biol 2021; 9:638477. [PMID: 33968925 PMCID: PMC8102983 DOI: 10.3389/fcell.2021.638477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/03/2021] [Indexed: 01/11/2023] Open
Abstract
Diabetic kidney disease (DKD) has become the leading cause of end-stage renal disease but the efficacy of current treatment remains unsatisfactory. The pathogenesis of DKD needs a more in-depth research. Ubiquitin specific proteases 36 (USP36), a member of deubiquitinating enzymes family, has aroused wide concerns for its role in deubiquitinating and stabilizing target proteins. Nevertheless, the role of USP36 in diabetes has never been reported yet. Herein, we identified an increased expression of USP36 both in vitro and in vivo in diabetic renal tubular epithelial cells (TECs), and its overexpression is related to the enhanced epithelial-to-mesenchymal transition (EMT). Further investigation into the mechanisms proved that USP36 could directly bind to and mediate the deubiquitination of dedicator of cytokinesis 4 (DOCK4), a guanine nucleotide exchange factor (GEF) that could activate Wnt/β-catenin signaling pathway and induce EMT. Our study revealed a new mechanism that USP36 participates in the pathogenesis of DKD, and provided potential intervening targets accordingly.
Collapse
Affiliation(s)
- Suwei Zhu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaoshuai Hou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Lu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Sheng
- Department of Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhengguo Cui
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tianyi Dong
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hong Feng
- Department of Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiang Wan
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
14
|
Prendecki M, McAdoo SP. New Therapeutic Targets in Antineutrophil Cytoplasm Antibody–Associated Vasculitis. Arthritis Rheumatol 2021; 73:361-370. [DOI: 10.1002/art.41407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
|
15
|
An HJ, Jang HN, Lee TW, Yoon C, Park DJ, Bae E. A case of vasculitis case with unusual renal pathological findings presenting with rapidly progressing renal failure. Clin Case Rep 2020; 8:2852-2856. [PMID: 33363836 PMCID: PMC7752485 DOI: 10.1002/ccr3.3277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/01/2020] [Accepted: 07/28/2020] [Indexed: 11/07/2022] Open
Abstract
Plasma cell infiltration may be an early lesion during renal invasion by ANCA-associated vasculitis (AAV), and AAV should be considered in the differential diagnosis with acute renal failure and systemic symptoms.
Collapse
Affiliation(s)
- Hyo Jung An
- Department of PathologyGyeongsang National University Changwon HospitalChangwonKorea
| | - Ha Nee Jang
- Department of Internal MedicineGyeongsang National College of Medicine and Gyeongsang National University HospitalJinjuKorea
| | - Tae Won Lee
- Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University Changwon HospitalChangwonKorea
| | - Changhyo Yoon
- Department of NeurologyGyeongsang National University Changwon HospitalGyeongsang National University School of MedicineJinjuKorea
| | - Dong Jun Park
- Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University Changwon HospitalChangwonKorea
| | - Eunjin Bae
- Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University Changwon HospitalChangwonKorea
| |
Collapse
|
16
|
Maruyama H, Hirayama K, Yamashita M, Ohgi K, Tsujimoto R, Takayasu M, Shimohata H, Kobayashi M. Serum 20S proteasome levels are associated with disease activity in MPO-ANCA-associated microscopic polyangiitis. BMC Rheumatol 2020; 4:36. [PMID: 32864569 PMCID: PMC7447580 DOI: 10.1186/s41927-020-00137-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/05/2020] [Indexed: 11/10/2022] Open
Abstract
Background Proteasomes are found in both the cell nucleus and cytoplasm and play a major role in the ubiquitin-dependent and -independent non-lysosomal pathways of intracellular protein degradation. Proteasomes are also involved in the turnover of various regulatory proteins, antigen processing, cell differentiation, and apoptosis. To determine the diagnostic value of serum proteasome in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), we investigated patients with AAV at various stages of the disease. Methods Serum 20S-proteasome was measured by ELISA in 44 patients with MPO-ANCA-associated microscopic polyangiitis (MPA) and renal involvement. Thirty of the patients provided serum samples before the initial treatment, and 30 provided samples during remission; 16 provided samples at both time points. Results The mean serum 20S-proteasome level was significantly higher in the active-vasculitis patients (3414.6 ± 2738.9 ng/mL; n = 30) compared to the inactive-vasculitis patients (366.4 ± 128.4 ng/mL; n = 30; p < 0.0001) and 40 controls (234.9 ± 90.1 ng/mL; p < 0.0001). There were significant positive correlations between the serum 20S-proteasome level and the Birmingham Vasculitis Activity Score (BVAS) (r = 0.581, p < 0.0001), the ANCA titer (r = 0.384, p < 0.0001), the white blood cell (WBC) count (r = 0.284, p = 0.0042), the platelet count (r = 0.369, p = 0.0002), and the serum C-reactive protein (CRP) level (r = 0.550, p < 0.0001). There were significant negative correlations between the serum 20S-proteasome level and both the hemoglobin concentration (r = - 0.351, p = 0.0003) and the serum albumin level (r = - 0.460, p < 0.0001). In a multiple regression analysis, there was a significant positive correlation between the serum 20S-proteasome level and only the BVAS results (β = 0.851, p = 0.0009). In a receiver operating curve analysis, the area under the curve for the serum 20S-proteasome level was 0.996, which is higher than those of the WBC count (0.738) and the serum CRP level (0.963). Conclusion The serum level of 20S-proteasome may be a useful marker for disease activity in AAV.
Collapse
Affiliation(s)
- Hiroshi Maruyama
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| | - Kouichi Hirayama
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| | - Marina Yamashita
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| | - Kentaro Ohgi
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan.,Department of Intensive Care Medicine, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Ryuji Tsujimoto
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan.,Department of Nephrology, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Mamiko Takayasu
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| | - Homare Shimohata
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| | - Masaki Kobayashi
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuo, Ami, Ibaraki, 300-0395 Japan
| |
Collapse
|
17
|
Li SS, Wang SL, Zhang HJ, He SH, Liang X, Li TF. The pathogenesis and treatment in antineutrophil cytoplasmic antibody associated vasculitis. Am J Transl Res 2020; 12:4094-4107. [PMID: 32913491 PMCID: PMC7476117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/04/2020] [Indexed: 06/11/2023]
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV) is a systemic autoimmune disease characterized by leukocytoclastic inflammation of small blood vessels. Commonly detected autoantibodies include anti-protease 3 (PR3) and anti-myeloperoxidase (MPO). Although cell necrosis plays an important role in the production of autoantibodies and the pathogenesis of AAV, the correlation between their titers and disease activity remains elusive. As improved detection techniques facilitate early diagnosis, a satisfactory efficacy can be achieved in patients with mild to medium severe AAV treated with glucocorticoids and immunosuppressants. However, resistant and relapsing AAV, sometimes life-threatening, do exist in clinical practice. In-depth understanding of pathogenesis of AAV may lend novel insight into the mechanism responsible for its formation and help find effective targeted therapies for refractory patients.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| | - Shuo-Lin Wang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| | - Hui-Juan Zhang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| | - Shi-Hao He
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| | - Xu Liang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| | - Tian-Fang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University Zhengzhou, China
| |
Collapse
|
18
|
Shochet L, Holdsworth S, Kitching AR. Animal Models of ANCA Associated Vasculitis. Front Immunol 2020; 11:525. [PMID: 32373109 PMCID: PMC7179669 DOI: 10.3389/fimmu.2020.00525] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/09/2020] [Indexed: 01/05/2023] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV) is a rare and severe autoimmune multisystemic disease. Its pathogenesis involves multiple arms of the immune system, as well as complex interactions between immune cells and target organs. Experimental animal models of disease can provide the crucial link from human disease to translational research into new therapies. This is particularly true in AAV, due to low disease incidence and substantial disease heterogeneity. Animal models allow for controlled environments in which disease mechanisms can be defined, without the clinical confounders of environmental and lifestyle factors. To date, multiple animal models have been developed, each of which shed light on different disease pathways. Results from animal studies of AAV have played a crucial role in enhancing our understanding of disease mechanisms, and have provided direction toward newer targeted therapies. This review will summarize our understanding of AAV pathogenesis as has been gleaned from currently available animal models, as well as address their strengths and limitations. We will also discuss the potential for current and new animal models to further our understanding of this important condition.
Collapse
Affiliation(s)
- Lani Shochet
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia
| | - Stephen Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia.,Department of Immunology, Monash Health, Clayton, VIC, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia.,Department of Pediatric Nephrology, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
19
|
Is There a Future for Anti-CD38 Antibody Therapy in Systemic Autoimmune Diseases? Cells 2019; 9:cells9010077. [PMID: 31892266 PMCID: PMC7016693 DOI: 10.3390/cells9010077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
CD38 is a type II glycoprotein highly expressed on plasmablasts, short-lived and long-lived plasma cells, but weakly expressed on other lymphoid cells, myeloid cells and non-hematopoietic cells. This expression pattern makes CD38 an interesting target for a targeted therapy aiming to deplete antibody-producing plasma cells. We present data suggesting that anti-CD38 therapy may be effective for the prevention at the preclinical stage and for the treatment of established autoimmune diseases, such as systemic lupus erythematosus, systemic sclerosis, Sjögren’s syndrome and anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Given the high unmet need for efficacious disease-modifying treatment in these diseases, studies are warranted to determine if anti-CD38 antibody-based therapies may delay or prevent the disease progression of systemic autoimmune diseases.
Collapse
|
20
|
|
21
|
Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol 2018; 15:91-101. [DOI: 10.1038/s41584-018-0145-y] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Hutton HL, Holdsworth SR, Kitching AR. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin Nephrol 2018; 37:418-435. [PMID: 28863790 DOI: 10.1016/j.semnephrol.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Our understanding of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis has developed greatly since the discovery of ANCA, directed against neutrophil components, in 1982. Observations in human disease, and increasingly sophisticated studies in vitro and in rodent models in vivo, have allowed a nuanced understanding of many aspects of the immunopathogenesis of disease, including the significance of ANCA as a diagnostic and monitoring tool as well as a mediator of microvascular injury. The mechanisms of leukocyte recruitment and tissue injury, and the role of T cells increasingly are understood. Unexpected findings, such as the role of complement, also have been uncovered through experimental studies and human observations. This review focusses on the pathogenesis of ANCA-associated vasculitis, highlighting the challenges in finding new, less-toxic treatments and potential therapeutic targets in this disease. The current suite of rodent models is reviewed, and future directions in the study of this complex and fascinating disease are suggested.
Collapse
Affiliation(s)
- Holly L Hutton
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Pediatric Nephrology, Monash Children's Hospital, Clayton, Victoria, Australia.
| |
Collapse
|
23
|
Salvadori M, Tsalouchos A. Antineutrophil cytoplasmic antibody associated vasculitides with renal involvement: Open challenges in the remission induction therapy. World J Nephrol 2018; 7:71-83. [PMID: 29736379 PMCID: PMC5937030 DOI: 10.5527/wjn.v7.i3.71] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/12/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023] Open
Abstract
Renal involvement with rapidly progressive glomerulonephritis is a common manifestation of antineutrophil cytoplasmic antibody (ANCA) associated vasculitides, which is characterized by end-stage renal disease and high mortality rates in untreated and/or late referral patients. The long-term renal survival has improved dramatically since the addition of cyclophosphamide (CYC) and recently of rituximab (RTX) in association with corticosteroids in the remission induction therapeutic regimens. However, renal prognosis remains unfavorable for many patients and the mortality rate is still significantly high. In this review, we analyze the open challenges to be addressed to optimize the induction remission therapy, principally in patients with advanced kidney failure. This concern the first-line therapy (CYC or RTX) based on different parameters (estimated glomerular filtration rate at baseline, new or relapsed disease, ANCA specificity, tissue injury, safety), the role of plasma exchange and the role of new therapies. Indeed, we discuss future perspectives in induction remission therapy by reporting recent advances in new targeted therapies with particular reference to avacopan, an orally administered selective C5a receptor inhibitor.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Nephrology and Renal Transplantation, Careggi University Hospital, Florence 50139, Italy
| | - Aris Tsalouchos
- Department of Nephrology and Dialysis, Saints Cosmas and Damian Hospital, Pescia 51017, Italy
| |
Collapse
|
24
|
Hofmann K, Clauder AK, Manz RA. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front Immunol 2018; 9:835. [PMID: 29740441 PMCID: PMC5924791 DOI: 10.3389/fimmu.2018.00835] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/05/2018] [Indexed: 12/29/2022] Open
Abstract
Success with B cell depletion using rituximab has proven the concept that B lineage cells represent a valid target for the treatment of autoimmune diseases, and has promoted the development of other B cell targeting agents. Present data confirm that B cell depletion is beneficial in various autoimmune disorders and also show that it can worsen the disease course in some patients. These findings suggest that B lineage cells not only produce pathogenic autoantibodies, but also significantly contribute to the regulation of inflammation. In this review, we will discuss the multiple pro- and anti-inflammatory roles of B lineage cells play in autoimmune diseases, in the context of recent findings using B lineage targeting therapies.
Collapse
Affiliation(s)
- Katharina Hofmann
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| |
Collapse
|
25
|
Schrezenmeier E, Jayne D, Dörner T. Targeting B Cells and Plasma Cells in Glomerular Diseases: Translational Perspectives. J Am Soc Nephrol 2018; 29:741-758. [PMID: 29326157 DOI: 10.1681/asn.2017040367] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The unique contributions of memory B cells and plasma cells in kidney diseases remain unclear. In this review, we evaluate the clinical experience with treatments directed at B cells, such as rituximab, and at plasma cells, such as proteasome inhibition, to shed light on the role of these two B lineage compartments in glomerular diseases. Specifically, analysis of these targeted interventions in diseases such as ANCA-associated vasculitis, SLE, and antibody-mediated transplant rejection permits insight into the pathogenetic effect of these cells. Notwithstanding the limitations of preclinical models and clinical studies (heterogeneous populations, among others), the data suggest that memory B and plasma cells represent two engines of autoimmunity, with variable involvement in these diseases. Whereas memory B cells and plasma cells appear to be key in ANCA-associated vasculitis and antibody-mediated transplant rejection, respectively, SLE seems likely to be driven by both autoimmune compartments. These conclusions have implications for the future development of targeted therapeutics in immune-mediated renal disease.
Collapse
Affiliation(s)
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Dörner
- Rheumatology and Clinical Immunology, Department of Medicine, Charité Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany; and
| |
Collapse
|
26
|
Pitarokoili K, Yoon MS, Kröger I, Reinacher-Schick A, Gold R, Schneider-Gold C. Severe refractory CIDP: a case series of 10 patients treated with bortezomib. J Neurol 2017; 264:2010-2020. [DOI: 10.1007/s00415-017-8599-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 11/29/2022]
|
27
|
Choi M, Schreiber A, Eulenberg-Gustavus C, Scheidereit C, Kamps J, Kettritz R. Endothelial NF- κB Blockade Abrogates ANCA-Induced GN. J Am Soc Nephrol 2017; 28:3191-3204. [PMID: 28687535 DOI: 10.1681/asn.2016060690] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 05/18/2017] [Indexed: 01/15/2023] Open
Abstract
ANCA-associated vasculitis (AAV) is a highly inflammatory condition in which ANCA-activated neutrophils interact with the endothelium, resulting in necrotizing vasculitis. We tested the hypothesis that endothelial NF-κB mediates necrotizing crescentic GN (NCGN) and provides a specific treatment target. Reanalysis of kidneys from previously examined murine NCGN disease models revealed NF-κB activation in affected kidneys, mostly as a p50/p65 heterodimer, and increased renal expression of NF-κB-dependent tumor necrosis factor α (TNF-α). NF-κB activation positively correlated with crescent formation, and nuclear phospho-p65 staining showed NF-κB activation within CD31-expressing endothelial cells (ECs) in affected glomeruli. Therefore, we studied the effect of ANCA on NF-κB activation in neutrophil/EC cocultures in vitro ANCA did not activate NF-κB in primed human neutrophils, but ANCA-stimulated primed neutrophils activated NF-κB in ECs, at least in part via TNF-α release. This effect increased endothelial gene transcription and protein production of NF-κB-regulated interleukin-8. Moreover, upregulation of endothelial NF-κB promoted neutrophil adhesion to EC monolayers, an effect that was inhibited by a specific IKKβ inhibitor. In a murine NCGN model, prophylactic application of E-selectin-targeted immunoliposomes packed with p65 siRNA to downregulate endothelial NF-κB significantly reduced urine abnormalities, renal myeloid cell influx, and NCGN. Increased glomerular endothelial phospho-p65 staining in patients with AAV indicated that NF-κB is activated in human NCGN also. We suggest that ANCA-stimulated neutrophils activate endothelial NF-κB, which contributes to NCGN and provides a potential therapeutic target in AAV.
Collapse
Affiliation(s)
- Mira Choi
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany; .,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany.,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany
| | | | - Jan Kamps
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ralph Kettritz
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany.,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
28
|
Abstract
In patients with membranous nephropathy, alkylating agents (cyclophosphamide or chlorambucil) alone or in combination with steroids achieve remission of nephrotic syndrome more effectively than conservative treatment or steroids alone, but can cause myelotoxicity, infections, and cancer. Calcineurin inhibitors can improve proteinuria, but are nephrotoxic. Most patients relapse after treatment withdrawal and can become treatment dependent, which increases the risk of nephrotoxicity. The discovery of nephritogenic autoantibodies against podocyte M-type phospholipase A2 receptor (PLA2R) and thrombospondin type-1 domain- containing protein 7A (THSD7A) antigens provides a clear pathophysiological rationale for interventions that specifically target B-cell lineages to prevent antibody production and subepithelial deposition. The anti-CD20 monoclonal antibody rituximab is safe and achieves remission of proteinuria in approximately two-thirds of patients with membranous nephropathy. In those with PLA2R-related disease, remission can be predicted by anti-PLA2R antibody depletion and relapse by antibody re-emergence into the circulation. Thus, integrated evaluation of serology and proteinuria could guide identification of affected patients and treatment with individually tailored protocols. Nonspecific and toxic immunosuppressive regimens will fall out of use. B-cell modulation by rituximab and second-generation anti-CD20 antibodies (or plasma cell-targeted therapy in anti-CD20 resistant forms of disease) will lead to a novel therapeutic paradigm for patients with membranous nephropathy.
Collapse
|
29
|
Abstract
The ubiquitin proteasome pathway was discovered in the 1980s to be a central component of the cellular protein-degradation machinery with essential functions in homeostasis, which include preventing the accumulation of misfolded or deleterious proteins. Cancer cells produce proteins that promote both cell survival and proliferation, and/or inhibit mechanisms of cell death. This notion set the stage for preclinical testing of proteasome inhibitors as a means to shift this fine equilibrium towards cell death. Since the late 1990s, clinical trials have been conducted for a variety of malignancies, leading to regulatory approvals of proteasome inhibitors to treat multiple myeloma and mantle-cell lymphoma. First-generation and second-generation proteasome inhibitors can elicit deep initial responses in patients with myeloma, for whom these drugs have dramatically improved outcomes, but relapses are frequent and acquired resistance to treatment eventually emerges. In addition, promising preclinical data obtained with proteasome inhibitors in models of solid tumours have not been confirmed in the clinic, indicating the importance of primary resistance. Investigation of the mechanisms of resistance is, therefore, essential to further maximize the utility of this class of drugs in the era of personalized medicine. Herein, we discuss the advances and challenges resulting from the introduction of proteasome inhibitors into the clinic.
Collapse
Affiliation(s)
- Elisabet E Manasanch
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
| |
Collapse
|
30
|
Woodle ES, Tremblay S, Driscoll J. Targeting Plasma Cells with Proteasome Inhibitors: Principles from Primates. J Am Soc Nephrol 2017; 28:1951-1953. [PMID: 28592425 DOI: 10.1681/asn.2017040443] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- E Steve Woodle
- Division of Transplantation, Department of Surgery, and .,Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Simon Tremblay
- Division of Transplantation, Department of Surgery, and.,Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Driscoll
- Division of Transplantation, Department of Surgery, and.,Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
31
|
Ludwig RJ, Vanhoorelbeke K, Leypoldt F, Kaya Z, Bieber K, McLachlan SM, Komorowski L, Luo J, Cabral-Marques O, Hammers CM, Lindstrom JM, Lamprecht P, Fischer A, Riemekasten G, Tersteeg C, Sondermann P, Rapoport B, Wandinger KP, Probst C, El Beidaq A, Schmidt E, Verkman A, Manz RA, Nimmerjahn F. Mechanisms of Autoantibody-Induced Pathology. Front Immunol 2017; 8:603. [PMID: 28620373 PMCID: PMC5449453 DOI: 10.3389/fimmu.2017.00603] [Citation(s) in RCA: 332] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
Autoantibodies are frequently observed in healthy individuals. In a minority of these individuals, they lead to manifestation of autoimmune diseases, such as rheumatoid arthritis or Graves' disease. Overall, more than 2.5% of the population is affected by autoantibody-driven autoimmune disease. Pathways leading to autoantibody-induced pathology greatly differ among different diseases, and autoantibodies directed against the same antigen, depending on the targeted epitope, can have diverse effects. To foster knowledge in autoantibody-induced pathology and to encourage development of urgently needed novel therapeutic strategies, we here categorized autoantibodies according to their effects. According to our algorithm, autoantibodies can be classified into the following categories: (1) mimic receptor stimulation, (2) blocking of neural transmission, (3) induction of altered signaling, triggering uncontrolled (4) microthrombosis, (5) cell lysis, (6) neutrophil activation, and (7) induction of inflammation. These mechanisms in relation to disease, as well as principles of autoantibody generation and detection, are reviewed herein.
Collapse
Affiliation(s)
- Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
- Department of Neurology, University of Kiel, Kiel, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Lars Komorowski
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | | | | | - Jon M. Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | - Peter Lamprecht
- Department of Rheumatology, University of Lübeck, Lübeck, Germany
| | - Andrea Fischer
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | | | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Klaus-Peter Wandinger
- Department of Neurology, Institute of Clinical Chemistry, University Medical-Centre Schleswig-Holstein, Lübeck, Germany
| | - Christian Probst
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Asmaa El Beidaq
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Alan Verkman
- Department of Medicine, University of California, San Francisco, CA, United States
- Department of Physiology, University of California, San Francisco, CA, United States
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
32
|
The proteasome - victim or culprit in autoimmunity. Clin Immunol 2016; 172:83-89. [PMID: 27475228 DOI: 10.1016/j.clim.2016.07.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
Abstract
The ubiquitin proteasome system is closely connected to apoptosis, autophagy, signaling of inflammatory cytokines and generation of ligands for MHC class I antigen presentation. Proteasome function in the innate immune response becomes particularly evident in patients with proteasome-associated autoinflammatory syndromes (PRAAS), where disease causing mutations result in reduced proteasome activity. PRAAS can be classified as a novel type of interferonopathy, however the molecular mechanism and signaling pathways leading from impaired proteasome capacity, the accumulation of damaged proteins, and the induction of type I IFN-genes remain to be determined. In contrast, several studies have confirmed an up-regulation of inducible subunits of the proteasome in systemic autoimmune diseases. Since proteasome inhibition was shown to be efficacious in several in-vitro studies and animal models of autoimmune diseases, it is justified to investigate the application of proteasome inhibitors in human disease. In this context, a number of available proteasome inhibitors has been characterized as potent immune-suppressants. The mode of action of proteasome inhibition interferes with the quality control of the huge amounts of synthetized antibodies causing an unfolded protein response. Further effects of proteasome inhibition includes inhibition of NFκB activation as well as direct activation of intrinsic and extrinsic pathways of apoptosis. The preliminary clinical work on proteasome inhibition in autoimmune diseases comprises only few studies in small cohorts with promising effects, which needs to be confirmed in controlled clinical trials.
Collapse
|
33
|
Van de Perre E, Smith RM, Bardsley V, Crawley C, Willcocks LC, Jayne DRW. Successful outcome using bortezomib in adult refractory IgA vasculitis: a case report. Rheumatology (Oxford) 2016; 55:2089-2091. [DOI: 10.1093/rheumatology/kew286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 06/27/2016] [Indexed: 11/14/2022] Open
|
34
|
Schreiber A, Eulenberg-Gustavus C, Bergmann A, Jerke U, Kettritz R. Lessons from a double-transgenic neutrophil approach to induce antiproteinase 3 antibody-mediated vasculitis in mice. J Leukoc Biol 2016; 100:1443-1452. [PMID: 27365530 DOI: 10.1189/jlb.5a0116-037r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022] Open
Abstract
ANCA to either PR3 or MPO are found in patients with necrotizing vasculitis and glomerulonephritis. ANCA binding to their target antigens on neutrophils and subsequent neutrophil activation are pivotal disease mechanisms that lead to vascular inflammation and necrosis. ANCA interaction with PR3 is more complex than with MPO as the neutrophil-specific CD177 receptor is involved in PR3 surface expression and PR3-ANCA-induced neutrophil activation. Modeling human disease is important to clinical research. Highly successful mouse models of MPO-ANCA vasculitis exist; however, recapitulating PR3-ANCA vasculitis has not been successful. We generated double-transgenic (DT) mice that expressed human PR3 and CD177 under a myeloid-specific huMRP8 promoter in an attempt to model PR3-ANCA vasculitis. DT mice strongly expressed the human transgenes in and on murine neutrophils and bound murine and human anti-PR3 antibodies. Nevertheless, passive transfer of these antibodies into LPS-primed DT mice or immunization of C57BL/6 mice with human PR3 followed by irradiation and transplantation of DT bone marrow failed to induce glomerulonephritis. Further analyses revealed that anti-PR3 antibodies did not activate DT neutrophils as shown by superoxide generation. Moreover, we found that mice did not properly process human pro-PR3 into mature PR3 and, consequently, the signaling complex between PR3, CD177, and CD11b, which promotes neutrophil activation by anti-PR3 antibodies, failed to form. We conclude that important species differences in PR3 and CD177 exist between men and mice that prevented successful generation of a murine anti-PR3 antibody model.
Collapse
Affiliation(s)
- Adrian Schreiber
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Astrid Bergmann
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; .,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
35
|
Hiepe F, Radbruch A. Plasma cells as an innovative target in autoimmune disease with renal manifestations. Nat Rev Nephrol 2016; 12:232-40. [DOI: 10.1038/nrneph.2016.20] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
36
|
Novikov P, Moiseev S, Bulanov N, Shchegoleva E. Bortezomib in refractory ANCA-associated vasculitis: a new option? Ann Rheum Dis 2016; 75:e9. [PMID: 26199397 DOI: 10.1136/annrheumdis-2015-207947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Pavel Novikov
- Clinic of Nephrology, Internal and Occupational Diseases, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergey Moiseev
- Clinic of Nephrology, Internal and Occupational Diseases, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nikolay Bulanov
- Clinic of Nephrology, Internal and Occupational Diseases, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elena Shchegoleva
- Clinic of Nephrology, Internal and Occupational Diseases, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
37
|
Souparnika S, D'Souza B, D'Souza V, Kumar S, Manjrekar P, Bairy M, Parthasarathy R, Kosuru S. Emerging Role of Myeloperoxidase in the Prognosis of Nephrotic Syndrome Patients Before and After Steroid Therapy. J Clin Diagn Res 2015; 9:BC01-4. [PMID: 26393116 DOI: 10.7860/jcdr/2015/12532.6222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/09/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND Myeloperoxidase (MPO) is a myelocyte derived iron containing enzyme particularly involved in host defense by destroying foreign micro organisms invading the body. Numerous evidences suggest that MPO is involved in the pathogenesis of many inflammatory diseases, especially atherosclerosis. AIM Present study deals with the role of MPO in the renal function and progression of disease in Nephrotic syndrome patients. STUDY DESIGN AND SETTINGS Case- Control Study carried out in Kasturba Medical College Hospital, Mangalore, India. MATERIALS AND METHODS Forty newly diagnosed Nephrotic syndrome cases, 40 age and sex matched healthy controls and 15 subjects in Nephrotic syndrome remission, were included in the study. Myeloperoxidase enzyme was assayed by 4 amino antipyrine methods in all the subjects. Other renal parameters like urea, creatinine, Blood Urea Nitrogen (BUN), BUN- Creatinine ratio (BUN/Cr) total protein, albumin, globulin, albumin - globulin ratio (A/G ratio) and estimated Glomerular Filtration Rate (eGFR) were also analysed. 24 hour urine protein-creatinine ratio was estimated in Nephrotic syndrome cases and remission group by turbidimetric assay. STATISTICAL ANALYSIS Students paired t-test and Wilcoxon Signed Rank test were used for the comparison of the data. Pearson and Spearman analyses were used for correlation of the parameters. RESULTS MPO levels were found to be high in Nephrotic syndrome cases when compared to healthy controls. Urea, creatinine, BUN, BUN/Cr ratio and eGFR were high in Nephrotic syndrome cases while total protein, albumin, globulin and A/G ratio showed decreased levels. MPO had a positive correlation with creatinine and urine protein-creatinine ratio in Nephrotic syndrome. During remission, MPO levels decreased significantly while total protein and albumin levels increased. CONCLUSION Myeloperoxidase enzyme is found to be elevated and it strongly correlated with the severity of disease in Nephrotic syndrome. Further studies can be done to use MPO as a therapeutic target in Nephrotic syndrome to ameliorate the symptoms.
Collapse
Affiliation(s)
- Sreelatha Souparnika
- Senior Research Fellow, Department of Biochemistry, Kasturba Medical College , Mangalore, Manipal University, India
| | - Benedicta D'Souza
- Professor, Department of Biochemistry, Kasturba Medical College , Mangalore, Manipal University, India
| | - Vivian D'Souza
- Professor, Department of Biochemistry, Kasturba Medical College , Mangalore, Manipal University, India
| | - Sushanth Kumar
- Associate Professor, Department of Nephrology, Kasturba Medical College , Mangalore, Manipal University, India
| | - Poornima Manjrekar
- Professor and Head, Department of Biochemistry, Kasturba Medical College , Mangalore, Manipal University, India
| | - Manohar Bairy
- Associate Professor, Department of Nephrology, Kasturba Medical College , Mangalore, Manipal University, India
| | | | - Srinivas Kosuru
- Post Graduate, Department of Nephrology, Kasturba Medical College , Mangalore, Manipal University, India
| |
Collapse
|
38
|
Smith RM. Update on the treatment of ANCA associated vasculitis. Presse Med 2015; 44:e241-9. [PMID: 26021670 DOI: 10.1016/j.lpm.2015.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 11/28/2022] Open
Abstract
The introduction of glucocorticoids and cyclophosphamide has transformed ANCA associated vasculitis (AAV) from a fatal to a largely treatable condition. Over the past 40 years, considerable progress has been made in refining immunosuppressive regimens with a focus on minimising toxicity. As knowledge of the pathogenesis of AAV grows, it is mirrored by the availability of biological agents. Lymphocyte and cytokine targeted agents have been evaluated for the treatment of AAV and are entering the routine therapeutic arena with the potential to improve patient outcomes. Rituximab has transformed management of AAV in the past decade. However, there remains unmet need in the treatment of AAV; the majority of patients will relapse within five years of diagnosis despite maintenance immunosuppression; a small number of patients remain refractory to current therapies and treatment toxicity continues to contribute to mortality and chronic disability. As in rare diseases, treatment advances in vasculitis depend on international collaborative research networks to both establish an evidence base for newer agents and develop recommendations for optimal patient management.
Collapse
Affiliation(s)
- Rona M Smith
- Addenbrooke's Hospital, Department of renal medicine, Box 57, Hills Road, Cambridge CB20QQ, United Kingdom.
| |
Collapse
|
39
|
Greenhall GHB, Salama AD. What is new in the management of rapidly progressive glomerulonephritis? Clin Kidney J 2015; 8:143-50. [PMID: 25815169 PMCID: PMC4370308 DOI: 10.1093/ckj/sfv008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/23/2015] [Indexed: 12/20/2022] Open
Abstract
Rapidly progressive glomerulonephritis (RPGN) results from severe crescentic damage to glomeruli and leads to irreversible kidney failure if not diagnosed and managed in a timely fashion. Traditional treatment has relied on glucocorticoids and cyclophosphamide, with additional plasmapheresis for certain conditions. Here we describe updates in the management of RPGN, according to the underlying renal pathology. However, there remains a paucity of trials that have enrolled patients with more advanced renal disease, dialysis dependence or with RPGN, and we are therefore still reliant on extrapolation of data from studies of patients with a less severe form of disease. In addition, reporting bias results in publication of cases or cohorts showing benefit for newer agents in advanced disease or RPGN, but it remains unclear how many unsuccessful outcomes in these circumstances take place. Since clinical trials specifically in RPGN are unlikely, use of biologic registries or combination of sufficient sized cohort series may provide indications of benefit outside of a clinical trial setting and should be encouraged, in order to provide some evidence for the efficacy of therapeutic regimens in RPGN and advanced renal disease.
Collapse
Affiliation(s)
| | - Alan D Salama
- UCL Centre for Nephrology , Royal Free Hospital , London , UK
| |
Collapse
|
40
|
Gomez AM, Willcox N, Vrolix K, Hummel J, Nogales-Gadea G, Saxena A, Duimel H, Verheyen F, Molenaar PC, Buurman WA, De Baets MH, Martinez-Martinez P, Losen M. Proteasome inhibition with bortezomib depletes plasma cells and specific autoantibody production in primary thymic cell cultures from early-onset myasthenia gravis patients. THE JOURNAL OF IMMUNOLOGY 2014; 193:1055-1063. [PMID: 24973445 DOI: 10.4049/jimmunol.1301555] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bortezomib is a potent inhibitor of proteasomes currently used to eliminate malignant plasma cells in multiple myeloma patients. It is also effective in depleting both alloreactive plasma cells in acute Ab-mediated transplant rejection and their autoreactive counterparts in animal models of lupus and myasthenia gravis (MG). In this study, we demonstrate that bortezomib at 10 nM or higher concentrations killed long-lived plasma cells in cultured thymus cells from nine early-onset MG patients and consistently halted their spontaneous production not only of autoantibodies against the acetylcholine receptor but also of total IgG. Surprisingly, lenalidomide and dexamethasone had little effect on plasma cells. After bortezomib treatment, they showed ultrastructural changes characteristic of endoplasmic reticulum stress after 8 h and were no longer detectable at 24 h. Bortezomib therefore appears promising for treating MG and possibly other Ab-mediated autoimmune or allergic disorders, especially when given in short courses at modest doses before the standard immunosuppressive drugs have taken effect.
Collapse
Affiliation(s)
- Alejandro M Gomez
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Nick Willcox
- Department of Clinical Neurology, University of Oxford, UK
| | - Kathleen Vrolix
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jonas Hummel
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Gisela Nogales-Gadea
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Neuromuscular Diseases Unit, Institut de Recerca del Hospital de la Santa Creu i Sant Pau, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Abhishek Saxena
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Hans Duimel
- ELMI Unit-CRISP, Department of Molecular Cell Biology, Maastricht University, Maastricht, the Netherlands
| | - Fons Verheyen
- ELMI Unit-CRISP, Department of Molecular Cell Biology, Maastricht University, Maastricht, the Netherlands
| | - Peter C Molenaar
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Wim A Buurman
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Marc H De Baets
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Pilar Martinez-Martinez
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mario Losen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
41
|
Tiburzy B, Kulkarni U, Hauser AE, Abram M, Manz RA. Plasma cells in immunopathology: concepts and therapeutic strategies. Semin Immunopathol 2014; 36:277-88. [PMID: 24740168 DOI: 10.1007/s00281-014-0426-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 04/01/2014] [Indexed: 12/12/2022]
Abstract
Plasma cells are terminally differentiated B cells that secrete antibodies, important for immune protection, but also contribute to any allergic and autoimmune disease. There is increasing evidence that plasma cell populations exhibit a considerable degree of heterogeneity with respect to their immunophenotype, migration behavior, lifetime, and susceptibility to immunosuppressive drugs. Pathogenic long-lived plasma cells are refractory to existing therapies. In contrast, short-lived plasma cells can be depleted by steroids and cytostatic drugs. Therefore, long-lived plasma cells are responsible for therapy-resistant autoantibodies and resemble a challenge for the therapy of antibody-mediated autoimmune diseases. Both lifetime and therapy resistance of plasma cells are supported by factors produced within their microenviromental niches. Current results suggest that plasma cell differentiation and survival factors such as IL-6 also signal via mammalian miRNAs within the plasma cell to modulate downstream transcription factors. Recent evidence also suggests that plasma cells and/or their immediate precursors (plasmablasts) can produce important cytokines and act as antigen-presenting cells, exhibiting so far underestimated roles in immune regulation and bone homeostasis. Here, we provide an overview on plasma cell biology and discuss exciting, experimental, and potential therapeutic approaches to eliminate pathogenic plasma cells.
Collapse
Affiliation(s)
- Benjamin Tiburzy
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | | | | | | | | |
Collapse
|
42
|
Beeken M, Lindenmeyer MT, Blattner SM, Radón V, Oh J, Meyer TN, Hildebrand D, Schlüter H, Reinicke AT, Knop JH, Vivekanandan-Giri A, Münster S, Sachs M, Wiech T, Pennathur S, Cohen CD, Kretzler M, Stahl RAK, Meyer-Schwesinger C. Alterations in the ubiquitin proteasome system in persistent but not reversible proteinuric diseases. J Am Soc Nephrol 2014; 25:2511-25. [PMID: 24722446 DOI: 10.1681/asn.2013050522] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Podocytes are the key cells affected in nephrotic glomerular kidney diseases, and they respond uniformly to injury with cytoskeletal rearrangement. In nephrotic diseases, such as membranous nephropathy and FSGS, persistent injury often leads to irreversible structural damage, whereas in minimal change disease, structural alterations are mostly transient. The factors leading to persistent podocyte injury are currently unknown. Proteolysis is an irreversible process and could trigger persistent podocyte injury through degradation of podocyte-specific proteins. We, therefore, analyzed the expression and functional consequence of the two most prominent proteolytic systems, the ubiquitin proteasome system (UPS) and the autophagosomal/lysosomal system, in persistent and transient podocyte injuries. We show that differential upregulation of both proteolytic systems occurs in persistent human and rodent podocyte injury. The expression of specific UPS proteins in podocytes differentiated children with minimal change disease from children with FSGS and correlated with poor clinical outcome. Degradation of the podocyte-specific protein α-actinin-4 by the UPS depended on oxidative modification in membranous nephropathy. Notably, the UPS was overwhelmed in podocytes during experimental glomerular disease, resulting in abnormal protein accumulation and compensatory upregulation of the autophagosomal/lysosomal system. Accordingly, inhibition of both proteolytic systems enhanced proteinuria in persistent nephrotic disease. This study identifies altered proteolysis as a feature of persistent podocyte injury. In the future, specific UPS proteins may serve as new biomarkers or therapeutic targets in persistent nephrotic syndrome.
Collapse
Affiliation(s)
| | - Maja T Lindenmeyer
- Institute of Physiology and Division of Nephrology, University of Zurich, Zurich, Switzerland
| | - Simone M Blattner
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan; and
| | | | | | - Tobias N Meyer
- Department of Internal Medicine, Nephrology, University Affiliated Asklepios Clinic Hamburg Barmbek, Hamburg, Germany
| | - Diana Hildebrand
- Clinical Chemistry, Mass Spectrometry and Proteome Analysis, and
| | - Hartmut Schlüter
- Clinical Chemistry, Mass Spectrometry and Proteome Analysis, and
| | | | | | - Anuradha Vivekanandan-Giri
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan; and
| | | | | | - Thorsten Wiech
- Pathology, Division of Renal Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Subramaniam Pennathur
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan; and
| | - Clemens D Cohen
- Institute of Physiology and Division of Nephrology, University of Zurich, Zurich, Switzerland
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan; and
| | | | | |
Collapse
|
43
|
Wang C, Chen S, Feng B, Guan Y. Proteasome inhibitors for malignancy-related Lambert-Eaton myasthenic syndrome. Muscle Nerve 2014; 49:325-8. [PMID: 24464710 DOI: 10.1002/mus.24122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/05/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022]
Abstract
Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune disorder characterized by autoantibodies against presynaptic voltage-gated calcium channels that impair neuromuscular transmission. Malignancies, especially small cell lung cancer (SCLC), have been associated with LEMS and account for approximately 60% of cases, making malignancy management a central step in LEMS therapy. In addition, immunosuppressive therapy is also recommended for symptomatic control. Interestingly, both pathological and epidemiological data suggest that the autoimmune response can inhibit progression of tumors in malignancy-associated LEMS. Thus, conventional broad-spectrum immunosuppressants may not be effective agents for treatment of LEMS, especially in those with malignancy-associated LEMS. Recent preclinical and clinical studies have indicated that proteasome inhibitors can eliminate antibody-producing cells efficiently, block dendritic cell maturation, and have anti-tumor activity. We hypothesize that proteasome inhibitors may be promising agents for treatment of malignancy-related LEMS.
Collapse
Affiliation(s)
- Chen Wang
- Department of Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | | | | | | |
Collapse
|
44
|
Zhou XJ, Cheng FJ, Zhu L, Lv JC, Qi YY, Hou P, Zhang H. Association of systemic lupus erythematosus susceptibility genes with IgA nephropathy in a Chinese cohort. Clin J Am Soc Nephrol 2014; 9:788-97. [PMID: 24458077 DOI: 10.2215/cjn.01860213] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES One hypothesis states that IgA nephropathy (IgAN) is a syndrome with an autoimmune component. Recent studies strongly support the notion of shared genetics between immune-related diseases. This study investigated single-nucleotide polymorphisms (SNPs) reported to be associated with systemic lupus erythematosus (SLE) in a Chinese cohort of patients with IgAN and in controls. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This study investigated whether SNP markers that had been reported to be associated with SLE were also associated with IgAN in a Chinese population. The study cohort consisted of 1194 patients with IgAN and 902 controls enrolled in Peking University First Hospital from 1997 to 2008. RESULTS Ninety-six SNPs mapping to 60 SLE loci with reported P values <1 × 10(-5) were investigated. CFH (P=8.41 × 10(-6)), HLA-DRA (P=4.91 × 10(-6)), HLA-DRB1 (P=9.46 × 10(-9)), PXK (P=3.62 × 10(-4)), BLK (P=9.32 × 10(-3)), and UBE2L3 (P=4.07 × 10(-3)) were identified as shared genes between IgAN and SLE. All associations reported herein were corroborated by associations at neighboring SNPs. Many of the alleles that are risk alleles for SLE are protective alleles for IgAN. By analyses of two open independent expression quantitative trait loci (eQTL) databases, correlations between genotypes and corresponding gene expression were observed (P<0.05 in multiple populations), suggesting a cis-eQTL effect. From gene-expression databases, differential expressions of these genes were observed in IgAN. Additive interactions between PXK rs6445961 and HLA-DRA rs9501626 (P=1.51 × 10(-2)), as well as multiplicative interactions between CFH rs6677604 and HLA-DRB1 rs9271366 (P=1.77 × 10(-2)), and between HLA-DRA rs9501626 and HLA-DRB1 rs9271366 (P=3.23 × 10(-2)) were observed. Disease risk decreased with accumulation of protective alleles. Network analyses highlighted four pathways: MHC class II antigen presentation, complement regulation, signaling by the B-cell receptor, and ubiquitin/proteasome-dependent degradation. CONCLUSION From this "systems genetics" perspective, these data provide important clues for future studies on pleiotropy in IgAN and lupus nephritis.
Collapse
Affiliation(s)
- Xu-Jie Zhou
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Rovin BH, Parikh SV. Lupus nephritis: the evolving role of novel therapeutics. Am J Kidney Dis 2014; 63:677-90. [PMID: 24411715 DOI: 10.1053/j.ajkd.2013.11.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 11/15/2013] [Indexed: 11/11/2022]
Abstract
Immune complex accumulation in the kidney is the hallmark of lupus nephritis and triggers a series of events that result in kidney inflammation and injury. Cytotoxic agents and corticosteroids are standard of care for lupus nephritis treatment, but are associated with considerable morbidity and suboptimal outcomes. Recently, there has been interest in using novel biologic agents and small molecules to treat lupus nephritis. These therapies can be broadly categorized as anti-inflammatory (laquinamod, anti-tumor necrosis factor-like weak inducer of apotosis, anti-C5, and retinoids), antiautoimmunity (anti-CD20, anti-interferon α, and costimulatory blockers), or both (anti-interleukin 6 and proteasome inhibitors). Recent lupus nephritis clinical trials applied biologics or small molecules of any category to induction treatment, seeking short-term end points of complete renal response. These trials in general have not succeeded. When lupus nephritis comes to clinical attention during the inflammatory stage of the disease, the autoimmune stage leading to kidney inflammation will have been active for some time. The optimal approach for using novel therapies may be to initially target kidney inflammation to preserve renal parenchyma, followed by suppression of autoimmunity. In this review, we discuss novel lupus nephritis therapies and how they fit into a combinatorial treatment strategy based on the pathogenic stage.
Collapse
Affiliation(s)
- Brad H Rovin
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH.
| | - Samir V Parikh
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
46
|
Jayne D. New-generation therapy for ANCA-associated vasculitis. Clin Exp Nephrol 2013; 17:694-696. [PMID: 24013763 DOI: 10.1007/s10157-013-0855-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 08/01/2013] [Indexed: 12/01/2022]
Abstract
Newer therapies for systemic vasculitis are required to improve the efficacy and reduce the toxicity associated with current agents. Our understanding of its pathogenesis is increasing with the availability of targeted therapies. B-cell depletion with rituximab is now a licensed therapy for ANCA vasculitis. Targets for therapy currently in development in ANCA associated vasculitis include blockade of complement and pro-inflammatory cytokines, and inhibition of T cell co-stimulation.
Collapse
Affiliation(s)
- David Jayne
- Department of Medicine, University of Cambridge, Cambridge, CB22QQ, UK.
| |
Collapse
|
47
|
van Roeyen CRC, Scurt FG, Brandt S, Kuhl VA, Martinkus S, Djudjaj S, Raffetseder U, Royer HD, Stefanidis I, Dunn SE, Dooley S, Weng H, Fischer T, Lindquist JA, Mertens PR. Cold shock Y-box protein-1 proteolysis autoregulates its transcriptional activities. Cell Commun Signal 2013; 11:63. [PMID: 24103640 PMCID: PMC3766096 DOI: 10.1186/1478-811x-11-63] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 08/12/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Y-box protein-1 (YB-1) fulfills pleiotropic functions relating to gene transcription, mRNA processing, and translation. It remains elusive how YB-1 shuttling into the nuclear and cytoplasmic compartments is regulated and whether limited proteolysis by the 20S proteasome releases fragments with distinct function(s) and subcellular distribution(s). RESULTS To address these questions, mapping of domains responsible for subcellular targeting was performed. Three nuclear localization signals (NLS) were identified. NLS-1 (aa 149-156) and NLS-2 (aa 185-194) correspond to residues with unknown function(s), whereas NLS-3 (aa 276-292) matches with a designated multimerization domain. Nuclear export signal(s) were not identified. Endoproteolytic processing by the 20S proteasome before glycine 220 releases a carboxy-terminal fragment (CTF), which localized to the nucleus, indicating that NLS-3 is operative. Genotoxic stress induced proteolytic cleavage and nuclear translocation of the CTF. Co-expression of the CTF and full-length YB-1 resulted in an abrogated transcriptional activation of the MMP-2 promoter, indicating an autoregulatory inhibitory loop, whereas it fulfilled similar trans-repressive effects on the collagen type I promoter. CONCLUSION Compartmentalization of YB-1 protein derivatives is controlled by distinct NLS, one of which targets a proteolytic cleavage product to the nucleus. We propose a model for an autoregulatory negative feedback loop that halts unlimited transcriptional activation.
Collapse
Affiliation(s)
- Claudia R C van Roeyen
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Leipziger Str 44, 39120 Magdeburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Grundmann F, Witthus M, Göbel H, Kisner T, Siewert R, Benzing T, Kurschat CE. Monoclonal gammopathy-associated pauci-immune extracapillary-proliferative glomerulonephritis successfully treated with bortezomib. Clin Kidney J 2013; 6:327-329. [PMID: 24282629 PMCID: PMC3840817 DOI: 10.1093/ckj/sft044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 03/26/2013] [Indexed: 12/23/2022] Open
Abstract
Extracapillary-proliferative glomerulonephritis is a rare complication of multiple myeloma. Partial remission of kidney involvement with cyclophosphamide therapy has previously been described. We report the case of a 60-year-old male patient diagnosed with rapidly progressive glomerulonephritis associated with IgG kappa monoclonal gammopathy. His kidney biopsy revealed pauci-immune extracapillary-proliferative glomerulonephritis without cryoglobulinaemia. Treatment with the proteasome inhibitor bortezomib induced rapid clinical and histological remission of his kidney disease. The patient's renal function remained stable on bortezomib maintenance therapy. Our findings suggest that bortezomib is a promising therapeutic approach to ameliorate severe kidney damage in monoclonal gammopathy- and myeloma-associated pauci-immune extracapillary-proliferative glomerulonephritis.
Collapse
Affiliation(s)
- Franziska Grundmann
- Department II of Internal Medicine , University of Cologne , Cologne , Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Schreiber A. L10. Animal models of ANCA-associated vasculitis: effector mechanisms and experimental therapies. Presse Med 2013; 42:520-3. [PMID: 23453497 DOI: 10.1016/j.lpm.2013.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Adrian Schreiber
- Department of Nephrology and Critical Care Medicine Charité Medical Faculty, Virchow Klinikum Campus and Experimental and Clinical Research Center, A joint cooperation between the Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.
| |
Collapse
|
50
|
Gomez AM, Willcox N, Molenaar PC, Buurman W, Martinez-Martinez P, De Baets MH, Losen M. Targeting plasma cells with proteasome inhibitors: possible roles in treating myasthenia gravis? Ann N Y Acad Sci 2012; 1274:48-59. [DOI: 10.1111/j.1749-6632.2012.06824.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|