1
|
Watanabe-Kusunoki K, Anders HJ. Balancing efficacy and safety of complement inhibitors. J Autoimmun 2024; 145:103216. [PMID: 38552408 DOI: 10.1016/j.jaut.2024.103216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 05/15/2024]
Abstract
Complement inhibitors have been approved for several immune-mediated diseases and they are considered the next paradigm-shifting approach in the treatment of glomerulonephritis. The hierarchical organization of the complement system offers numerous molecular targets for therapeutic intervention. However, complement is an integral element of host defense and therefore complement inhibition can be associated with serious infectious complications. Here we give a closer look to the hierarchical complement system and how interfering with proximal versus distal or selective versus unselective molecular targets could determine efficacy and safety. Furthermore, we propose to consider the type of disease, immunological activity, and patient immunocompetence when stratifying patients, e.g., proximal/unselective targets for highly active and potentially fatal diseases while distal and selective targets may suit more chronic disease conditions with low or moderate disease activity requiring persistent complement blockade in patients with concomitant immunodeficiency. Certainly, there exists substantial promise for anti-complement therapeutics. However, balancing efficacy and safety will be key to establish powerful treatment effects with minimal adverse events, especially when complement blockade is continued over longer periods of time in chronic disorders.
Collapse
Affiliation(s)
- Kanako Watanabe-Kusunoki
- Renal Division, Department of Medicine IV, Ludwig-Maximilians (LMU) University Hospital, LMU Munich, Germany; Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, Ludwig-Maximilians (LMU) University Hospital, LMU Munich, Germany.
| |
Collapse
|
2
|
Zhao H, Zheng D. Revealing common differential mRNAs, signaling pathways, and immune cells in blood, glomeruli, and tubulointerstitium of lupus nephritis patients based on transcriptomic data. Ren Fail 2023; 45:2215344. [PMID: 37334926 DOI: 10.1080/0886022x.2023.2215344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/11/2023] [Accepted: 05/14/2023] [Indexed: 06/21/2023] Open
Abstract
Lupus nephritis (LN) is a potentially fatal autoimmune disease. The purpose of this study was to find potential key molecular markers of LN to aid in the early diagnosis and management of the disease. Datasets GSE99967_blood, GSE32591_glomeruli, and GSE32591_tubulointerstitium were included in this study. Differentially expressed mRNAs (DEmRNAs) were identified between the normal control and LN groups using the limma package in R. Common DEmRNAs in the three datasets were taken. Subsequently, functional enrichment analysis, immune correlation analysis, receiver operating characteristic (ROC) curve analysis and real-time polymerase chain reaction (RT-PCR) verification were performed. In this study, 11 common DEmRNAs were obtained and all of them were up-regulated. In protein-protein interaction (PPI) networks, we found that MX dynamin like GTPase 1 (MX1) and radical S-adenosyl methionine domain containing 2 (RSAD2) had the highest interaction score (0.997). Functional enrichment analysis revealed that MX1 and RSAD2 were enriched in influenza A and hepatitis C signaling pathways. The area under the curve (AUC) values of interferon-induced protein 44 (IFI44) and MX1 in GSE32591_glomeruli and GSE32591_tubulointerstitium datasets are 1, which is worthy of further study on their diagnostic value and molecular mechanism. The xCell analysis showed abnormal distribution of granulocyte-macrophage progenitor (GMP) cells in blood, glomeruli, and tubulointerstitium. Pearson's correlation analysis found that GMP cells were significantly correlated with lactotransferrin (LTF) and cell cycle. Identification of common DEmRNAs and key pathways in the blood, glomeruli, and tubulointerstitium of patients with LN provides potential research directions for exploring the molecular mechanisms of the disease.
Collapse
Affiliation(s)
- Haifang Zhao
- Department of Nephrology, Dongying People's Hospital, Dongying, China
| | - Dongxia Zheng
- Department of Nephrology, Dongying People's Hospital, Dongying, China
| |
Collapse
|
3
|
Liang X, Li P, Jiang J, Xin J, Luo J, Li J, Chen P, Ren K, Zhou Q, Guo B, Zhou X, Chen J, He L, Yang H, Hu W, Ma S, Li B, Chen X, Shi D, Li J. Transcriptomics unveils immune metabolic disruption and a novel biomarker of mortality in patients with HBV-related acute-on-chronic liver failure. JHEP Rep 2023; 5:100848. [PMID: 37583946 PMCID: PMC10424217 DOI: 10.1016/j.jhepr.2023.100848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/16/2023] [Accepted: 07/01/2023] [Indexed: 08/17/2023] Open
Abstract
Background & Aims HBV-related acute-on-chronic liver failure (HBV-ACLF) is a complex syndrome associated with high short-term mortality. This study aims to reveal the molecular basis and identify novel HBV-ACLF biomarkers. Methods Seventy patients with HBV-ACLF and different short-term (28 days) outcomes underwent transcriptome sequencing using peripheral blood mononuclear cells. Candidate biomarkers were confirmed in two external cohorts using ELISA. Results Cellular composition analysis with peripheral blood mononuclear cell transcriptomics showed that the proportions of monocytes, T cells and natural killer cells were significantly correlated with 28-day mortality. Significant metabolic dysregulation of carbohydrate, energy and amino acid metabolism was observed in ACLF non-survivors. V-set and immunoglobulin domain-containing 4 (VSIG4) was the most robust predictor of patient survival (adjusted p = 1.74 × 10-16; variable importance in the projection = 1.21; AUROC = 0.89) and was significantly correlated with pathways involved in the progression of ACLF, including inflammation, oxidative phosphorylation, tricarboxylic acid cycle and T-cell activation/differentiation. Plasma VSIG4 analysis externally validated its diagnostic value in ACLF (compared with chronic liver disease and healthy groups, AUROC = 0.983). The prognostic performance for 28-/90-day mortality (AUROCs = 0.769/0.767) was comparable to that of three commonly used scores (COSSH-ACLFs, 0.867/0.884; CLIF-C ACLFs, 0.840/0.835; MELD-Na, 0.710/0.737). Plasma VSIG4 level, as an independent predictor, could be used to improve the prognostic performance of clinical scores. Risk stratification based on VSIG4 expression levels (>122 μg/ml) identified patients with ACLF at a high risk of death. The generality of VSIG4 in other etiologies was validated. Conclusions This study reveals that immune-metabolism disorder underlies poor ACLF outcomes. VSIG4 may be helpful as a diagnostic and prognostic biomarker in clinical practice. Impact and implications Acute-on-chronic liver failure (ACLF) is a lethal clinical syndrome associated with high mortality. We found significant immune cell alterations and metabolic dysregulation that were linked to high mortality in patients with HBV-ACLF based on transcriptomics using peripheral blood mononuclear cells. We identified VSIG4 (V-set and immunoglobulin domain-containing 4) as a diagnostic and prognostic biomarker in ACLF, which could specifically identify patients with ACLF at a high risk of death.
Collapse
Affiliation(s)
- Xi Liang
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Peng Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaojiao Xin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinjin Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaqi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Pengcheng Chen
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xingping Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaxian Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lulu He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wen Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shiwen Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Bingqi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Chen
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital, Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jun Li
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chinese Group on the Study of Severe Hepatitis B (COSSH)
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital, Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Ebstein SY, Rafique A, Zhou Y, Krasco A, Montalvo-Ortiz W, Yu L, Custodio L, Adam RC, Bloch N, Lee K, Adewale F, Vergata D, Luz A, Coquery S, Daniel B, Ullman E, Franklin MC, Hermann A, Huang T, Olson W, Davis S, Murphy AJ, Sleeman MA, Wei J, Skokos D. VSIG4 interaction with heparan sulfates inhibits VSIG4-complement binding. Glycobiology 2023; 33:591-604. [PMID: 37341346 PMCID: PMC10426322 DOI: 10.1093/glycob/cwad050] [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: 07/18/2022] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023] Open
Abstract
V-set and immunoglobulin domain-containing 4 (VSIG4) is a complement receptor of the immunoglobulin superfamily that is specifically expressed on tissue resident macrophages, and its many reported functions and binding partners suggest a complex role in immune function. VSIG4 is reported to have a role in immune surveillance as well as in modulating diverse disease phenotypes such as infections, autoimmune conditions, and cancer. However, the mechanism(s) governing VSIG4's complex, context-dependent role in immune regulation remains elusive. Here, we identify cell surface and soluble glycosaminoglycans, specifically heparan sulfates, as novel binding partners of VSIG4. We demonstrate that genetic deletion of heparan sulfate synthesis enzymes or cleavage of cell-surface heparan sulfates reduced VSIG4 binding to the cell surface. Furthermore, binding studies demonstrate that VSIG4 interacts directly with heparan sulfates, with a preference for highly sulfated moieties and longer glycosaminoglycan chains. To assess the impact on VSIG4 biology, we show that heparan sulfates compete with known VSIG4 binding partners C3b and iC3b. Furthermore, mutagenesis studies indicate that this competition occurs through overlapping binding epitopes for heparan sulfates and complement on VSIG4. Together these data suggest a novel role for heparan sulfates in VSIG4-dependent immune modulation.
Collapse
Affiliation(s)
- Sarah Y Ebstein
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Ashique Rafique
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Yi Zhou
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Amanda Krasco
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Welby Montalvo-Ortiz
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Lola Yu
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Luisaidy Custodio
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Rene C Adam
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Nicolin Bloch
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Ken Lee
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Funmilola Adewale
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Dominic Vergata
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Antonio Luz
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Sebastien Coquery
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Benjamin Daniel
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Erica Ullman
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Matthew C Franklin
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Aynur Hermann
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Tammy Huang
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - William Olson
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Samuel Davis
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Andrew J Murphy
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Matthew A Sleeman
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Joyce Wei
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Dimitris Skokos
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| |
Collapse
|
5
|
Holers VM. Complement therapeutics are coming of age in rheumatology. Nat Rev Rheumatol 2023; 19:470-485. [PMID: 37337038 DOI: 10.1038/s41584-023-00981-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/21/2023]
Abstract
The complement system was described over 100 years ago, and it is well established that activation of this pathway accompanies the great majority of autoimmune and inflammatory diseases. In addition, over three decades of work in murine models of human disease have nearly universally demonstrated that complement activation is upstream of tissue injury and the engagement of pro-inflammatory mechanisms such as the elaboration of cytokines and chemokines, as well as myeloid cell recruitment and activation. With that background, and taking advantage of advances in the development of biologic and small-molecule therapeutics, the creation and clinical evaluation of complement therapeutics is now rapidly expanding. This article reviews the current state of the complement therapeutics field, with a focus on their use in diseases cared for or consulted upon by rheumatologists. Included is an overview of the activation mechanisms and components of the system, in addition to the mechanisms by which the complement system interacts with other immune system constituents. The various therapeutic approaches to modulating the system in rheumatic and autoimmune diseases are reviewed. To understand how best to clinically assess the complement system, methods of its evaluation are described. Finally, next-generation therapeutic and diagnostic advances that can be envisioned for the future are discussed.
Collapse
Affiliation(s)
- V Michael Holers
- Medicine/Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
6
|
Liu B, Cheng L, Gao H, Zhang J, Dong Y, Gao W, Yuan S, Gong T, Huang W. The biology of VSIG4: Implications for the treatment of immune-mediated inflammatory diseases and cancer. Cancer Lett 2023; 553:215996. [PMID: 36343787 DOI: 10.1016/j.canlet.2022.215996] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
V-set and immunoglobulin domain containing 4 (VSIG4), a type I transmembrane receptor exclusively expressed in a subset of tissue-resident macrophages, plays a pivotal role in clearing C3-opsonized pathogens and their byproducts from the circulation. VSIG4 maintains immune homeostasis by suppressing the activation of complement pathways or T cells and inducing regulatory T-cell differentiation, thereby inhibiting the development of immune-mediated inflammatory diseases but enhancing cancer progression. Consequently, VSIG4 exhibits a potential therapeutic effect for immune-mediated inflammatory diseases, but also is regarded as a novel target of immune checkpoint inhibition in cancer therapy. Recently, soluble VSIG4, the extracellular domain of VSIG4, shed from the surface of macrophages, has been found to be a biomarker to define macrophage activation-related diseases. This review mainly summarizes recent new findings of VSIG4 in macrophage phagocytosis and immune homeostasis, and discusses its potential diagnostic and therapeutic usage in infection, inflammation, and cancer.
Collapse
Affiliation(s)
- Bei Liu
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; PLA 307 Clinical College of Anhui Medical University, Beijing, 100071, China
| | - Li Cheng
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Honghao Gao
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Jiale Zhang
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital, Fuchenglu 6#, Haidian District, Beijing, 100048, China
| | - Yanxin Dong
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital, Fuchenglu 6#, Haidian District, Beijing, 100048, China
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Boston, MA, 02021, USA
| | - Shunzong Yuan
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; PLA 307 Clinical College of Anhui Medical University, Beijing, 100071, China.
| | - Taiqian Gong
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital, Fuchenglu 6#, Haidian District, Beijing, 100048, China.
| | - Wenrong Huang
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
7
|
Li Y, Wang Q, Li J, Li A, Wang Q, Zhang Q, Chen Y. Therapeutic modulation of V Set and Ig domain-containing 4 (VSIG4) signaling in immune and inflammatory diseases. Cytotherapy 2023; 25:561-572. [PMID: 36642683 DOI: 10.1016/j.jcyt.2022.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
Inflammation is the result of acute and chronic stresses, caused by emotional or physical trauma, or nutritional or environmental pollutants, and brings serious harm to human life and health. As an important cellular component of the innate immune barrier, the macrophage plays a key role in maintaining tissue homeostasis and promoting tissue repair by controlling infection and resolving inflammation. Several studies suggest that V Set and Ig domain-containing 4 is specifically expressed in tissue macrophages and is associated with a variety of inflammatory diseases. In this paper, we mainly summarize the recent research on V Set and Ig domain-containing 4 structures, functions, function and roles in acute and chronic inflammatory diseases, and provide a novel therapeutic avenue for the treatment of inflammatory diseases, including nervous system, urinary, respiratory and metabolic diseases.
Collapse
Affiliation(s)
- You Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Jiaxin Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Aohan Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China.
| | - Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China.
| |
Collapse
|
8
|
Holers VM. Contributions of animal models to mechanistic understandings of antibody-dependent disease and roles of the amplification loop. Immunol Rev 2023; 313:181-193. [PMID: 36111456 DOI: 10.1111/imr.13136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The complement system plays an important pathophysiologic role in human diseases associated with immune or ischemic insults. In addition to understanding the effector mechanisms that are important for the biological effects of the system, substantial efforts have gone into understanding which specific complement activation pathways generate these potent effects. These approaches include the use of gene-targeted mice and specific pathway inhibitors, as well as the integration of human disease genetic and biomarker studies. In some disease states, it is quite clear that the alternate pathway plays a unique role in the initiation of the complement system. However, although initially a widely unexpected finding, it has now been shown in many tissue-based disease models and in initial human studies that engagement of the amplification loop is also essential for tissue injury when the classical and/or lectin pathways initiate pathway activation through pathogenic autoantibodies. This review provides evidence for such a conclusion through animal models, focusing on pathogenic antibody passive transfer models but also other relevant experimental systems. These data, along with initial biomarkers and clinical trial outcomes in human diseases that are associated with pathogenic autoantibodies, suggest that targeting the alternative pathway amplification loop may have near-universal therapeutic utility for tissue-based diseases.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
9
|
Abstract
Dysregulation and accelerated activation of the alternative pathway (AP) of complement is known to cause or accentuate several pathologic conditions in which kidney injury leads to the appearance of hematuria and proteinuria and ultimately to the development of chronic renal failure. Multiple genetic and acquired defects involving plasma- and membrane-associated proteins are probably necessary to impair the protection of host tissues and to confer a significant predisposition to AP-mediated kidney diseases. This review aims to explore how our current understanding will make it possible to identify the mechanisms that underlie AP-mediated kidney diseases and to discuss the available clinical evidence that supports complement-directed therapies. Although the value of limiting uncontrolled complement activation has long been recognized, incorporating complement-targeted treatments into clinical use has proved challenging. Availability of anti-complement therapy has dramatically transformed the outcome of atypical hemolytic uremic syndrome, one of the most severe kidney diseases. Innovative drugs that directly counteract AP dysregulation have also opened new perspectives for the management of other kidney diseases in which complement activation is involved. However, gained experience indicates that the choice of drug should be tailored to each patient's characteristics, including clinical, histologic, genetic, and biochemical parameters. Successfully treating patients requires further research in the field and close collaboration between clinicians and researchers who have special expertise in the complement system.
Collapse
Affiliation(s)
- Erica Daina
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
10
|
Zhou X, Khan S, Huang D, Li L. V-Set and immunoglobulin domain containing (VSIG) proteins as emerging immune checkpoint targets for cancer immunotherapy. Front Immunol 2022; 13:938470. [PMID: 36189222 PMCID: PMC9520664 DOI: 10.3389/fimmu.2022.938470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The development of immune checkpoint inhibitors is becoming a promising approach to fight cancers. Antibodies targeting immune checkpoint proteins such as CTLA-4 and PD-1 can reinvigorate endogenous antitumor T-cell responses and bring durable advantages to several malignancies. However, only a small subset of patients benefit from these checkpoint inhibitors. Identification of new immune checkpoints with the aim of combination blockade of multiple immune inhibitory pathways is becoming necessary to improve efficiency. Recently, several B7 family-related proteins, TIGIT, VSIG4, and VSIG3, which belong to the VSIG family, have attracted substantial attention as coinhibitory receptors during T-cell activation. By interacting with their corresponding ligands, these VSIG proteins inhibit T-cell responses and maintain an immune suppressive microenvironment in tumors. These results indicated that VSIG family members are becoming putative immune checkpoints in cancer immunotherapy. In this review, we summarized the function of each VSIG protein in regulating immune responses and in tumor progression, thus providing an overview of our current understanding of VSIG family members.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sohail Khan
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dabing Huang
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Dabing Huang, ; Lu Li,
| | - Lu Li
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Dabing Huang, ; Lu Li,
| |
Collapse
|
11
|
Intertwined pathways of complement activation command the pathogenesis of lupus nephritis. Transl Res 2022; 245:18-29. [PMID: 35296451 PMCID: PMC9167748 DOI: 10.1016/j.trsl.2022.03.005] [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: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022]
Abstract
The complement system is involved in the origin of autoimmunity and systemic lupus erythematosus. Both genetic deficiency of complement components and excessive activation are involved in primary and secondary renal diseases, including lupus nephritis. Among the pathways, the classical pathway has long been accepted as the main pathway of complement activation in systemic lupus erythematosus. However, more recent studies have shown the contribution of factors B and D which implies the involvement of the alternative pathway. While there is evidence on the role of the lectin pathway in systemic lupus erythematosus, it is yet to be demonstrated whether this pathway is protective or harmful in lupus nephritis. Complement is being explored for the development of disease biomarkers and therapeutic targeting. In the current review we discuss the involvement of complement in lupus nephritis.
Collapse
|
12
|
Antunes Andrade F, Goeldner Eibofner I, Pieczarka C, van Tong H, Sena L, Skare T, Ramos da Rosa Utiyama S, Jose de Messias-Reason I, P Velavan T. Impact of VSIG4 gene polymorphisms on susceptibility and functional status of rheumatoid arthritis. Int J Immunogenet 2021; 48:260-265. [PMID: 33645007 DOI: 10.1111/iji.12533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 02/03/2021] [Accepted: 02/13/2021] [Indexed: 01/31/2023]
Abstract
The complement receptor of the immunoglobulin superfamily (CRIg, encoded by the VSIG4 gene) is a macrophage receptor involved in the clearance of immune complexes and autologous cells. Our results suggest that the VSIG4 rs1044165T allele is a risk factor for severe functional status of rheumatoid arthritis in women, possibly by affecting VSIG4 gene expression.
Collapse
Affiliation(s)
- Fabiana Antunes Andrade
- Laboratory of Molecular Immunopathology, Clinic Hospital, Federal University of Paraná, Curitiba, Brazil.,Department of Medicine, Positive University, Curitiba, Brazil
| | - Isabela Goeldner Eibofner
- Laboratory of Molecular Immunopathology, Clinic Hospital, Federal University of Paraná, Curitiba, Brazil.,Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Cristhine Pieczarka
- Laboratory of Molecular Immunopathology, Clinic Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Hoang van Tong
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Vietnamese-German Center for Medical Research, Hanoi, Vietnam
| | - Leia Sena
- Laboratory of Molecular Immunopathology, Clinic Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Thelma Skare
- Rheumatology Unit, Evangelical Hospital, Curitiba, Brazil
| | | | | | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Vietnamese-German Center for Medical Research, Hanoi, Vietnam.,Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo.,Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
13
|
Vandendriessche S, Cambier S, Proost P, Marques PE. Complement Receptors and Their Role in Leukocyte Recruitment and Phagocytosis. Front Cell Dev Biol 2021; 9:624025. [PMID: 33644062 PMCID: PMC7905230 DOI: 10.3389/fcell.2021.624025] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
The complement system is deeply embedded in our physiology and immunity. Complement activation generates a multitude of molecules that converge simultaneously on the opsonization of a target for phagocytosis and activation of the immune system via soluble anaphylatoxins. This response is used to control microorganisms and to remove dead cells, but also plays a major role in stimulating the adaptive immune response and the regeneration of injured tissues. Many of these effects inherently depend on complement receptors expressed on leukocytes and parenchymal cells, which, by recognizing complement-derived molecules, promote leukocyte recruitment, phagocytosis of microorganisms and clearance of immune complexes. Here, the plethora of information on the role of complement receptors will be reviewed, including an analysis of how this functionally and structurally diverse group of molecules acts jointly to exert the full extent of complement regulation of homeostasis.
Collapse
Affiliation(s)
- Sofie Vandendriessche
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Pedro E Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
14
|
Expanding the Role of Complement Therapies: The Case for Lupus Nephritis. J Clin Med 2021; 10:jcm10040626. [PMID: 33562189 PMCID: PMC7915321 DOI: 10.3390/jcm10040626] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
The complement system is an innate immune surveillance network that provides defense against microorganisms and clearance of immune complexes and cellular debris and bridges innate and adaptive immunity. In the context of autoimmune disease, activation and dysregulation of complement can lead to uncontrolled inflammation and organ damage, especially to the kidney. Systemic lupus erythematosus (SLE) is characterized by loss of tolerance, autoantibody production, and immune complex deposition in tissues including the kidney, with inflammatory consequences. Effective clearance of immune complexes and cellular waste by early complement components protects against the development of lupus nephritis, while uncontrolled activation of complement, especially the alternative pathway, promotes kidney damage in SLE. Therefore, complement plays a dual role in the pathogenesis of lupus nephritis. Improved understanding of the contribution of the various complement pathways to the development of kidney disease in SLE has created an opportunity to target the complement system with novel therapies to improve outcomes in lupus nephritis. In this review, we explore the interactions between complement and the kidney in SLE and their implications for the treatment of lupus nephritis.
Collapse
|
15
|
Alaridhee H, Alharbi A, Saeed Z, Thomas RC, Stover CM. Complement Properdin Determines Disease Activity in MRL/ lpr Mice. ACTA ACUST UNITED AC 2020; 56:medicina56090430. [PMID: 32867176 PMCID: PMC7559332 DOI: 10.3390/medicina56090430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/04/2022]
Abstract
Background and objects: In systemic lupus erythematosus, circulating immune complexes activate complement and, when trapped in renal capillaries, cause glomerulonephritis. Mouse models have been used in the preclinical assessment of targeting complement activation pathways to manage chronic inflammation in lupus. Properdin is the only known positive regulator of complement activation, but its role in the severity of lupus nephritis has not been studied yet. Materials and Methods: Fully characterized properdin-deficient mice were crossed with lupus prone MRL/lpr mice on C57Bl/6 background. Results: Compared to MRL/lpr properdin wildtype mice, MRL/lpr properdin-deficient mice had significantly lower anti-DNA antibody titres, TNFα and BAFF levels in serum. The qualitative glomerulonephritic score was less severe and there was significantly less serum creatinine in MRL/lpr properdin-deficient mice compared to MRL/lpr properdin wildtype littermate mice. Conclusion: Properdin plays a significant role in the severity of lupus overall and specifically in the extent of glomerulonephritis observed in MRL/lpr mice. Because MRL/lpr properdin-deficient mice had lower levels of anti-DNA antibodies, inflammatory mediators and markers of renal impairment, the study implies that properdin could constitute a novel therapy target in lupus disease.
Collapse
Affiliation(s)
- Hasanain Alaridhee
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK; (H.A.); (A.A.); (Z.S.); (R.C.T.)
| | - Azzah Alharbi
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK; (H.A.); (A.A.); (Z.S.); (R.C.T.)
- Department of Medical Microbiology and Parasitology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Zeayd Saeed
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK; (H.A.); (A.A.); (Z.S.); (R.C.T.)
- Department of Nursing, Technical Institute of Samawa, Al-Furat Al-Awsat Technical University, Najaf 54003, Iraq
| | - Róisín C. Thomas
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK; (H.A.); (A.A.); (Z.S.); (R.C.T.)
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Cordula M. Stover
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK; (H.A.); (A.A.); (Z.S.); (R.C.T.)
- Correspondence: ; Tel.: +44-116-252-5032
| |
Collapse
|
16
|
Update on the cellular and molecular aspects of lupus nephritis. Clin Immunol 2020; 216:108445. [PMID: 32344016 DOI: 10.1016/j.clim.2020.108445] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/26/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022]
Abstract
Recent progress has highlighted the involvement of a variety of innate and adaptive immune cells in lupus nephritis. These include activated neutrophils producing extracellular chromatin traps that induce type I interferon production and endothelial injury, metabolically-rewired IL-17-producing T-cells causing tissue inflammation, follicular and extra-follicular helper T-cells promoting the maturation of autoantibody-producing B-cells that may also sustain the formation of germinal centers, and alternatively activated monocytes/macrophages participating in tissue repair and remodeling. The role of resident cells such as podocytes and tubular epithelial cells is increasingly recognized in regulating the local immune responses and determining the kidney function and integrity. These findings are corroborated by advanced, high-throughput genomic studies, which have revealed an unprecedented amount of data highlighting the molecular heterogeneity of immune and non-immune cells implicated in lupus kidney disease. Importantly, this research has led to the discovery of putative pathogenic pathways, enabling the rationale design of novel treatments.
Collapse
|
17
|
Munawara U, Perveen K, Small AG, Putty T, Quach A, Gorgani NN, Hii CS, Abbott CA, Ferrante A. Human Dendritic Cells Express the Complement Receptor Immunoglobulin Which Regulates T Cell Responses. Front Immunol 2019; 10:2892. [PMID: 31921153 PMCID: PMC6914870 DOI: 10.3389/fimmu.2019.02892] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 11/25/2019] [Indexed: 01/09/2023] Open
Abstract
The B7 family-related protein V-set and Ig containing 4 (VSIG4), also known as Z39Ig and Complement Immunoglobulin Receptor (CRIg), is the most recent of the complement receptors to be identified, with substantially distinct properties from the classical complement receptors. The receptor displays both phagocytosis-promoting and anti-inflammatory properties. The receptor has been reported to be exclusively expressed in macrophages. We now present evidence, that CRIg is also expressed in human monocyte-derived dendritic cells (MDDC), including on the cell surface, implicating its role in adaptive immunity. Three CRIg transcripts were detected and by Western blotting analysis both the known Long (L) and Short (S) forms were prominent but we also identified another form running between these two. Cytokines regulated the expression of CRIg on dendritic cells, leading to its up- or down regulation. Furthermore, the steroid dexamethasone markedly upregulated CRIg expression, and in co-culture experiments, the dexamethasone conditioned dendritic cells caused significant inhibition of the phytohemagglutinin-induced and alloantigen-induced T cell proliferation responses. In the alloantigen-induced response the production of IFNγ, TNF-α, IL-13, IL-4, and TGF-β1, were also significantly reduced in cultures with dexamethasone-treated DCs. Under these conditions dexamethasone conditioned DCs did not increase the percentage of regulatory T cells (Treg). Interestingly, this suppression could be overcome by the addition of an anti-CRIg monoclonal antibody to the cultures. Thus, CRIg expression may be a control point in dendritic cell function through which drugs and inflammatory mediators may exert their tolerogenic- or immunogenic-promoting effects on dendritic cells.
Collapse
Affiliation(s)
- Usma Munawara
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,College of Science and Engineering, Flinders University, Bedford Park, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Annabelle G Small
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Trishni Putty
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Alex Quach
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Nick N Gorgani
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Charles S Hii
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Catherine A Abbott
- College of Science and Engineering, Flinders University, Bedford Park, SA, Australia
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA, Australia.,School of Medicine, School of Biological Sciences and The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
18
|
Shi Y, Yao W, Sun L, Li G, Liu H, Ding P, Hu W, Xu H. The new complement inhibitor CRIg/FH ameliorates lupus nephritis in lupus-prone MRL/lpr mice. BMC Nephrol 2019; 20:424. [PMID: 31752725 PMCID: PMC6873683 DOI: 10.1186/s12882-019-1599-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/23/2019] [Indexed: 01/24/2023] Open
Abstract
Backgrounds The aberrant activation of complement system is critically involved in lupus nephropathy. Recent study showed complement C3 inhibitor was effective in the treatment of lupus nephropathy. In this study, we investigate the effect of a novel complement C3 inhibitor, CRIg/FH, in the treatment of lupus nephropathy in MRL/lpr lupus mice. Methods We treated MRL/lpr female mice with a dose escalation of CRIg/FH (10, 5 and 2 mg/kg) by intraperitoneal injection twice weekly since 12 weeks age. In addition, MRL/lpr mice treated with intraperitoneal injection of normal saline or oral prednisone, along with C57BL/6 J healthy mice were maintained to serve as controls. We started 8-h urine collection weekly to screen proteinuria by measuring the levels of urine urea/creatinine. Serum samples was collected at week 16 and 20 to measure levels of urea nitrogen, creatinine, and immunological markers (C3, C4, A-ds-DNA) before the mice were sacrificed at 20 weeks age to collect kidneys for histopathological examinations. Results Overt skin lesions were observed in MRL/lpr mice treated with normal saline, while skin lesion was not observed in CRIg/FH treated MRL/lpr mice. There was no overt proteinuria observed in MRL/lpr mice treated with CRIg/FH. Serum creatinine and BUN levels in MRL/lpr mice was maintained in highest CRIg/FH dose (10 mg/kg twice a week) to be significantly lower than that in prednisone treated MRL/lpr mice at 20 weeks age. In addition, CRIg/FH treatment in MRL/lpr mice results in a significantly elevated serum C3 and C4 levels when compared to prednisone treatment at both 16 and 20 weeks. Furthermore, our study identified that serum level of A-ds-DNA was also significantly lower in CRIg/FH treatment than that in predisone treated MRL/lpr mice. Renal pathology confirmed that kidneys from CRIg/FH treated MRL/lpr mice suffered less from nephritis and complement disposition. Conclusion Our results showed that the complement inhibitor CRIg/FH can protect MRL/lpr mice from lupus nephropathy by preserving renal function and glomerulus complement activation. Our findings support the positive effect of complement inhibitors in the treatment of lupus nephropathy.
Collapse
Affiliation(s)
- Yu Shi
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Wen Yao
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Li Sun
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Guomin Li
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Haimei Liu
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Hong Xu
- Division of Rheumatology, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
19
|
Wang X, Van Lookeren Campagne M, Katschke KJ, Gullipalli D, Miwa T, Ueda Y, Wang Y, Palmer M, Xing G, Song WC. Prevention of Fatal C3 Glomerulopathy by Recombinant Complement Receptor of the Ig Superfamily. J Am Soc Nephrol 2018; 29:2053-2059. [PMID: 29895552 DOI: 10.1681/asn.2018030270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022] Open
Abstract
Background C3 glomerulopathy (C3G) is a life-threatening kidney disease caused by dysregulation of the alternative pathway of complement (AP) activation. No approved specific therapy is available for C3G, although an anti-C5 mAb has been used off-label in some patients with C3G, with mixed results. Thus, there is an unmet medical need to develop other inhibitors of complement for C3G.Methods We used a murine model of lethal C3G to test the potential efficacy of an Fc fusion protein of complement receptor of the Ig superfamily (CRIg-Fc) in the treatment of C3G. CRIg-Fc binds C3b and inhibits C3 and C5 convertases of the AP. Mice with mutations in the factor H and properdin genes (FHm/mP-/-) develop early-onset C3G, with AP consumption, high proteinuria, and lethal crescentic GN.Results Treatment of FHm/mP-/- mice with CRIg-Fc, but not a control IgG, inhibited AP activation and diminished the consumption of plasma C3, factor B, and C5. CRIg-Fc-treated FHm/mP-/- mice also had significantly improved survival and reduced proteinuria, hematuria, BUN, glomerular C3 fragment, C9 and fibrin deposition, and GN pathology scores.Conclusions Therapeutics developed on the basis of the mechanism of action of soluble CRIg may be effective for the treatment of C3G and should be explored clinically.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Departments of Systems Pharmacology and Translational Therapeutics and
| | | | | | | | - Takashi Miwa
- Departments of Systems Pharmacology and Translational Therapeutics and
| | - Yoshiyasu Ueda
- Departments of Systems Pharmacology and Translational Therapeutics and
| | - Yuan Wang
- Departments of Systems Pharmacology and Translational Therapeutics and
| | - Matthew Palmer
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Guolan Xing
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wen-Chao Song
- Departments of Systems Pharmacology and Translational Therapeutics and
| |
Collapse
|
20
|
Nagre N, Cong X, Terrazas C, Pepper I, Schreiber JM, Fu H, Sill JM, Christman JW, Satoskar AR, Zhao X. Inhibition of Macrophage Complement Receptor CRIg by TRIM72 Polarizes Innate Immunity of the Lung. Am J Respir Cell Mol Biol 2018; 58:756-766. [PMID: 29268030 PMCID: PMC6002657 DOI: 10.1165/rcmb.2017-0236oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022] Open
Abstract
The complement system plays a critical role in immune responses against pathogens. However, its identity and regulation in the lung are not fully understood. This study aimed to explore the role of tripartite motif protein (TRIM) 72 in regulating complement receptor (CR) of the Ig superfamily (CRIg) in alveolar macrophage (AM) and innate immunity of the lung. Imaging, absorbance quantification, and flow cytometry were used to evaluate in vitro and in vivo AM phagocytosis with normal, or altered, TRIM72 expression. Pulldown, coimmunoprecipitation, and gradient binding assays were applied to examine TRIM72 and CRIg interaction. A pneumonia model was established by intratracheal injection of Pseudomonas aeruginosa. Mortality, lung bacterial burden, and cytokine levels in BAL fluid and lung tissues were examined. Our data show that TRIM72 inhibited CR-mediated phagocytosis, and release of TRIM72 inhibition led to increased AM phagocytosis. Biochemical assays identified CRIg as a binding partner of TRIM72, and TRIM72 inhibited formation of the CRIg-phagosome. Genetic ablation of TRIM72 led to improved pathogen clearance, reduced cytokine storm, and improved survival in murine models of severe pneumonia, specificity of which was confirmed by adoptive transfer of wild-type or TRIM72KO AMs to AM-depleted TRIM72KO mice. TRIM72 overexpression promoted bacteria-induced NF-κB activation in murine alveolar macrophage cells. Our data revealed a quiescent, noninflammatory bacterial clearance mechanism in the lung via AM CRIg, which is suppressed by TRIM72. In vivo data suggest that targeted suppression of TRIM72 in AM may be an effective measure to treat fatal pulmonary bacterial infections.
Collapse
Affiliation(s)
- Nagaraja Nagre
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Xiaofei Cong
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - César Terrazas
- Departments of Pathology and Microbiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ian Pepper
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - John M. Schreiber
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Hongyun Fu
- Division of Community Health and Research, Pediatrics Department, Eastern Virginia Medical School, Norfolk, Virginia
| | - Joshua M. Sill
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia; and
| | - John W. Christman
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Abhay R. Satoskar
- Departments of Pathology and Microbiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
21
|
van Lookeren Campagne M, Verschoor A. Pathogen clearance and immune adherence "revisited": Immuno-regulatory roles for CRIg. Semin Immunol 2018; 37:4-11. [PMID: 29573978 DOI: 10.1016/j.smim.2018.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 01/30/2023]
Abstract
Rapid elimination of microbes from the bloodstream, along with the ability to mount an adaptive immune response, are essential for optimal host-defense. Kupffer cells are strategically positioned in the liver sinusoids and efficiently capture circulating microbes from the hepatic artery and portal vein, thus preventing bacterial dissemination. In vivo and in vitro studies have probed how complement receptor of the immunoglobulin superfamily (CRIg), also referred to as Z39Ig and V-set and Ig domain-containing 4 (VSIG4), acts as a critical player in pathogen recognition and clearance. While recent data suggested that CRIg may bind bacterial cell wall components directly, the single transmembrane receptor is best known for its interaction with complement C3 opsonization products on the microbial surface. On Kupffer cells, CRIg must capture opsonized microbes against the shear forces of the blood flow. In vivo work reveals how immune adherence (IA), a process in which blood platelets or erythrocytes associate with circulating bacteria, plays a critical role in regulating pathogen capture by CRIg under flow conditions. In addition to its typical innate immune functions, CRIg was shown to directly and indirectly influence adaptive immune responses. Here, we review our current understanding of the diverse roles of CRIg in pathogen elimination, anti-microbial immunity and autoimmunity. In particular, we will explore how, through selective capturing by CRIg, an important balance is achieved between the immunological and clearance functions of liver and spleen.
Collapse
Affiliation(s)
| | - Admar Verschoor
- Institute for Systemic Inflammation Research, Universität zu Lübeck, 23538 Lübeck, Germany.
| |
Collapse
|
22
|
Guo DD, Hu B, Tang HY, Sun YY, Liu B, Tian QM, Bi HS. Proteomic Profiling Analysis Reveals a Link between Experimental Autoimmune Uveitis and Complement Activation in Rats. Scand J Immunol 2017; 85:331-342. [DOI: 10.1111/sji.12539] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/08/2017] [Indexed: 01/01/2023]
Affiliation(s)
- D. D. Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong; Eye Institute of Shandong University of Traditional Chinese Medicine; Jinan China
| | - B. Hu
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine; Jinan China
| | - H. Y. Tang
- The Second Clinical Medical College; Shandong University of Traditional Chinese Medicine; Jinan China
| | - Y. Y. Sun
- The Second Clinical Medical College; Shandong University of Traditional Chinese Medicine; Jinan China
| | - B. Liu
- The Second Clinical Medical College; Shandong University of Traditional Chinese Medicine; Jinan China
| | - Q. M. Tian
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine; Jinan China
| | - H. S. Bi
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong; Eye Institute of Shandong University of Traditional Chinese Medicine; Jinan China
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine; Jinan China
| |
Collapse
|
23
|
A Systems Biology-Based Investigation into the Pharmacological Mechanisms of Sheng-ma-bie-jia-tang Acting on Systemic Lupus Erythematosus by Multi-Level Data Integration. Sci Rep 2015; 5:16401. [PMID: 26560501 PMCID: PMC4642335 DOI: 10.1038/srep16401] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/12/2015] [Indexed: 11/08/2022] Open
Abstract
Sheng-ma-bie-jia-tang (SMBJT) is a Traditional Chinese Medicine (TCM) formula that is widely used for the treatment of Systemic Lupus Erythematosus (SLE) in China. However, molecular mechanism behind this formula remains unknown. Here, we systematically analyzed targets of the ingredients in SMBJT to evaluate its potential molecular mechanism. First, we collected 1,267 targets from our previously published database, the Traditional Chinese Medicine Integrated Database (TCMID). Next, we conducted gene ontology and pathway enrichment analyses for these targets and determined that they were enriched in metabolism (amino acids, fatty acids, etc.) and signaling pathways (chemokines, Toll-like receptors, adipocytokines, etc.). 96 targets, which are known SLE disease proteins, were identified as essential targets and the rest 1,171 targets were defined as common targets of this formula. The essential targets directly interacted with SLE disease proteins. Besides, some common targets also had essential connections to both key targets and SLE disease proteins in enriched signaling pathway, e.g. toll-like receptor signaling pathway. We also found distinct function of essential and common targets in immune system processes. This multi-level approach to deciphering the underlying mechanism of SMBJT treatment of SLE details a new perspective that will further our understanding of TCM formulas.
Collapse
|