1
|
Tanaka R, Murakami Y, Ellis D, Seita J, Yinga W, Kakuta S, Kumano K, Fukui R, Miyake K. TLR7 responses in glomerular macrophages accelerate the progression of glomerulonephritis in NZBWF1 mice. Int Immunol 2025; 37:339-353. [PMID: 40401698 DOI: 10.1093/intimm/dxaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/24/2025] [Indexed: 01/28/2025] Open
Abstract
Systemic lupus erythematosus is a systemic autoimmune disease characterized by the production of autoantibodies and damage to multiple organs. Glomerulonephritis, a manifestation involving glomerular deposition of immune complexes and complement components, significantly contributes to disease morbidity. Although an endosomal single-stranded RNA sensor [Toll-like receptor 7 (TLR7)] is known to drive glomerulonephritis by promoting autoantibody production in B cells, the contribution of macrophage TLR7 responses to glomerulonephritis remains poorly understood. Here, we have examined Tlr7‒/‒ NZBWF1 (New Zealand Black/New Zealand White F1) mice and found that TLR7 deficiency ameliorates lupus nephritis by abolishing autoantibody production against RNA-associated antigens, C3 deposition, and macrophage accumulation in glomeruli. Furthermore, TLR7 signaling increased CD31 expression on glomerular endothelial cells and Ly6Clow macrophages but not on T and B cells, suggesting that CD31 mediates TLR7-dependent migration of monocytes into glomeruli. Compared to their splenic counterparts, glomerular macrophages produced IL-1β in a TLR7-dependent manner. In addition, single-cell RNA sequencing of glomerular macrophages revealed that TLR7 signaling induced expression of lupus-associated genes, including those encoding Chitinase 3 like 1, ferritin heavy chain 1, IKKε, and complement factor B (CfB). Although serum CfB did not increase in NZBWF1 mice, TLR7-dependent CfB protein expression was detected in glomerular macrophages. In addition, TLR7 signaling promoted C3 cleavage and deposition predominantly on mesangial cells. These findings suggest that TLR7 responses in glomerular macrophages accelerate the progression of glomerulonephritis in NZBWF1 mice.
Collapse
Affiliation(s)
- Reika Tanaka
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yusuke Murakami
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan
| | - Dorothy Ellis
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Jun Seita
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Wu Yinga
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Shigeru Kakuta
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo 113-8657, Japan
| | - Keiki Kumano
- Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
2
|
Carter MJ, Bogdanov YD, Smith RC, Cox KL, Frampton S, Ferson L, Foxall RB, Hussain K, Strefford JC, Beers SA, Cragg MS. The ETS-family transcription factor PU.1 is a critical regulator of the inhibitory Fcγ receptor IIB expression in humans. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf109. [PMID: 40420414 DOI: 10.1093/jimmun/vkaf109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 05/28/2025]
Abstract
The inhibitory Fc gamma receptor IIB (FcγRIIB) is a critical determinant of humoral immunity. By providing feedback inhibition, through inhibitory signalling or competition for antibody Fc engagement, it counterbalances and contextualises cellular responses to signals emanating from co-ligated activating receptors, such as the B-cell receptor and activating FcγR. These activities collectively suppress the emergence of B- cell-mediated autoimmune disease and immune complex-mediated pathologies. However, FcγRIIB upregulation within the tumour microenvironment limits the efficacy of monoclonal antibody (mAb)-mediated immunotherapy of cancer. While the functional significance of FcγRIIB is well established in mice, its physiological roles and the regulatory mechanisms governing its expression remain incompletely understood in humans. Here we characterise the molecular determinants of FcγRIIB expression in human immune models and primary cells. Our findings reveal that the ETS-family transcription factor PU.1 plays a crucial role in regulating basal and inducible FcγRIIB expression. Moreover, when co-expressed, PU.1 co-operates with the related ETS-family member SPIB to drive FcγRIIB expression. PU.1 binding to the proximal FcγRIIB promoter elicits transcription, at least in part, through recruitment of the CBP/p300 transcriptional co-activators. Interestingly, similar mechanisms are also observed at the proximal promoters of the activating FcγRI and FcγRIIA, suggesting that additional, potentially lineage specific, factors cooperate with PU.1 to drive the distinct expression patterns of these FcγR. These insights pave the way for future investigations aimed at understanding the molecular mechanisms responsible for cell lineage-specific FcγR expression and subsequently manipulating them for therapeutic purposes.
Collapse
Affiliation(s)
- Matthew J Carter
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Yury D Bogdanov
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Rosanna C Smith
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Kerry L Cox
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Sarah Frampton
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Cancer Genomics Group, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Lili Ferson
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Cancer Genomics Group, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Russel B Foxall
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Khiyam Hussain
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford, United Kingdom
| | - Jonathan C Strefford
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Cancer Genomics Group, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Stephen A Beers
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Mark S Cragg
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
3
|
Thurman JM. Nearing the Finish Line: Steady Progress in the Development of Complement Inhibitors for Glomerular Disease. Kidney Int Rep 2025; 10:302-305. [PMID: 39990875 PMCID: PMC11843313 DOI: 10.1016/j.ekir.2024.11.1370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/25/2025] Open
Affiliation(s)
- Joshua M. Thurman
- Department of Medicine, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
4
|
De la Cruz A, Garcés M, Larios E, Madera-Salcedo IK, Crispín JC, Rosetti F. Immune complex deposition promotes NK cell accumulation in the kidney. PLoS One 2024; 19:e0312141. [PMID: 39570975 PMCID: PMC11581347 DOI: 10.1371/journal.pone.0312141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/01/2024] [Indexed: 11/24/2024] Open
Abstract
In systemic lupus erythematosus, immune complexes deposited in the kidney vasculature represent a potent inflammatory trigger with a high potential to progress to glomerulonephritis and organ failure. These immune complexes can be recognized by multiple effector cells via complement and Fcγ receptors. The transcriptome of CD16-bearing NK cells has been documented in kidneys from patients with SLE. In this study, we show that NK cells accumulate in the kidney in response to immune complex deposition and modulate the behavior of local T cells. Depletion of NK cells transiently ameliorated disease, suggesting NK cells may play a role in lupus nephritis and other immune complex-mediated conditions.
Collapse
Affiliation(s)
- Abigail De la Cruz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, UNAM, Mexico City, Mexico
| | - Marco Garcés
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Becario de la Dirección General de Calidad y Educación en Salud, Secretaría de Salud, México
| | - Emiliano Larios
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Becario de la Dirección General de Calidad y Educación en Salud, Secretaría de Salud, México
| | - Iris K. Madera-Salcedo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C. Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Mexico
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
5
|
Jin Y, Wang Y, Ma X, Li H, Zhang M. Identification of NET formation and the renoprotective effect of degraded NETs in lupus nephritis. Am J Physiol Renal Physiol 2024; 327:F637-F654. [PMID: 39205658 PMCID: PMC11483074 DOI: 10.1152/ajprenal.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
To explore molecular biomarkers associated with the pathophysiology and therapy of lupus nephritis (LN), we conducted a joint analysis of transcriptomic data from 40 peripheral blood mononuclear cells (PBMCs) (GSE81622) and 21 kidney samples (GSE112943) from the Gene Expression Omnibus database using bioinformatics. A total of 976 and 2,427 differentially expressed genes (DEGs) were identified in PBMCs and renal tissues. Seven and two functional modules closely related to LN were identified. Further enrichment analysis revealed that the neutrophil activation pathway was highly active in both PBMCs and the kidney. Subsequently, 16 core genes closely associated with LN were verified by protein-protein interaction screening and quantitative PCR. In vitro cell models and MRL/lpr mouse models confirmed that the abnormal expression of these core genes was closely linked to neutrophil extracellular traps (NETs) generated by neutrophil activation, while degradation of NETs led to downregulation of core gene expression, thereby improving pathological symptoms of LN. Therefore, identification of patients with systemic lupus erythematosus exhibiting abnormal expression patterns for these core genes may serve as a useful indicator for kidney involvement. In addition, targeting neutrophils to modulate their activation levels and inhibit aberrant expression of these genes represents a potential therapeutic strategy for treating LN. NEW & NOTEWORTHY The mechanisms by which immune cells cause kidney injury in lupus nephritis are poorly understood. We integrated and analyzed the transcriptomic features of PBMCs and renal tissues from the GEO database to identify key molecular markers associated with neutrophil activation. We confirmed that neutrophil extracellular traps (NETs) formed by neutrophil activation promoted the upregulation of key genes in cell and animal models. Targeted degradation of NETs significantly ameliorated kidney injury in MRL/lpr mice.
Collapse
Affiliation(s)
- Yong Jin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yutong Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xu Ma
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hongbin Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Manling Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
6
|
Aringer M, Finzel S, Voll RE. [Immunopathogenesis of systemic lupus erythematosus]. Z Rheumatol 2024; 83:68-76. [PMID: 35551439 PMCID: PMC10847069 DOI: 10.1007/s00393-022-01214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 11/28/2022]
Abstract
Insights into the immunopathogenesis of systemic lupus erythematosus (SLE) help to understand the complex disease patterns and to develop new treatment strategies. The disease manifestations essentially result from autoantibodies, immune complexes and cytokines. Particularly the propensity towards developing various autoantibodies is central to the disease itself; autoantibody specificities lead to highly variable organ manifestations. This review article delineates the clinically relevant state of knowledge on SLE pathogenesis, with the goal to establish a model useful for clinical practice, which also helps to classify the novel therapeutic approaches.
Collapse
Affiliation(s)
- Martin Aringer
- Rheumatologie, Medizinische Klinik III und UniversitätsCentrum für Autoimmun- und Rheumatische Erkrankungen (UCARE), Universitätsklinikum und Medizinische Fakultät Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | - Stephanie Finzel
- Klinik für Rheumatologie und Klinische Immunologie & Centrum für chronische Immundefizienz, Universitätsklinikum Freiburg, Freiburg, Deutschland
| | - Reinhard E Voll
- Klinik für Rheumatologie und Klinische Immunologie & Centrum für chronische Immundefizienz, Universitätsklinikum Freiburg, Freiburg, Deutschland
| |
Collapse
|
7
|
Tabrizi S, Martin-Alonso C, Xiong K, Blewett T, Sridhar S, An Z, Patel S, Rodriguez-Aponte S, Naranjo CA, Wang ST, Shea D, Golub TR, Bhatia SN, Adalsteinsson V, Love JC. An intravenous DNA-binding priming agent protects cell-free DNA and improves the sensitivity of liquid biopsies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523947. [PMID: 36711455 PMCID: PMC9882106 DOI: 10.1101/2023.01.13.523947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Blood-based, or "liquid," biopsies enable minimally invasive diagnostics but have limits on sensitivity due to scarce cell-free DNA (cfDNA). Improvements to sensitivity have primarily relied on enhancing sequencing technology ex vivo . Here, we sought to augment the level of circulating tumor DNA (ctDNA) detected in a blood draw by attenuating the clearance of cfDNA in vivo . We report a first-in-class intravenous DNA-binding priming agent given 2 hours prior to a blood draw to recover more cfDNA. The DNA-binding antibody minimizes nuclease digestion and organ uptake of cfDNA, decreasing its clearance at 1 hour by over 150-fold. To improve plasma persistence and limit potential immune interactions, we abrogated its Fc-effector function. We found that it protects GC-rich sequences and DNase-hypersensitive sites, which are ordinarily underrepresented in cfDNA. In tumor-bearing mice, priming improved tumor DNA recovery by 19-fold and sensitivity for detecting cancer from 6% to 84%. These results suggest a novel method to enhance the sensitivity of existing DNA-based cancer testing using blood biopsies.
Collapse
|
8
|
Harsini S, Rezaei N. Autoimmune diseases. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
9
|
Kronbichler A, Bajema I, Geetha D, Säemann M. Novel aspects in the pathophysiology and diagnosis of glomerular diseases. Ann Rheum Dis 2022; 82:585-593. [PMID: 36535746 DOI: 10.1136/ard-2022-222495] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Immune deposits/complexes are detected in a multitude of tissues in autoimmune disorders, but no organ has attracted as much attention as the kidney. Several kidney diseases are characterised by the presence of specific configurations of such deposits, and many of them are under a 'shared care' between rheumatologists and nephrologists. This review focuses on five different diseases commonly encountered in rheumatological and nephrological practice, namely IgA vasculitis, lupus nephritis, cryoglobulinaemia, anti-glomerular basement membrane disease and anti-neutrophil cytoplasm-antibody glomerulonephritis. They differ in disease aetiopathogenesis, but also the potential speed of kidney function decline, the responsiveness to immunosuppression/immunomodulation and the deposition of immune deposits/complexes. To date, it remains unclear if deposits are causing a specific disease or aim to abrogate inflammatory cascades responsible for tissue damage, such as neutrophil extracellular traps or the complement system. In principle, immunosuppressive therapies have not been developed to tackle immune deposits/complexes, and repeated kidney biopsy studies found persistence of deposits despite reduction of active inflammation, again highlighting the uncertainty about their involvement in tissue damage. In these studies, a progression of active lesions to chronic changes such as glomerulosclerosis was frequently reported. Novel therapeutic approaches aim to mitigate these changes more efficiently and rapidly. Several new agents, such as avacopan, an oral C5aR1 inhibitor, or imlifidase, that dissolves IgG within minutes, are more specifically reducing inflammatory cascades in the kidney and repeat tissue sampling might help to understand their impact on immune cell deposition and finally kidney function recovery and potential impact of immune complexes/deposits.
Collapse
Affiliation(s)
- Andreas Kronbichler
- Department of Medicine, University of Cambridge, Cambridge, UK .,Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, UK
| | - Ingeborg Bajema
- Department of Pathology, Leiden University Medical Center, Leiden and Department of Pathology and Medical Biology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Duvuru Geetha
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcus Säemann
- 6th Medical Department, Nephrology and Dialysis, Clinic Ottakring, Vienna, Austria.,Medical Faculty, Sigmund Freud University, Vienna, Austria
| |
Collapse
|
10
|
David BL, Ivan GNJ, Emilio PGE, Daniela MSJ, Betsabe CH, Luisa VVM, Selene FRN, Guadalupe ACEA, Miriam SCA, Alfredo C, Arturo NVC, Mercedes HCD, German CME, Gonzalez-Lopez L. Low serum uromodulin levels and their association with lupus flares. PLoS One 2022; 17:e0276481. [PMID: 36301848 PMCID: PMC9612514 DOI: 10.1371/journal.pone.0276481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/07/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Only two previous studies in systemic lupus erythematosus (SLE) patients have identified that the blood concentrations of uromodulin are lower in nephritis. However, none of them had evaluated whether a low serum uromodulin adjusted by the glomerular filtration rate (sUromod/eGFR index) contributed to identify patients in risk of lupus nephritis (LN) using multivariable models. AIM Therefore, this study aimed two objectives to evaluate the association between low serum uromodulin levels and low sUromod adjusted by eGFR with renal flares in SLE excluding effects of potential confounders in multivariable analyses; and to identify the value of low sUmod and low sUmod/eGFR index as a potential diagnostic marker of LN. PATIENTS AND METHODS Design: Cross-sectional study. SLE patients (n = 114) were investigated for lupus flare with renal SLEDAI. Two groups: a) SLE with renal flare (renal-SLEDAI≥4, n = 41) and b) SLE non-renal flare (renal SLEDAI<4, n = 73). SLE patients were evaluated by other indices including a global disease activity index (SLEDAI) and SLICC renal disease activity score. Serum uromodulin levels (ng/mL) were quantified by ELISA. Serum uromodulin was adjusted by eGFR (sUromod/eGFR index). Cutt-offs of low sUromodulin and low sUromod/eGFR index were computed, ROC curves were performed and values of diagnostic tests were obtained. Multivariable logistic regression models were performed to identify if low sUromod/eGFR index is associated to renal flares. RESULTS Low serum uromodulin and low sUromod/eGFR index correlated to high scores of renal-SLEDAI, SLICC-renal and proteinuria. SLE patients with a renal flare had lower uromodulin levels compared to SLE patients without renal flare (p = 0.004). After adjusting by potential confounders, the low sUromod/eGFR index (<0.80 ng/mL) increased the risk of a renal flare (OR, 2.91; 95%CI, 1.21 to 6.98; p = 0.02). CONCLUSIONS We propose the low sUromod/eGFR index as a potential new marker of renal disease activity in SLE.
Collapse
Affiliation(s)
- Bonilla-Lara David
- Programa de Doctorado en Farmacología, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Gamez-Nava Jorge Ivan
- Programa de Doctorado en Farmacología, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Programa de Doctorado en Salud Publica, Departamento de Salud Pública, Centro Universitario de Ciencias de la Salud, Departamento de Salud Pública, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Research Group of Factors Related to Therapeutic Outcomes in Autoimmune Diseases, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Perez-Guerrero Edsaul Emilio
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Murillo-Saich Jessica Daniela
- Department of Medicine, School of Medicine, University of California, San Diego, California, United States of America
| | - Contreras-Haro Betsabe
- División de Ciencias de la Salud, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá, Jalisco, México
| | - Vazquez-Villegas Maria Luisa
- Programa de Doctorado en Salud Publica, Departamento de Salud Pública, Centro Universitario de Ciencias de la Salud, Departamento de Salud Pública, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Research Group of Factors Related to Therapeutic Outcomes in Autoimmune Diseases, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Epidemiología, Unidad de Medicina Familiar N°, 4, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Fajardo-Robledo Nicte Selene
- Research Group of Factors Related to Therapeutic Outcomes in Autoimmune Diseases, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Laboratorio de Investigación y Desarrollo Farmacéutico, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Aguilar-Chavez Erika Anita Guadalupe
- Department of Medicine, School of Medicine, University of California, San Diego, California, United States of America
- Unidad de Medicina Familiar No. 2, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Saldaña-Cruz Ana Miriam
- Research Group of Factors Related to Therapeutic Outcomes in Autoimmune Diseases, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Celis Alfredo
- Programa de Doctorado en Salud Publica, Departamento de Salud Pública, Centro Universitario de Ciencias de la Salud, Departamento de Salud Pública, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Nava-Valdivia Cesar Arturo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | | | - Cardona-Muñoz Ernesto German
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Laura Gonzalez-Lopez
- Programa de Doctorado en Farmacología, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Research Group of Factors Related to Therapeutic Outcomes in Autoimmune Diseases, Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
- Departamento de Medicina Interna-Reumatología, Hospital General Regional 110 Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
- * E-mail: ,
| |
Collapse
|
11
|
Bi Y, Su J, Zhou S, Zhao Y, Zhang Y, Zhang H, Liu M, Zhou A, Xu J, Pan M, Zhao Y, Li F. Distinct impact of IgG subclass on autoantibody pathogenicity in different IgG4-mediated diseases. eLife 2022; 11:76223. [PMID: 35920621 PMCID: PMC9385207 DOI: 10.7554/elife.76223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
IgG4 is the least potent human IgG subclass for the FcγR-mediated antibody effector function. Paradoxically, IgG4 is also the dominant IgG subclass of pathogenic autoantibodies in IgG4-mediated diseases. Here, we show that the IgG subclass and Fc-FcγR interaction have a distinct impact on the pathogenic function of autoantibodies in different IgG4-mediated diseases in mouse models. While IgG4 and its weak Fc-FcγR interaction have an ameliorative role in the pathogenicity of anti-ADAMTS13 autoantibodies isolated from thrombotic thrombocytopenic purpura (TTP) patients, they have an unexpected exacerbating effect on anti-Dsg1 autoantibody pathogenicity in pemphigus foliaceus (PF) models. Strikingly, a non-pathogenic anti-Dsg1 antibody variant optimized for FcγR-mediated effector function can attenuate the skin lesions induced by pathogenic anti-Dsg1 antibodies by promoting the clearance of dead keratinocytes. These studies suggest that IgG effector function contributes to the clearance of autoantibody-Ag complexes, which is harmful in TTP, but beneficial in PF and may provide new therapeutic opportunity.
Collapse
Affiliation(s)
- Yanxia Bi
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Su
- Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengru Zhou
- Department of Dermatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yingjie Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Huihui Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Mingdong Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Aiwu Zhou
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jianrong Xu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Pan
- Department of Dermatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yiming Zhao
- Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fubin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Miyake K, Shibata T, Fukui R, Sato R, Saitoh SI, Murakami Y. Nucleic Acid Sensing by Toll-Like Receptors in the Endosomal Compartment. Front Immunol 2022; 13:941931. [PMID: 35812450 PMCID: PMC9259784 DOI: 10.3389/fimmu.2022.941931] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptors (TLRs) respond to pathogen constituents, such as microbial lipids and nucleic acids (NAs). TLRs recognize NAs in endosomal compartments. Structural and functional studies have shown that recognition of NAs by TLRs depends on NA processing by RNases and DNases. DNase II-dependent DNA degradation is required for TLR9 responses to single-stranded DNAs, whereas RNase T2-dependent RNA degradation enables TLR7 and TLR8 to respond to nucleosides and oligoribonucleotides. In contrast, RNases and DNases negatively regulate TLR responses by degrading their ligands. RNase T2 negatively regulates TLR3 responses to degrading the TLR3 ligand double-stranded RNAs. Therefore, NA metabolism in the endosomal compartments affects the endosomal TLR responses. Dysregulation of NA metabolism in the endosomal compartment drives the TLR-dependent pathologies in human diseases.
Collapse
Affiliation(s)
- Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
- *Correspondence: Kensuke Miyake,
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| |
Collapse
|
13
|
Tout I, Miossec P. The role of B cells and their interactions with stromal cells in the context of inflammatory autoimmune diseases. Clin Exp Rheumatol 2022; 21:103098. [PMID: 35417796 DOI: 10.1016/j.autrev.2022.103098] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/08/2022] [Indexed: 02/07/2023]
Abstract
Interactions between B cells and stromal cells have essential functions in immune cell development and responses. During chronic inflammation, the pro-inflammatory microenvironment leads to changes in stromal cells, which acquire a pathogenic phenotype specific to each organ and disease. B cells are recruited to the site of inflammation and interact with these pathogenic stromal cells contributing to the disease's severity. In addition to producing autoantibodies, B cells contribute to the pathogenesis of autoimmune inflammatory diseases by serving as professional antigen-presenting cells, producing cytokines, and through additional mechanisms. This review describes the role of B cells and their interactions with stromal cells in chronic inflammation, with a focus on human disease, using three selected autoimmune inflammatory diseases: rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis. Understanding B cells roles and their interaction with stromal cells will help develop new therapeutic options for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Issam Tout
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit, University of Lyon, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437 Lyon, France
| | - Pierre Miossec
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit, University of Lyon, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437 Lyon, France.
| |
Collapse
|
14
|
Vivarelli M, van de Kar N, Labbadia R, Diomedi-Camassei F, Thurman JM. A clinical approach to children with C3 glomerulopathy. Pediatr Nephrol 2022; 37:521-535. [PMID: 34002292 DOI: 10.1007/s00467-021-05088-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/28/2021] [Accepted: 04/20/2021] [Indexed: 11/28/2022]
Abstract
C3 glomerulopathy is a relatively new clinical entity that represents a challenge both to diagnose and to treat. As new therapeutic agents that act as complement inhibitors become available, many with an oral formulation, a better understanding of this disease and of the underlying complement dysregulation driving it has become increasingly useful to optimize patient care. Moreover, recent advances in research have clarified the role of complement in other glomerular diseases in which its role was less established, namely in immune-complex membranoproliferative glomerulonephritis (IC-MPGN), ANCA-vasculitis, IgA nephropathy, and idiopathic membranous nephropathy. Complement inhibitors are being studied in adult and adolescent clinical trials for these indications. This review summarizes current knowledge and future perspectives on every aspect of the diagnosis and management of C3 glomerulopathy and elucidates current understanding of the role of complement in this condition and in other glomerular diseases in children. An overview of ongoing trials involving therapeutic agents targeting complement in glomerular diseases is also provided.
Collapse
Affiliation(s)
- Marina Vivarelli
- Division of Nephrology and Dialysis, Department of Pediatric Subspecialties, Bambino Gesù Pediatric Hospital IRCCS, Piazza S Onofrio 4, 00165, Rome, Italy.
| | - Nicole van de Kar
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Raffaella Labbadia
- Division of Nephrology and Dialysis, Department of Pediatric Subspecialties, Bambino Gesù Pediatric Hospital IRCCS, Piazza S Onofrio 4, 00165, Rome, Italy
| | | | - Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
15
|
Venkatadri R, Sabapathy V, Dogan M, Sharma R. Targeting Regulatory T Cells for Therapy of Lupus Nephritis. Front Pharmacol 2022; 12:806612. [PMID: 35069220 PMCID: PMC8775001 DOI: 10.3389/fphar.2021.806612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Lupus glomerulonephritis (LN) is a complex autoimmune disease characterized by circulating autoantibodies, immune-complex deposition, immune dysregulation and defects in regulatory T cell (Tregs). Treatment options rely on general immunosuppressants and steroids that have serious side effects. Approaches to target immune cells, such as B cells in particular, has had limited success and new approaches are being investigated. Defects in Tregs in the setting of autoimmunity is well known and Treg-replacement strategies are currently being explored. The aim of this minireview is to rekindle interest on Treg-targeting strategies. We discuss the existing evidences for Treg-enhancement strategies using key cytokines interleukin (IL)-2, IL-33 and IL-6 that have shown to provide remission in LN. We also discuss strategies for indirect Treg-modulation for protection from LN.
Collapse
Affiliation(s)
- Rajkumar Venkatadri
- Center for Immunity, Inflammation and Regenerative Medicine (CIIR), Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Vikram Sabapathy
- Center for Immunity, Inflammation and Regenerative Medicine (CIIR), Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Murat Dogan
- Center for Immunity, Inflammation and Regenerative Medicine (CIIR), Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Rahul Sharma
- Center for Immunity, Inflammation and Regenerative Medicine (CIIR), Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
16
|
Koga T, Ichinose K, Tsokos GC. Tissue resident cell processes determine organ damage in systemic lupus erythematosus. Clin Immunol 2022; 234:108919. [PMID: 34974170 DOI: 10.1016/j.clim.2021.108919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects almost any organ. Multiple immunological abnormalities involving every domain of the immune system contribute to the expression of the disease. It is now recognized that elements of the immune system instigate processes in tissue resident cells which execute organ damage. Although correction of ongoing immune aberrations is important in the control of disease activity, targeting tissue specific injurious processes may prove desirable in limiting organ damage.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Güngörer V, Ertan K, Arslan Ş. Diagnosis that clinicians don't think of, serum sickness-like reaction. Pediatr Int 2022; 64:e14740. [PMID: 34676635 DOI: 10.1111/ped.14740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Vildan Güngörer
- Departments of Pediatric Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Kübra Ertan
- Departments of Pediatrics, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Şükrü Arslan
- Departments of Pediatric Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| |
Collapse
|
18
|
Murakami Y, Fukui R, Tanaka R, Motoi Y, Kanno A, Sato R, Yamaguchi K, Amano H, Furukawa Y, Suzuki H, Suzuki Y, Tamura N, Yamashita N, Miyake K. Anti-TLR7 Antibody Protects Against Lupus Nephritis in NZBWF1 Mice by Targeting B Cells and Patrolling Monocytes. Front Immunol 2021; 12:777197. [PMID: 34868046 PMCID: PMC8632649 DOI: 10.3389/fimmu.2021.777197] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 01/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and multiple organ damage. Toll-like receptor 7 (TLR7), an innate immune RNA sensor expressed in monocytes/macrophages, dendritic cells (DCs), and B cells, promotes disease progression. However, little is known about the cellular mechanisms through which TLR7 drives lupus nephritis. Here, we show that the anti-mouse TLR7 mAb, but not anti-TLR9 mAb, protected lupus-prone NZBWF1 mice from nephritis. The anti-TLR7 mAb reduced IgG deposition in glomeruli by inhibiting the production of autoantibodies to the RNA-associated antigens. We found a disease-associated increase in Ly6Clow patrolling monocytes that expressed high levels of TLR7 and had upregulated expression of lupus-associated IL-10, CD115, CD31, and TNFSF15 in NZBWF1 mice. Anti-TLR7 mAb abolished this lupus-associated increase in patrolling monocytes in the circulation, spleen, and glomeruli. These results suggested that TLR7 drives autoantibody production and lupus-associated monocytosis in NZBWF1 mice and, that anti-TLR7 mAb is a promising therapeutic tool targeting B cells and monocytes/macrophages.
Collapse
Affiliation(s)
- Yusuke Murakami
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Reika Tanaka
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuji Motoi
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuo Kanno
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Amano
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Naomi Yamashita
- Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Innate Immunity, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Toll-like Receptor Signaling Inhibitory Peptide Improves Inflammation in Animal Model and Human Systemic Lupus Erythematosus. Int J Mol Sci 2021; 22:ijms222312764. [PMID: 34884569 PMCID: PMC8657918 DOI: 10.3390/ijms222312764] [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: 10/28/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
Toll-like receptors (TLRs) play a major role in the innate immune system. Several studies have shown the regulatory effects of TLR-mediated pathways on immune and inflammatory diseases. Dysregulated functions of TLRs within the endosomal compartment, including TLR7/9 trafficking, may cause systemic lupus erythematosus (SLE). TLR signaling pathways are fine-tuned by Toll/interleukin-1 receptor (TIR) domain-containing adapters, leading to interferon (IFN)-α production. This study describes a TLR inhibitor peptide 1 (TIP1) that primarily suppresses the downstream signaling mediated by TIR domain-containing adapters in an animal model of lupus and patients with SLE. The expression of most downstream proteins of the TLR7/9/myeloid differentiation factor 88 (MyD88)/IFN regulatory factor 7 signaling was downregulated in major tissues such as the kidney, spleen, and lymph nodes of treated mice. Furthermore, the pathological analysis of the kidney tissue confirmed that TIP1 could improve inflammation in MRL/lpr mice. TIP1 treatment downregulated many downstream proteins associated with TLR signaling, such as MyD88, interleukin-1 receptor-associated kinase, tumor necrosis factor receptor-associated factor 6, and IFN-α, in the peripheral blood mononuclear cells of patients with SLE. In conclusion, our data suggest that TIP1 can serve as a potential candidate for the treatment of SLE.
Collapse
|
20
|
Zheng C, Sleiman MM, Yang X, He S, Atkinson C, Tomlinson S. Increasing the efficacy and safety of a human complement inhibitor for treating post-transplant cardiac ischemia reperfusion injury by targeting to a graft-specific neoepitope. J Heart Lung Transplant 2021; 40:1112-1121. [PMID: 34334299 PMCID: PMC10587835 DOI: 10.1016/j.healun.2021.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 07/03/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Post-transplant ischemia reperfusion injury (IRI) is a recognized risk factor for subsequent organ dysfunction, alloresponsiveness, and rejection. The complement system is known to play a role in IRI and represents a therapeutic target. Complement is activated in transplanted grafts when circulating IgM antibodies bind to exposed ischemia-induced neoepitopes upon reperfusion, and we investigated the targeting of a human complement inhibitor, CR1, to a post-transplant ischemia-induced neoepitope. METHODS A fragment of human CR1 was linked to a single chain antibody construct (C2 scFv) recognizing an injury-specific neoepitope to yield C2-CR1. This construct, along with a soluble untargeted counterpart, was characterized in a cardiac allograft transplantation model of IRI in terms of efficacy and safety. RESULTS CR1 was similarly effective against mouse and human complement. C2-CR1 provided effective protection against cardiac IRI at a lower dose than untargeted CR1. The increased efficacy of C2-CR1 relative to CR1 correlated with decreased C3 deposition, and C2-CR1, but not CR1, targeted to cardiac allografts. At a dose necessary to reduce IRI, C2-CR1 had minimal impact on serum complement activity, in contrast to CR1 which resulted in a high level of systemic inhibition. The circulatory half-life of CR1 was markedly longer than that of C2-CR1, and whereas a minimum therapeutic dose of CR1 severely impaired host susceptibility to infection, C2-CR1 had no impact. CONCLUSION We show the translational potential of a human complement inhibitor targeted to a universal ischemia-induced graft-specific epitope, and demonstrate advantages compared to an untargeted counterpart in terms of efficacy and safety.
Collapse
Affiliation(s)
- Chaowen Zheng
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Mohamad Mahdi Sleiman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaofeng Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Songqing He
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- The Lee Patterson Allen Transplant Immunobiology Laboratory, Department of Transplant Surgery, Medical University of South Carolina, Department of Surgery, Charleston, SC, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Department of Pulmonary Medicine, University of Florida, Gainesville, FL, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
21
|
Stein MC, Braun F, Krebs CF, Bunders MJ. Kidney organoid systems for studies of immune-mediated kidney diseases: challenges and opportunities. Cell Tissue Res 2021; 385:457-473. [PMID: 34309728 PMCID: PMC8310776 DOI: 10.1007/s00441-021-03499-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022]
Abstract
Acute and chronic kidney diseases are major contributors to morbidity and mortality in the global population. Many nephropathies are considered to be immune-mediated with dysregulated immune responses playing an important role in the pathogenesis. At present, targeted approaches for many kidney diseases are still lacking, as the underlying mechanisms remain insufficiently understood. With the recent development of organoids—a three-dimensional, multicellular culture system, which recapitulates important aspects of human tissues—new opportunities to investigate interactions between renal cells and immune cells in the pathogenesis of kidney diseases arise. To date, kidney organoid systems, which reflect the structure and closer resemble critical aspects of the organ, have been established. Here, we highlight the recent advances in the development of kidney organoid models, including pluripotent stem cell-derived kidney organoids and primary epithelial cell-based tubuloids. The employment and further required advances of current organoid models are discussed to investigate the role of the immune system in renal tissue development, regeneration, and inflammation to identify targets for the development of novel therapeutic approaches of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Melissa C Stein
- Research Department Virus Immunology, Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madeleine J Bunders
- Research Department Virus Immunology, Leibniz-Institute for Experimental Virology, Hamburg, Germany.
| |
Collapse
|
22
|
Gile JJ, Sara JDS, Mueller MR. Systemic lupus erythematosus multiorgan flare with quiescent serologic markers. BMJ Case Rep 2021; 14:14/3/e239048. [PMID: 33653839 PMCID: PMC7929838 DOI: 10.1136/bcr-2020-239048] [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] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) can affect almost every organ with differing degrees of severity. Typically, SLE activity is associated with hypocomplimentaemia and elevated double-stranded DNA (dsDNA) levels. We describe a case of a severe multiorgan lupus flare including lupus cerebritis, autoimmune haemolytic anaemia, lupus nephritis and lupus myopericarditis with normal complement and dsDNA levels. This highlights the importance of understanding the heterogeneous nature of SLE flares.
Collapse
|
23
|
Kucuksezer UC, Aktas Cetin E, Esen F, Tahrali I, Akdeniz N, Gelmez MY, Deniz G. The Role of Natural Killer Cells in Autoimmune Diseases. Front Immunol 2021; 12:622306. [PMID: 33717125 PMCID: PMC7947192 DOI: 10.3389/fimmu.2021.622306] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells, the large granular lymphocytes differentiated from the common lymphoid progenitors, were discovered in early 1970's. They are members of innate immunity and were initially defined by their strong cytotoxicity against virus-infected cells and by their important effector functions in anti-tumoral immune responses. Nowadays, NK cells are classified among the recently discovered innate lymphoid cell subsets and have capacity to influence both innate and adaptive immune responses. Therefore, they can be considered as innate immune cells that stands between the innate and adaptive arms of immunity. NK cells don't express T or B cell receptors and are recognized by absence of CD3. There are two major subgroups of NK cells according to their differential expression of CD16 and CD56. While CD16+CD56dim subset is best-known by their cytotoxic functions, CD16-CD56bright NK cell subset produces a bunch of cytokines comparable to CD4+ T helper cell subsets. Another subset of NK cells with production of interleukin (IL)-10 was named as NK regulatory cells, which has suppressive properties and could take part in immune-regulatory responses. Activation of NK cells is determined by a delicate balance of cell-surface receptors that have either activating or inhibitory properties. On the other hand, a variety of cytokines including IL-2, IL-12, IL-15, and IL-18 influence NK cell activity. NK-derived cytokines and their cytotoxic functions through induction of apoptosis take part in regulation of the immune responses and could contribute to the pathogenesis of many immune mediated diseases including ankylosing spondylitis, Behçet's disease, multiple sclerosis, rheumatoid arthritis, psoriasis, systemic lupus erythematosus and type-1 diabetes. Dysregulation of NK cells in autoimmune disorders may occur through multiple mechanisms. Thanks to the rapid developments in biotechnology, progressive research in immunology enables better characterization of cells and their delicate roles in the complex network of immunity. As NK cells stand in between innate and adaptive arms of immunity and "bridge" them, their contribution in inflammation and immune regulation deserves intense investigations. Better understanding of NK-cell biology and their contribution in both exacerbation and regulation of inflammatory disorders is a requisite for possible utilization of these multi-faceted cells in novel therapeutic interventions.
Collapse
Affiliation(s)
- Umut Can Kucuksezer
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Aktas Cetin
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Fehim Esen
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
- Department of Ophthalmology, Medical Faculty, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ilhan Tahrali
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Nilgun Akdeniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Metin Yusuf Gelmez
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
24
|
Zhang Y, Liu D, Liu Z. Fine Particulate Matter (PM 2.5) and Chronic Kidney Disease. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 254:183-215. [PMID: 34529145 DOI: 10.1007/398_2020_62] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The impact of ambient particulate matter (PM) on public health has become a great global concern, which is especially prominent in developing countries. For health purposes, PM is typically defined by size, with the smaller particles having more health impacts. Particles with a diameter <2.5 μm are called PM2.5. Initial research studies have focused on the impact of PM2.5 on respiratory and cardiovascular diseases; nevertheless, an increasing number of data suggested that PM2.5 may affect every organ system in the human body, and the kidney is of no exception. The kidney is vulnerable to particulate matter because most environmental toxins are concentrated by the kidney during filtration. According to the high morbidity and mortality related to chronic kidney disease, it is necessary to determine the effect of PM2.5 on kidney disease and its mechanism that needs to be identified. To understand the current status of PM2.5 in the atmosphere and their potential harmful kidney effects in different regions of the world this review article was prepared based on peer-reviewed scientific papers, scientific reports, and database from government organizations published after the year 1998. In this review, we focus on the worldwide epidemiological evidence linking PM2.5 with chronic kidney disease and the effect of PM2.5 on the chronic kidney disease (CKD) progression. At the same time, we also discuss the possible mechanisms of PM2.5 exposure leading to kidney damage, in order to emphasize the contribution of PM2.5 to kidney damage. A global database on PM2.5 and kidney disease should be developed to provide new ideas for the prevention and treatment of kidney disease.
Collapse
Affiliation(s)
- Yilin Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P. R. China
- Research Center for Kidney Disease, Zhengzhou, Henan Province, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P. R. China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, P. R. China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P. R. China.
- Research Center for Kidney Disease, Zhengzhou, Henan Province, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P. R. China.
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, P. R. China.
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P. R. China.
- Research Center for Kidney Disease, Zhengzhou, Henan Province, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P. R. China.
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, P. R. China.
| |
Collapse
|
25
|
Melki I, Allaeys I, Tessandier N, Mailhot B, Cloutier N, Campbell RA, Rowley JW, Salem D, Zufferey A, Laroche A, Lévesque T, Patey N, Rauch J, Lood C, Droit A, McKenzie SE, Machlus KR, Rondina MT, Lacroix S, Fortin PR, Boilard E. FcγRIIA expression accelerates nephritis and increases platelet activation in systemic lupus erythematosus. Blood 2020; 136:2933-2945. [PMID: 33331924 PMCID: PMC7751357 DOI: 10.1182/blood.2020004974] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune inflammatory disease characterized by deposits of immune complexes (ICs) in organs and tissues. The expression of FcγRIIA by human platelets, which is their unique receptor for immunoglobulin G antibodies, positions them to ideally respond to circulating ICs. Whereas chronic platelet activation and thrombosis are well-recognized features of human SLE, the exact mechanisms underlying platelet activation in SLE remain unknown. Here, we evaluated the involvement of FcγRIIA in the course of SLE and platelet activation. In patients with SLE, levels of ICs are associated with platelet activation. Because FcγRIIA is absent in mice, and murine platelets do not respond to ICs in any existing mouse model of SLE, we introduced the FcγRIIA (FCGR2A) transgene into the NZB/NZWF1 mouse model of SLE. In mice, FcγRIIA expression by bone marrow cells severely aggravated lupus nephritis and accelerated death. Lupus onset initiated major changes to the platelet transcriptome, both in FcγRIIA-expressing and nonexpressing mice, but enrichment for type I interferon response gene changes was specifically observed in the FcγRIIA mice. Moreover, circulating platelets were degranulated and were found to interact with neutrophils in FcγRIIA-expressing lupus mice. FcγRIIA expression in lupus mice also led to thrombosis in lungs and kidneys. The model recapitulates hallmarks of human SLE and can be used to identify contributions of different cellular lineages in the manifestations of SLE. The study further reveals a role for FcγRIIA in nephritis and in platelet activation in SLE.
Collapse
Affiliation(s)
- Imene Melki
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Allaeys
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Nicolas Tessandier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Benoit Mailhot
- Département de Médecine Moléculaire, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
- Axe Neurosciences, Université Laval, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Nathalie Cloutier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Robert A Campbell
- Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT
- University of Utah Molecular Medicine Program, Eccles Institute of Human Genetics, Salt Lake City, UT
| | - Jesse W Rowley
- Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT
- University of Utah Molecular Medicine Program, Eccles Institute of Human Genetics, Salt Lake City, UT
| | - David Salem
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Anne Zufferey
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Audrée Laroche
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Tania Lévesque
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Natalie Patey
- Centre Hospitalier Universitaire de Sainte-Justine, Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montreal, Montreal, QC, Canada
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Christian Lood
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA
| | - Arnaud Droit
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Steven E McKenzie
- Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA
| | - Kellie R Machlus
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; and
| | - Matthew T Rondina
- Axe Neurosciences, Université Laval, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- University of Utah Molecular Medicine Program, Eccles Institute of Human Genetics, Salt Lake City, UT
- Department of Internal Medicine-Geriatric Research Education and Clinical Center (GRECC), George E. Wahlen Veterans Affairs Medical Center (VAMC), Salt Lake City, UT
| | - Steve Lacroix
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Paul R Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Centre de Recherche Arthrite, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| |
Collapse
|
26
|
Liu S, Wang C, Yang H, Zhu T, Jiang H, Chen J. Weighted gene co-expression network analysis identifies FCER1G as a key gene associated with diabetic kidney disease. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1427. [PMID: 33313172 PMCID: PMC7723642 DOI: 10.21037/atm-20-1087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Diabetic kidney disease (DKD) is the primary cause of end-stage renal disease. However, the pathogenesis of DKD remains unclarified, and there is an urgent need for improved treatments. Recently, many crucial genes closely linked to the molecular mechanism underlying various diseases were discovered using weighted gene co-expression network analysis. Methods We used a gene expression omnibus series dataset GSE104948 with 12 renal glomerular DKD tissue samples and 18 control samples obtained from the gene expression omnibus database and performed weighted gene co-expression network analysis. After obtaining the trait-related modules, gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses of the modules were conducted and the key gene associated with DKD was selected from the top two most significant gene ontology terms using the maximal clique centrality method. Finally, we verified the key gene using protein-protein interaction analysis, additional datasets, and explored the relationship between the key gene and DKD renal function using the Nephroseq v5 online database. Results Among the 10 gene co-expression modules identified, the darkorange2 and red modules were highly related to DKD and the normal biological process, respectively. Majority of the genes in the darkorange2 module were related to immune and inflammatory responses, and potentially related to the progression of DKD due to their abnormal up-regulation. After performing sub-network analysis of the genes extracted from the top two most significant gene ontology terms and calculating the maximal clique centrality values of each gene, FCER1G, located at the center of the protein-protein interaction network, was identified as a key gene related to DKD. Furthermore, gene expression omnibus validation in additional datasets also showed that FCER1G was overexpressed in DKD compared with normal tissues. Finally, Pearson’s correlation analysis between the expression of FCER1G and DKD renal function revealed that the abnormal upregulation of FCER1G was related to diabetic glomerular lesions. Conclusions Our study demonstrated for the first time that FCER1G is a crucial gene associated with the pathogenesis of DKD.
Collapse
Affiliation(s)
- Shanshan Liu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Cuili Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Huiying Yang
- Department of Nephrology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Zhu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Lou H, Wojciak-Stothard B, Ruseva MM, Cook HT, Kelleher P, Pickering MC, Mongkolsapaya J, Screaton GR, Xu XN. Autoantibody-dependent amplification of inflammation in SLE. Cell Death Dis 2020; 11:729. [PMID: 32908129 PMCID: PMC7481301 DOI: 10.1038/s41419-020-02928-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Anti-double stranded DNA antibodies (anti-dsDNA) are a hallmark of SLE but their role in disease pathogenesis is not fully resolved. Anti-dsDNA in serum are highly heterogeneous therefore in this study, we aimed to dissect the functional specificities of anti-dsDNA using a panel of human monoclonal antibodies (humAbs) generated from patients with active lupus nephritis. A total of 46 ANA reactive humAbs were isolated and divided into four broad classes based on their reactivity to histones, DNA and Crithidia. Functional analysis indicated that one subclass of antibodies bound strongly to decondensed DNA areas in neutrophil extracellular traps (NETs) and protected NETs from nuclease digestion, similar to the sera from active SLE patients. In addition, these anti-dsDNA antibodies could stimulate type I interferon responses in mononuclear phagocytic cells, or NF-kB activity in endothelial cells, by uptake of NETs-anti-NETs immune complexes and subsequently trigging inflammatory responses in an Fc-gamma receptor (Fcg-R)-dependant manner. Together our data suggest that only a subset of anti-dsDNA antibodies is capable to amplify inflammatory responses by deposit in the nephritic kidney in vivo, protecting NETs digestion as well as uptake of NETs immune complexes into Fcg-R-expressing cells in vitro.
Collapse
Affiliation(s)
- Hantao Lou
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK.
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK.
| | - Beata Wojciak-Stothard
- Department of Medicine, Centre for Pharmacology and Therapeutics, Imperial College London, London, W12 0HS, UK
| | - Marieta M Ruseva
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - H Terence Cook
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - Peter Kelleher
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK
- Department of Infection and Immunity, Charing Cross Hospital North West London Pathology, London, W6 8RF, UK
| | - Matthew C Pickering
- Division of Immunology and Inflammation, Centre for Complement and Inflammation Research, Imperial College London, London, W12 0NN, UK
| | - Juthathip Mongkolsapaya
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
- Division of Medical Sciences, John Radcliffe Hospital, Oxford University, Oxford, OX3 9DU, UK
| | - Gavin R Screaton
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
- Division of Medical Sciences, John Radcliffe Hospital, Oxford University, Oxford, OX3 9DU, UK
| | - Xiao-Ning Xu
- Department of Medicine, Centre for Immunology & Vaccinology, Chelsea and Westminster Hospital, Imperial College London, London, SW10 9NH, UK.
| |
Collapse
|
28
|
Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol 2020; 20:633-643. [PMID: 32782358 PMCID: PMC7418887 DOI: 10.1038/s41577-020-00410-0] [Citation(s) in RCA: 396] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
Antibody-dependent enhancement (ADE) is a mechanism by which the pathogenesis of certain viral infections is enhanced in the presence of sub-neutralizing or cross-reactive non-neutralizing antiviral antibodies. In vitro modelling of ADE has attributed enhanced pathogenesis to Fcγ receptor (FcγR)-mediated viral entry, rather than canonical viral receptor-mediated entry. However, the putative FcγR-dependent mechanisms of ADE overlap with the role of these receptors in mediating antiviral protection in various viral infections, necessitating a detailed understanding of how this diverse family of receptors functions in protection and pathogenesis. Here, we discuss the diversity of immune responses mediated upon FcγR engagement and review the available experimental evidence supporting the role of FcγRs in antiviral protection and pathogenesis through ADE. We explore FcγR engagement in the context of a range of different viral infections, including dengue virus and SARS-CoV, and consider ADE in the context of the ongoing SARS-CoV-2 pandemic. Antibody-dependent enhancement (ADE) has been described as a mechanism that contributes to the pathogenesis of dengue virus infection. Limited evidence also suggests that it can also occur in other viral infections. Here, the authors explore the history of the ADE phenomenon, discuss the diversity of Fc effector functions and consider its potential relevance in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Aaron Gupta
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
29
|
Rubin SJS, Bloom MS, Robinson WH. B cell checkpoints in autoimmune rheumatic diseases. Nat Rev Rheumatol 2020; 15:303-315. [PMID: 30967621 DOI: 10.1038/s41584-019-0211-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
B cells have important functions in the pathogenesis of autoimmune diseases, including autoimmune rheumatic diseases. In addition to producing autoantibodies, B cells contribute to autoimmunity by serving as professional antigen-presenting cells (APCs), producing cytokines, and through additional mechanisms. B cell activation and effector functions are regulated by immune checkpoints, including both activating and inhibitory checkpoint receptors that contribute to the regulation of B cell tolerance, activation, antigen presentation, T cell help, class switching, antibody production and cytokine production. The various activating checkpoint receptors include B cell activating receptors that engage with cognate receptors on T cells or other cells, as well as Toll-like receptors that can provide dual stimulation to B cells via co-engagement with the B cell receptor. Furthermore, various inhibitory checkpoint receptors, including B cell inhibitory receptors, have important functions in regulating B cell development, activation and effector functions. Therapeutically targeting B cell checkpoints represents a promising strategy for the treatment of a variety of autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Samuel J S Rubin
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Michelle S Bloom
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - William H Robinson
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA. .,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. .,VA Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
30
|
Abstract
Immune complexes (ICs) formed by foreign or self-antigens and antibodies in biological fluids affect various tissues and are thought to cause several diseases. Biological and physical properties of IC, abnormal IC amounts, IC deposition and their relationships with disease pathogenesis had been studied. However, the relationship between ICs and each disease is not well understood and little is known of what determined ICs deposition in particular organ and why different organs are affected in different diseases. Recent technological advance enables identification of ICs in particular its antigens in tissues and body fluids, which may provide a key to discover an important trigger for immunological abnormality occurrence. Further identification of their epitopes, that are the exact origin of antigenicity, is developing and may be useful for diagnosis, elucidation of pathogenesis and treatment against IC-induced diseases. Here, we first make an overview of clearance of ICs, IC-induced pathogenesis and biological properties of ICs. Then, we introduce various methods developed to recover ICs from biological fluids or to identify antigens incorporated into ICs. Furthermore, several methods that can be used in epitope mapping for IC antigens are also documented.
Collapse
Affiliation(s)
- Nozomi Aibara
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kaname Ohyama
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
31
|
Aringer M. Inflammatory markers in systemic lupus erythematosus. J Autoimmun 2019; 110:102374. [PMID: 31812331 DOI: 10.1016/j.jaut.2019.102374] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 01/04/2023]
Abstract
While systemic lupus erythematosus (SLE) is an autoantibody and immune complex disease by nature, most of its organ manifestations are in fact inflammatory. SLE activity scores thus heavily rely on assessing inflammation in the various organs. This focus on clinical items demonstrates that routine laboratory markers of inflammation are still limited in their impact. The erythrocyte sedimentation rate (ESR) is used, but represents a rather crude overall measure. Anemia and diminished serum albumin play a role in estimating inflammatory activity, but both are reflecting more than one mechanism, and the association with inflammation is complex. C-reactive protein (CRP) is a better marker for infections than for SLE activity, where there is only a limited association, and procalcitonin (PCT) is also mainly used for detecting severe bacterial infection. Of the cytokines directly induced by immune complexes, type I interferons, interleukin-18 (IL-18) and tumor necrosis factor (TNF) are correlated with inflammatory disease activity. Still, precise and timely measurement is an issue, which is why they are not currently used for routine purposes. While somewhat more robust in the assays, IL-18 binding protein (IL-18BP) and soluble TNF-receptor 2 (TNF-R2), which are related to the respective cytokines, have not yet made it into clinical routine. The same is true for several chemokines that are increased with activity and relatively easy to measure, but still experimental parameters. In the urine, proteinuria leads and is essential for assessing kidney involvement, but may also result from damage. Similar to the situation in serum and plasma, several cytokines and chemokines perform reasonably well in scientific studies, but are not routine parameters. Cellular elements in the urine are more difficult to assess in the routine laboratory, where sufficient routine is not always available. Therefore, the analysis of urinary T cells may have potential for better monitoring renal inflammation.
Collapse
Affiliation(s)
- Martin Aringer
- University Medical Center and Faculty of Medicine Carl Gustav Carus at the TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
32
|
Verbeek JS, Hirose S, Nishimura H. The Complex Association of FcγRIIb With Autoimmune Susceptibility. Front Immunol 2019; 10:2061. [PMID: 31681256 PMCID: PMC6803437 DOI: 10.3389/fimmu.2019.02061] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that Fc γ RIIb -/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the Fc γ R gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the Fc γ RIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.
Collapse
Affiliation(s)
- J Sjef Verbeek
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Sachiko Hirose
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Hiroyuki Nishimura
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| |
Collapse
|
33
|
IL233, an IL-2-IL-33 hybrid cytokine induces prolonged remission of mouse lupus nephritis by targeting Treg cells as a single therapeutic agent. J Autoimmun 2019; 102:133-141. [PMID: 31103267 DOI: 10.1016/j.jaut.2019.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 12/29/2022]
Abstract
Lupus glomerulonephritis (GN) is an autoimmune disease characterized by immune complex-deposition, complement activation and glomerular inflammation. In lupus-prone NZM2328 mice, the occurrence of lupus GN was accompanied by a decrease in Treg cells and an increase in proinflammatory cytokine-producing T cells. Because IL-33 in addition to IL-2 has been shown to be important for Treg cell proliferation and ST2 (IL-33 receptor) positive Treg cells are more potent in suppressor activity, a hybrid cytokine with active domains of IL-2 and IL-33 was generated to target the ST2+ Treg cells as a therapeutic agent to treat lupus GN. Three mouse models were used: spontaneous and Ad-IFNα- accelerated lupus GN in NZM2328 and the lymphoproliferative autoimmune GN in MRL/lpr mice. Daily injections of IL233 for 5 days prevented Ad-IFNα-induced lupus GN and induced remission of spontaneous lupus GN. The remission was permanent in that no relapses were detected. The remission was accompanied by persistent elevation of Treg cells in the renal lymph nodes. IL233 is more potent than IL-2 and IL-33 either singly or in combination in the treatment of lupus GN. The results of this study support the thesis that IL233 should be considered as a novel agent for treating lupus GN.
Collapse
|
34
|
Kuriakose J, Redecke V, Guy C, Zhou J, Wu R, Ippagunta SK, Tillman H, Walker PD, Vogel P, Häcker H. Patrolling monocytes promote the pathogenesis of early lupus-like glomerulonephritis. J Clin Invest 2019; 129:2251-2265. [PMID: 31033479 DOI: 10.1172/jci125116] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 03/12/2019] [Indexed: 12/24/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease with genetic and environmental contributions. Hallmarks of the disease are the appearance of immune complexes (IC) containing autoreactive Abs and TLR-activating nucleic acids, whose deposition in kidney glomeruli is suspected to promote tissue injury and glomerulonephritis (GN). Here, using a mouse model based on the human SLE susceptibility locus TNFAIP3-interacting protein 1 (TNIP1, also known as ABIN1), we investigated the pathogenesis of GN. We found that GN was driven by TLRs but, remarkably, proceeded independently of ICs. Rather, disease in 3 different mouse models and patients with SLE was characterized by glomerular accumulation of patrolling monocytes (PMos), a cell type with an emerging key function in vascular inflammation. Consistent with such function in GN, monocyte-specific deletion of ABIN1 promoted kidney disease, whereas selective elimination of PMos provided protection. In contrast to GN, PMo elimination did not protect from reduced survival or disease symptoms such as IC generation and splenomegaly, suggesting that GN and other inflammatory processes are governed by distinct pathogenic mechanisms. These data identify TLR-activated PMos as the principal component of an intravascular process that contributes to glomerular inflammation and kidney injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Heather Tillman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
35
|
Thurman JM, Yapa R. Complement Therapeutics in Autoimmune Disease. Front Immunol 2019; 10:672. [PMID: 31001274 PMCID: PMC6456694 DOI: 10.3389/fimmu.2019.00672] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022] Open
Abstract
Many autoimmune diseases are characterized by generation of autoantibodies that bind to host proteins or deposit within tissues as a component of immune complexes. The autoantibodies can activate the complement system, which can mediate tissue damage and trigger systemic inflammation. Complement inhibitory drugs may, therefore, be beneficial across a large number of different autoimmune diseases. Many new anti-complement drugs that target specific activation mechanisms or downstream activation fragments are in development. Based on the shared pathophysiology of autoimmune diseases, some of these complement inhibitory drugs may provide benefit across multiple different diseases. In some antibody-mediated autoimmune diseases, however, unique features of the autoantibodies, the target antigens, or the affected tissues may make it advantageous to block individual components or pathways of the complement system. This paper reviews the evidence that complement is involved in various autoimmune diseases, as well as the studies that have examined whether or not complement inhibitors are effective for treating these diseases.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Roshini Yapa
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
36
|
Characteristics of membranoproliferative glomerulonephritis based on a new classification at a single center. Clin Exp Nephrol 2019; 23:852-858. [PMID: 30854618 DOI: 10.1007/s10157-019-01716-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/13/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND Recently, a new classification has been established for membranoproliferative glomerulonephritis (MPGN). However, the effect of the new classification on MPGN treatment is not fully understood. METHODS We conducted a retrospective study of 87 patients with biopsies diagnosed as MPGN. We reclassified 87 MPGN patients diagnosed between 1977 and 2014 at our hospital, according to the new classification, and analyzed both primary immune complex (IC)- and Alternative pathway (AP)-mediated MPGN [corrected] in terms of clinicopathological features, treatment, and renal prognosis. RESULTS Proteinuria was abundant in the IC-mediated MPGN group (p = 0.0063), and the serum albumin level was significantly lower in the IC-mediated MPGN group (p = 0.0186). The serum C3 value was significantly lower in the CP-mediated MPGN group (p = 0.0317). Serum CH50 values were also lower in the CP-mediated MPGN group (p = 0.0404). However, glomerular deposition of C3 showed no significant differences in immunofluorescence findings. The 148.6-month renal survival rate was similar in both groups (p = 0.445). CONCLUSION These results suggested no significant differences in complement activation of the solid phase in local glomeruli and therefore equivalent in renal prognosis [corrected].
Collapse
|
37
|
Chen B, Vousden KA, Naiman B, Turman S, Sun H, Wang S, Vinall LMK, Kemp BP, Kasturiangan S, Rees DG, Grant E, Hinrichs MJ, Eck S, DiGiandomenico A, Jack Borrok M, Ly N, Xiong X, Gonzalez C, Morehouse C, Wang Y, Zhou Y, Cann J, Zhao W, Koelkebeck H, Okubo K, Mayadas TN, Howe D, Griffiths J, Kolbeck R, Herbst R, Sims GP. Humanised effector-null FcγRIIA antibody inhibits immune complex-mediated proinflammatory responses. Ann Rheum Dis 2019; 78:228-237. [PMID: 30459279 PMCID: PMC6352406 DOI: 10.1136/annrheumdis-2018-213523] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/25/2018] [Accepted: 10/30/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.
Collapse
Affiliation(s)
- Bo Chen
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Katherine A Vousden
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Great Abington, UK
| | - Brian Naiman
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Sean Turman
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Hong Sun
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Shu Wang
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
- Viela Bio, Gaithersburg, Maryland, USA
| | - Lisa M K Vinall
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Great Abington, UK
| | - Benjamin P Kemp
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Great Abington, UK
| | - Srinath Kasturiangan
- Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland, USA
| | - D Gareth Rees
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Great Abington, UK
| | - Ethan Grant
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Mary Jane Hinrichs
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Steven Eck
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | | | - M Jack Borrok
- Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Neang Ly
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Ximing Xiong
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Carlos Gonzalez
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | | | - Yue Wang
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Yebin Zhou
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Jennifer Cann
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Weiguang Zhao
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Holly Koelkebeck
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Koshu Okubo
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - David Howe
- Department of Clinical Development, MedImmune Ltd, Granta Park, Great Abington, UK
| | - Janet Griffiths
- Department of Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Ronald Herbst
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Gary P Sims
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| |
Collapse
|
38
|
Nakajima R, Miyagaki T, Morimura S, Fukasawa T, Oka T, Yoshizaki A, Sugaya M, Sato S. Exacerbated Immune Complex-Mediated Vascular Injury in Mice with Heterozygous Deficiency of Aryl Hydrocarbon Receptor through Upregulation of Fcγ Receptor III Expression on Macrophages. J Invest Dermatol 2018; 138:2195-2204. [DOI: 10.1016/j.jid.2018.03.1520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 03/13/2018] [Accepted: 03/23/2018] [Indexed: 11/28/2022]
|
39
|
Establishment and evaluation of a general dissociation technique for antibodies in circulating immune complexes. Clin Exp Med 2018; 19:65-75. [PMID: 30120614 PMCID: PMC6394588 DOI: 10.1007/s10238-018-0523-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/08/2018] [Indexed: 11/06/2022]
Abstract
This study aimed to establish a general and efficient dissociation technique for detecting antibodies in circulating immune complexes (CICs) in serum and to evaluate its clinical application. CICs were efficiently separated from specimens using polyethylene glycol double-precipitation. The best conditions for anti-HBs dissociation from HBsAg-ICs were a pH of 1.80, incubation at 15 °C for 5–10 min, and detection within 10 min after neutralization. The mean dissociation rate, reproducibility, mean dissociation recovery rate and specificity of the new technique were 64.3%, < 5.97, 95.4 and 100%, respectively. They had a favourable linear relationship (r = 0.9932), and the stability of the reagents exceeded 24 months, except the CIC antibody dissociation reagent (> 12 months). Conditions for the dissociation of other CICs tested were similar, but there were differences in the rate of antibody dissociation. Different HBV-M patterns had significantly different levels and rates of antibody dissociation from HBsAg-IC (P < 0.05), and the detection rates of the corresponding antibodies in HCV, core-anti-HCV core antibody (HCV-ICs), HIV P24-anti-HIV P24 antibody (HIV-ICs), insulin-anti-insulin antibody (INS-ICs) and thyroid globulin-anti-thyroid globulin antibody CICs (TG-ICs) were 34.8, 66.7, 20 and 14.3%, respectively. These data suggest that our CIC antibody dissociation technique is a good general pretreatment technique for the detection of antibodies after the precipitation, separation and dissociation of multiple CICs.
Collapse
|
40
|
Zhang H, Liu L, Li L. Lentivirus-mediated knockdown of FcγRI (CD64) attenuated lupus nephritis via inhibition of NF-κB regulating NLRP3 inflammasome activation in MRL/lpr mice. J Pharmacol Sci 2018; 137:342-349. [DOI: 10.1016/j.jphs.2018.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/21/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022] Open
|
41
|
Olaru F, Döbel T, Lonsdorf AS, Oehrl S, Maas M, Enk AH, Schmitz M, Gröne EF, Gröne HJ, Schäkel K. Intracapillary immune complexes recruit and activate slan-expressing CD16+ monocytes in human lupus nephritis. JCI Insight 2018; 3:96492. [PMID: 29875315 DOI: 10.1172/jci.insight.96492] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 04/24/2018] [Indexed: 12/30/2022] Open
Abstract
Lupus nephritis is a major cause of morbidity in patients with systemic lupus erythematosus. Among the different types of lupus nephritis, intracapillary immune complex (IC) deposition and accumulation of monocytes are hallmarks of lupus nephritis class III and IV. The relevance of intracapillary ICs in terms of monocyte recruitment and activation, as well as the nature and function of these monocytes are not well understood. For the early focal form of lupus nephritis (class III) we demonstrate a selective accumulation of the proinflammatory population of 6-sulfo LacNAc+ (slan) monocytes (slanMo), which locally expressed TNF-α. Immobilized ICs induced a direct recruitment of slanMo from the microcirculation via interaction with Fc γ receptor IIIA (CD16). Interestingly, intravenous immunoglobulins blocked CD16 and prevented cell recruitment. Engagement of immobilized ICs by slanMo induced the production of neutrophil-attracting chemokine CXCL2 as well as TNF-α, which in a forward feedback loop stimulated endothelial cells to produce the slanMo-recruiting chemokine CX3CL1 (fractalkine). In conclusion, we observed that expression of CD16 equips slanMo with a unique capacity to orchestrate early IC-induced inflammatory responses in glomeruli and identified slanMo as a pathogenic proinflammatory cell type in lupus nephritis.
Collapse
Affiliation(s)
- Florina Olaru
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anke S Lonsdorf
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie Oehrl
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Maas
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander H Enk
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty, Technische Universität (TU) Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), Medical Faculty, TU Dresden, Dresden, Germany
| | - Elisabeth F Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann-J Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
42
|
Flores-Mendoza G, Sansón SP, Rodríguez-Castro S, Crispín JC, Rosetti F. Mechanisms of Tissue Injury in Lupus Nephritis. Trends Mol Med 2018. [PMID: 29526595 DOI: 10.1016/j.molmed.2018.02.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Disease heterogeneity remains a major challenge for the understanding of systemic lupus erythematosus (SLE). Recent work has revealed the important role of nonimmune factors in the development of end-organ damage involvement, shifting the current paradigm that views SLE as a disease inflicted by a disturbed immune system on passive target organs. Here, we discuss the pathogenesis of lupus nephritis in a comprehensive manner, by incorporating the role that target organs play by withstanding and modulating the local inflammatory response. Moreover, we consider the effects that genetic variants exert on immune and nonimmune cells in order to shape the phenotype of the disease in each affected individual.
Collapse
Affiliation(s)
- Giovanna Flores-Mendoza
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; Doctorado en Ciencias Biológicas, Facultad de Medicina, UNAM, Mexico City, Mexico
| | - Stephanie P Sansón
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, UNAM, Mexico City, Mexico
| | - Santiago Rodríguez-Castro
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, UNAM, Mexico City, Mexico
| | - José C Crispín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.
| | - Florencia Rosetti
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.
| |
Collapse
|
43
|
Dai Y, Hu Z, Chen Y, Lou B, Cui D, Xu A, Rao Y, He J, Yang J, Zeng X, Xu X, Wang G, Xu J, Zhou T, Sun C, Cheng J. A novel general and efficient technique for dissociating antigen in circulating immune complexes. Electrophoresis 2018; 39:406-416. [PMID: 28972666 DOI: 10.1002/elps.201700246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 02/05/2023]
Abstract
Circulating immune complexes (CICs) are produced during the immune response. It is more clinically important to establish a general and efficient CICs dissociation technique for the detection of antigens for CICs other than the detection of free antigens in the serum. Polyethylene glycol (PEG) two-precipitation separation and glycine-HCl as a buffer system were employed to develop a general and efficient buffer dissociation technique to separate CICs from serum and dissociate antigens from CICs. The measurement value of new PEG two-precipitation separation technique was higher than traditional PEG precipitation separation technique. There were slight differences in the dissociation conditions of HCV Core-IC, HIV P24-IC, Ins-IC and TG-IC as compared to HBsAg-IC. The detection of antigens in HBsAg-IC, HCV Core-IC, HIV P24-IC, Ins-IC and TG-IC with this technique was superior to that with HCl Dissociation, Trypsin Digestion or Immune Complex Transfer technique. PEG two-precipitation dissociation technique may reduce macromolecular protein and the adhesion of free antigens during the co-precipitation, which increases the efficiency of separation and precipitation of CICs. This technique also avoids the damage of reagents to antigens, assuring the repeatability, reliability and validity. Thus, this technique is application in samples negative or positive for free antigens.
Collapse
Affiliation(s)
- Yuzhu Dai
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
| | - Zhengjun Hu
- Department of Clinical Laboratory, First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine, Hangzhou, P. R. China
| | - Yu Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Bin Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Dawei Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Aifang Xu
- Department of Clinical Laboratory Science, Hangzhou Xixi Hospital, Hangzhou, P. R. China
| | - Yueli Rao
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
| | - Jiahui He
- Department of Clinical Laboratory Science, Hangzhou Xixi Hospital, Hangzhou, P. R. China
| | - Jiezuan Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Xianming Zeng
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
| | - Xujian Xu
- Department of Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Guozheng Wang
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
| | - Jian Xu
- Medical Technology College, Zhejiang University of Traditional Chinese Medicine, Hangzhou, P. R. China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, P. R. China
| | - Changgui Sun
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
| | - Jun Cheng
- Department of Clinical Laboratory, The 117th Hospital of PLA, Hangzhou, P. R. China
- Medical Technology College, Zhejiang University of Traditional Chinese Medicine, Hangzhou, P. R. China
- Faculty of Graduate Studies, Wenzhou Medical University, Wenzhou, P. R. China
| |
Collapse
|
44
|
Qi Q, Li H, Lin ZM, Yang XQ, Zhu FH, Liu YT, Shao MJ, Zhang LY, Xu YS, Yan YX, Sun LL, He SJ, Tang W, Zuo JP. (5R)-5-hydroxytriptolide ameliorates anti-glomerular basement membrane glomerulonephritis in NZW mice by regulating Fcγ receptor signaling. Acta Pharmacol Sin 2018; 39:107-116. [PMID: 28880016 DOI: 10.1038/aps.2017.88] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/17/2017] [Indexed: 12/20/2022]
Abstract
(5R)-5-hydroxytriptolide (LLDT-8) is a novel triptolide analog that has been identified as a promising candidate for treating autoimmune diseases and has been shown to be effective in treating murine collagen-induced arthritis and lupus nephritis. In the present study, we investigated the therapeutic effect and possible mechanism of action of LLDT-8 in a murine anti-glomerular basement membrane (GBM) glomerulonephritis model. NZW mice were injected with rabbit anti-GBM serum (500 μL, ip). The mice were orally treated with LLDT-8 (0.125 mg/kg, every other day) or a positive control prednisolone (2 mg/kg every day) for 14 d. Blood and urine samples as well as spleen and kidney tissues were collected for analyses. LLDT-8 treatment did not affect the generation of mouse anti-rabbit antibodies. LLDT-8 significantly reversed established proteinuria, improved renal histopathology and attenuated renal dysfunction in glomerulonephritis mice. Furthermore, LLDT-8 inhibited inflammation in the kidney evidenced by significantly decreasing C3 and IgG deposition, reducing the levels of the pathogenic cytokines TNF-α, IL-6, IL-17, and IFN-γ, and reducing related chemokine expression and leukocyte infiltration in kidneys. Moreover, LLDT-8 treatment significantly increased the expression of FcγRIIB in the kidney and spleen. In addition, the treatment restored the reduced expression of FcγRIIB on the surface of kidney effector cells, CD11b+ cells, and interfered with FcγR-dependent signaling, especially FcγRIIB-mediated downstream kinases, such as BTK. These results demonstrate that LLDT-8 ameliorates anti-GBM glomerulonephritis by regulating the Fcγ receptor signaling.
Collapse
|
45
|
Kim YY, Park KT, Jang SY, Lee KH, Byun JY, Suh KH, Lee YM, Kim YH, Hwang KW. HM71224, a selective Bruton's tyrosine kinase inhibitor, attenuates the development of murine lupus. Arthritis Res Ther 2017; 19:211. [PMID: 28950886 PMCID: PMC5615432 DOI: 10.1186/s13075-017-1402-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/01/2017] [Indexed: 01/16/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) is associated with B cell hyperactivity, and lupus nephritis (LN), in particular, is promoted by the production of autoantibodies and immune complex deposition. Bruton’s tyrosine kinase (BTK) plays critical roles in B cell receptor-related and Fc receptor-related signaling. We aimed to investigate the impact of therapeutic intervention with HM71224 (LY3337641), a selective BTK inhibitor, on the development of murine SLE-like disease features. Methods We examined the therapeutic effects of HM71224 on SLE-like disease features in MRL/lpr and NZB/W F1 mice. The disease-related skin lesion was macroscopically observed in MRL/lpr mice, and the impact on splenomegaly and lymphadenopathy was determined by the weight of the spleen and cervical lymph node. The renal function was evaluated by measuring blood urea nitrogen, serum creatinine, and urine protein, and the renal damage was assessed by histopathological grading. Survival rate was observed during the administration period. The impact of B cell inhibition was investigated in splenocytes from both mice using flow cytometry. Autoantibody was measured in serum by ELISA. Results HM71224 effectively suppressed splenic B220+GL7+, B220+CD138+, and B220+CD69+ B cell counts, and anti-dsDNA IgG and reduced splenomegaly and lymph node enlargement. The compound also prevented skin lesions caused by lupus development, ameliorated renal inflammation and damage with increased blood urea nitrogen and creatinine, and decreased proteinuria. Furthermore, HM71224 also decreased mortality from lupus development in both mouse models. Conclusion Our results indicate that inhibition of BTK by HM71224 effectively reduced B cell hyperactivity and significantly attenuated the development of SLE and LN in rodent SLE models.
Collapse
Affiliation(s)
- Yu-Yon Kim
- Host Defense Modulation Lab, College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea.,Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Ki Tae Park
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Sun Young Jang
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Kyu Hang Lee
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Joo-Yun Byun
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Kwee Hyun Suh
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Young-Mi Lee
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea
| | - Young Hoon Kim
- Hanmi Research Center, Hanmi Pharm. Co. Ltd, 550 Dongtangiheung-Ro, Hwaseong-Si, Gyeonggi-Do, 18469, Republic of Korea.
| | - Kwang Woo Hwang
- Host Defense Modulation Lab, College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
46
|
McDonald G, Medina CO, Pilichowska M, Kearney JF, Shinkura R, Selsing E, Wortis HH, Honjo T, Imanishi-Kari T. Accelerated Systemic Autoimmunity in the Absence of Somatic Hypermutation in 564Igi: A Mouse Model of Systemic Lupus with Knocked-In Heavy and Light Chain Genes. Front Immunol 2017; 8:1094. [PMID: 28955333 PMCID: PMC5601273 DOI: 10.3389/fimmu.2017.01094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
564Igi mice have knocked-in immunoglobulin (Ig) heavy (H) and light (L) chain genes that encode an autoantibody recognizing RNA. Previously, we showed that these mice produce pathogenic IgG autoantibodies when activation-induced deaminase (AID) is expressed in pre-B and immature B cells but not when it is expressed only in mature B cells. AID has two functions; it is necessary for somatic hypermutation (SHM) and class switch recombination (CSR). To determine the role of each of these functions in the generation of pathogenic autoantibodies, we generated 564Igi mice that carry a mutant AID-encoding gene, Aicda (AicdaG23S), which is capable of promoting CSR but not SHM. We found that 564Igi AicdaG23S mice secreted class-switched antibodies (Abs) at levels approximately equal to 564Igi mice. However, compared to 564Igi mice, 564Igi AicdaG23S mice had increased pathogenic IgG Abs and severe systemic lupus erythematosus-like disease, including, glomerulonephritis, and early death. We suggest that in 564Igi mice SHM by AID changes Ig receptors away from self reactivity, thereby mitigating the production of autoantibody, providing a novel mechanism of tolerance.
Collapse
Affiliation(s)
- Gabrielle McDonald
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Carlos O Medina
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Monika Pilichowska
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, United States
| | - John F Kearney
- Department of Microbiology, University of Alabama, Birmingham, AL, United States
| | - Reiko Shinkura
- Department of Immunology, Nagahama Institute of Bioscience and Technology, Nagahama, Japan
| | - Erik Selsing
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Henry H Wortis
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Thereza Imanishi-Kari
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
47
|
|
48
|
Gordon RA, Herter JM, Rosetti F, Campbell AM, Nishi H, Kashgarian M, Bastacky SI, Marinov A, Nickerson KM, Mayadas TN, Shlomchik MJ. Lupus and proliferative nephritis are PAD4 independent in murine models. JCI Insight 2017; 2:92926. [PMID: 28515361 DOI: 10.1172/jci.insight.92926] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022] Open
Abstract
Though recent reports suggest that neutrophil extracellular traps (NETs) are a source of antigenic nucleic acids in systemic lupus erythematosus (SLE), we recently showed that inhibition of NETs by targeting the NADPH oxidase complex via cytochrome b-245, β polypeptide (cybb) deletion exacerbated disease in the MRL.Faslpr lupus mouse model. While these data challenge the paradigm that NETs promote lupus, it is conceivable that global regulatory properties of cybb and cybb-independent NETs confound these findings. Furthermore, recent reports indicate that inhibitors of peptidyl arginine deiminase, type IV (Padi4), a distal mediator of NET formation, improve lupus in murine models. Here, to clarify the contribution of NETs to SLE, we employed a genetic approach to delete Padi4 in the MRL.Faslpr model and used a pharmacological approach to inhibit PADs in both the anti-glomerular basement membrane model of proliferative nephritis and a human-serum-transfer model of SLE. In contrast to prior inhibitor studies, we found that deletion of Padi4 did not ameliorate any aspect of nephritis, loss of tolerance, or immune activation. Pharmacological inhibition of PAD activity had no effect on end-organ damage in inducible models of glomerulonephritis. These data provide a direct challenge to the concept that NETs promote autoimmunity and target organ injury in SLE.
Collapse
Affiliation(s)
- Rachael A Gordon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jan M Herter
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Florencia Rosetti
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Hiroshi Nishi
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony Marinov
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin M Nickerson
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Immunobiology.,Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
49
|
Kitai M, Fukuda N, Ueno T, Endo M, Maruyama T, Abe M, Okada K, Soma M, Matsumoto K. Effects of a spleen tyrosine kinase inhibitor on progression of the lupus nephritis in mice. J Pharmacol Sci 2017; 134:29-36. [PMID: 28479222 DOI: 10.1016/j.jphs.2017.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 01/14/2023] Open
Abstract
The Fc receptors (FcR) have pivotal roles in the pathogenesis of the autoimmune glomerulonephritis. We therefore investigated the effects of a Syk inhibitor on the progression of lupus nephritis and SH3 domain binding protein 2 and p38MAP kinase signalings in mice. NZB/W F1 mice, a model of lupus nephritis, received a Syk inhibitor R406. Western blotting and immunohistochemistry revealed that R406 treatment significantly delayed the appearance of proteinuria, histologically improved their glomerulosclerosis and inhibited the increased the expression of MCP-1 and TGF-β1 mRNAs and the nephrin and podocin proteins in the kidney. The treatment suppressed the phosphorylation of 3BP2 in white blood cells from the spleen and significantly inhibited the phosphorylation of p38MAPK in the kidney but did not affect expression of neonatal Fc receptor. These findings indicate the important roles and mechanisms of Fcγ receptors I and III in the development of autoimmune glomerulonephritis and suggest the possible application of Syk inhibitors as novel medicines for the glomerulonephritis.
Collapse
Affiliation(s)
- Maki Kitai
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Noboru Fukuda
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan; Research Center of Nihon University, Tokyo, Japan.
| | - Takahiro Ueno
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Morito Endo
- Faculty of Human Health Science, Hachinohe Gakuin University, Hachinohe, Aomori, Japan
| | - Takashi Maruyama
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Masanori Abe
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuyoshi Okada
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Masayoshi Soma
- Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Koichi Matsumoto
- Division of Nephrology Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Morawski PA, Bolland S. Expanding the B Cell-Centric View of Systemic Lupus Erythematosus. Trends Immunol 2017; 38:373-382. [PMID: 28274696 DOI: 10.1016/j.it.2017.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/26/2017] [Accepted: 02/08/2017] [Indexed: 12/29/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by a breakdown of self-tolerance in B cells and the production of antibodies against nuclear self-antigens. Increasing evidence supports the notion that additional cellular contributors beyond B cells are important for lupus pathogenesis. In this review we consider recent advances regarding both the pathogenic and the regulatory role of lymphocytes in SLE beyond the production of IgG autoantibodies. We also discuss various inflammatory effector cell types involved in cytokine production, removal of self-antigens, and responses to autoreactive IgE antibodies. We aim to integrate these ideas to expand the current understanding of the cellular components that contribute to disease progression and ultimately help in the design of novel, targeted therapeutics.
Collapse
Affiliation(s)
- Peter A Morawski
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Silvia Bolland
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|