1
|
Jeon C, Kim D, Kim KM, Lee SH, Lee JH, Kim SH, Kim JS, Kang YM, Jo S, Kim TH, Son CN. Complement factor H-related protein 5 alleviates joint inflammation and osteoclast differentiation by disrupting RANK-JNK signaling in collagen antibody-induced arthritis mouse model. Cytokine 2024; 184:156790. [PMID: 39461285 DOI: 10.1016/j.cyto.2024.156790] [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: 07/15/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Complement Factor H-Related protein 5 (CFHR5) belongs to the factor H/CFHR family and regulates the complement system by modulating factor H's inhibitory activity against C3b. Despite its known role, the impact of CFHR5 on autoimmune arthritis and its relationship to pathophysiological changes in arthritis and bone loss remain unclear. This study aimed to assess the effect of CFHR5 on aggressive osteoclast activity and arthritis using a murine model of collagen antibody-induced arthritis (CAIA). METHODS The effect of recombinant CFHR5 protein (rCFHR5) on arthritis were evaluated in CAIA. The mice were divided into three group and intraperitoneally treated with rCFHR5, methotrexate (MTX) as positive control or PBS as negative control. In the CAIA mouse model, the rCFHR5-treated group significantly reduced the incidence and clinical arthritis equivalent to the MTX group. Clinical arthritis scores, incidence and body weight were measured, and histological analysis of ankle joints was performed by Hematoxylin and Eosin (H&E) and Safranin O - Fast green (SOFG), Tartrate-resistant acid phosphatase (TRAP) staining and Immunohistochemistry. Moreover, to investigate the rCFHR5 role, we isolated murine osteoclast precursor cells (OCPs) from each group, induced osteoclasts with M-CSF and RANKL, and performed TRAP and F-actin staining. To verify the mechanism, mRNA and protein analyses were performed in OCPs. RESULTS Histological examination of ankle joints revealed substantial reductions in synovial hyperplasia, bone marrow inflammation, bone erosion, cartilage destruction and TRAP-positive cells in the rCFHR5 group compared to the vehicle group. The ankle joints of the rCFHR5 group showed markedly decreased expression of proinflammatory cytokines (TNF-α, IL-1β and IL-6). Mechanically, treatment with rCFHR5 inhibited RANKL-mediated osteoclast differentiation from OCPs and disrupted the RANK-JNK signaling. These findings demonstrate that treatment with rCFHR5 attenuates joint inflammation and reduces osteoclast differentiation, indicating its potential anti-inflammatory effect in autoimmune arthritis models.
Collapse
Affiliation(s)
- Chanhyeok Jeon
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dongju Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Me Kim
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea
| | - Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea
| | - Ji-Hyun Lee
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea
| | - Sang-Hyon Kim
- Division of Rheumatology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Mo Kang
- Preclina Inc, Incheon, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sungsin Jo
- Department of Biology, Soonchunhyang University, Asan, Republic of Korea.
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea; Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea.
| | - Chang-Nam Son
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea.
| |
Collapse
|
2
|
Li L, Ding P, Dong Y, Shen S, Lv X, Yu J, Li L, Chen J, Wang P, Han B, Xu T, Hu W. CG001, a C3b-targeted complement inhibitor, blocks 3 complement pathways: development and preclinical evaluation. Blood Adv 2024; 8:4181-4193. [PMID: 38865712 PMCID: PMC11334799 DOI: 10.1182/bloodadvances.2024012874] [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: 02/07/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024] Open
Abstract
ABSTRACT Excessively activated or dysregulated complement activation may contribute to the pathogenesis of a wide range of human diseases, thus leading to a surge in complement inhibitors. Herein, we developed a human-derived and antibody-like C3b-targeted fusion protein (CRIg-FH-Fc) x2, termed CG001, that could potently block all 3 complement pathways. Complement receptor of the immunoglobulin superfamily (CRIg) and factor H (FH) bind to distinct sites in C3b and synergistically inhibit complement activation. CRIg occupancy in C3b prevents the recruitment of C3 and C5 substrates, whereas FH occupancy in C3b accelerates the decay of C3/C5 convertases and promotes the factor I-mediated degradation and inactivation of C3b. CG001 also showed therapeutic effects in alternative pathways-induced hemolytic mouse and classical pathways-induced mesangial proliferative glomerulonephritis rat models. In the pharmacological/toxicological evaluation in rats and cynomolgus monkeys, CG001 displayed an antibody-like pharmacokinetic profile, a convincing complement inhibitory effect, and no observable toxic effects. Therefore, CG001 holds substantial potential for human clinical studies.
Collapse
Affiliation(s)
- Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | | | - Shupei Shen
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Xinyue Lv
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jie Yu
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Luying Li
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pilin Wang
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Ting Xu
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Small A, Lowe K, Wechalekar MD. Immune checkpoints in rheumatoid arthritis: progress and promise. Front Immunol 2023; 14:1285554. [PMID: 38077329 PMCID: PMC10704353 DOI: 10.3389/fimmu.2023.1285554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent autoimmune inflammatory conditions, and while the mechanisms driving pathogenesis are yet to be completely elucidated, self-reactive T cells and immune checkpoint pathways have a clear role. In this review, we provide an overview of the importance of checkpoint pathways in the T cell response and describe the involvement of these in RA development and progression. We discuss the relationship between immune checkpoint therapy in cancer and autoimmune adverse events, draw parallels with the involvement of immune checkpoints in RA pathobiology, summarise emerging research into some of the lesser-known pathways, and the potential of targeting checkpoint-related pathways in future treatment approaches to RA management.
Collapse
Affiliation(s)
- Annabelle Small
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Katie Lowe
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia
| |
Collapse
|
4
|
Duan H, Abram TG, Cruz AR, Rooijakkers SHM, Geisbrecht BV. New Insights into the Complement Receptor of the Ig Superfamily Obtained from Structural and Functional Studies on Two Mutants. Immunohorizons 2023; 7:806-818. [PMID: 38032267 PMCID: PMC10696418 DOI: 10.4049/immunohorizons.2300064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
The extracellular region of the complement receptor of the Ig superfamily (CRIg) binds to certain C3 cleavage products (C3b, iC3b, C3c) and inhibits the alternative pathway (AP) of complement. In this study, we provide further insight into the CRIg protein and describe two CRIg mutants that lack multiple lysine residues as a means of facilitating chemical modifications of the protein. Structural analyses confirmed preservation of the native CRIg architecture in both mutants. In contrast to earlier reports suggesting that CRIg binds to C3b with an affinity of ∼1 μM, we found that wild-type CRIg binds to C3b and iC3b with affinities <100 nM, but to C3c with an affinity closer to 1 μM. We observed this same trend for both lysine substitution mutants, albeit with an apparent ∼2- to 3-fold loss of affinity when compared with wild-type CRIg. Using flow cytometry, we confirmed binding to C3 fragment-opsonized Staphylococcus aureus cells by each mutant, again with an ∼2- to 3-fold decrease when compared with wild-type. Whereas wild-type CRIg inhibits AP-driven lysis of rabbit erythrocytes with an IC50 of 1.6 μM, we observed an ∼3-fold reduction in inhibition for both mutants. Interestingly, we found that amine-reactive crosslinking of the CRIg mutant containing only a single lysine results in a significant improvement in inhibitory potency across all concentrations examined when compared with the unmodified mutant, but in a manner sensitive to the length of the crosslinker. Collectively, our findings provide new insights into the CRIg protein and suggest an approach for engineering increasingly potent CRIg-based inhibitors of the AP.
Collapse
Affiliation(s)
- Huiquan Duan
- Department of Biochemistry and Molecular Biophysics, Kansas State University; Manhattan, KS
| | - Troy G. Abram
- Department of Biochemistry and Molecular Biophysics, Kansas State University; Manhattan, KS
| | - Ana Rita Cruz
- Department of Medical Microbiology and Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology and Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Brian V. Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University; Manhattan, KS
| |
Collapse
|
5
|
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
|
6
|
Tao S, Yu H, You T, Kong X, Wei X, Zheng Z, Zheng L, Feng Z, Huang B, Zhang X, Chen F, Chen X, Song H, Li J, Chen B, Chen J, Yao Q, Zhao F. A Dual-Targeted Metal-Organic Framework Based Nanoplatform for the Treatment of Rheumatoid Arthritis by Restoring the Macrophage Niche. ACS NANO 2023. [PMID: 37429012 DOI: 10.1021/acsnano.3c03828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Inflammatory infiltration and bone destruction are important pathological features of rheumatoid arthritis (RA), which originate from the disturbed niche of macrophages. Here, we identified a niche-disrupting process in RA: due to overactivation of complement, the barrier function of VSIg4+ lining macrophages is disrupted and mediates inflammatory infiltration within the joint, thereby activating excessive osteoclastogenesis and bone resorption. However, complement antagonists have poor biological applications due to superphysiologic dose requirements and inadequate effects on bone resorption. Therefore, we developed a dual-targeted therapeutic nanoplatform based on the MOF framework to achieve bone-targeted delivery of the complement inhibitor CRIg-CD59 and pH-responsive sustained release. The surface-mineralized zoledronic acid (ZA) of ZIF8@CRIg-CD59@HA@ZA targets the skeletal acidic microenvironment in RA, and the sustained release of CRIg-CD59 can recognize and prevent the complement membrane attack complex (MAC) from forming on the surface of healthy cells. Importantly, ZA can inhibit osteoclast-mediated bone resorption, and CRIg-CD59 can promote the repair of the VSIg4+ lining macrophage barrier to achieve sequential niche remodeling. This combination therapy is expected to treat RA by reversing the core pathological process, circumventing the pitfalls of traditional therapy.
Collapse
Affiliation(s)
- Siyue Tao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027 Zhejiang, China
| | - Tao You
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026 Anhui, China
| | - Xiangxi Kong
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Xiaoan Wei
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Zeyu Zheng
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Lin Zheng
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Zhenhua Feng
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Bao Huang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Xuyang Zhang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Feng Chen
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026 Anhui, China
| | - Xiao Chen
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026 Anhui, China
| | - Haixin Song
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
| | - Jie Li
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, 315100 Zhejiang, China
| | - Binhui Chen
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, 315100 Zhejiang, China
| | - Jian Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
- Department of Orthopedic Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, 325000 Zhejiang, China
| | - Qingqing Yao
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027 Zhejiang, China
| | - Fengdong Zhao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016 Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310016 Zhejiang, China
- Department of Orthopedic Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, 325000 Zhejiang, China
| |
Collapse
|
7
|
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: 7.0] [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
|
8
|
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: 1.0] [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
|
9
|
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: 1.0] [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
|
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
|
Ruocco A, Sirico A, Novelli R, Iannelli S, Van Breda SV, Kyburz D, Hasler P, Aramini A, Amendola PG. The role of C5a-C5aR1 axis in bone pathophysiology: A mini-review. Front Cell Dev Biol 2022; 10:957800. [PMID: 36003145 PMCID: PMC9393612 DOI: 10.3389/fcell.2022.957800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Bone remodeling is a physiological, dynamic process that mainly depends on the functions of 2 cell types: osteoblasts and osteoclasts. Emerging evidence suggests that complement system is crucially involved in the regulation of functions of these cells, especially during inflammatory states. In this context, complement component 5a (C5a), a powerful pro-inflammatory anaphylatoxin that binds the receptor C5aR1, is known to regulate osteoclast formation and osteoblast inflammatory responses, and has thus been proposed as potential therapeutic target for the treatment of inflammatory bone diseases. In this review, we will analyze the role of C5a-C5aR1 axis in bone physiology and pathophysiology, describing its involvement in the pathogenesis of some of the most frequent inflammatory bone diseases such as rheumatoid arthritis, and also in osteoporosis and bone cancer and metastasis. Moreover, we will examine C5aR1-based pharmacological approaches that are available and have been tested so far for the treatment of these conditions. Given the growing interest of the scientific community on osteoimmunology, and the scarcity of data regarding the role of C5a-C5aR1 axis in bone pathophysiology, we will highlight the importance of this axis in mediating the interactions between skeletal and immune systems and its potential use as a therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | - Diego Kyburz
- Departement Biomedizin, University of Basel, Basel, Switzerland
| | - Paul Hasler
- Division of Rheumatology, Kantonsspital Aarau AG, Aarau, Switzerland
| | | | | |
Collapse
|
12
|
Kurowska-Stolarska M, Alivernini S. Synovial tissue macrophages in joint homeostasis, rheumatoid arthritis and disease remission. Nat Rev Rheumatol 2022; 18:384-397. [PMID: 35672464 DOI: 10.1038/s41584-022-00790-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Synovial tissue macrophages (STMs) were principally recognized as having a pro-inflammatory role in rheumatoid arthritis (RA), serving as the main producers of pathogenic tumour necrosis factor (TNF). Recent advances in single-cell omics have facilitated the discovery of distinct STM populations, providing an atlas of discrete phenotypic clusters in the context of healthy and inflamed joints. Interrogation of the functions of distinct STM populations, via ex vivo and experimental mouse models, has re-defined our understanding of STM biology, opening up new opportunities to better understand the pathology of the arthritic joint. These works have identified STM subpopulations that form a protective lining barrier within the synovial membrane and actively participate in the remission of RA. We discuss how distinct functions of STM clusters shape the synovial tissue environment in health, during inflammation and in disease remission, as well as how an increased understanding of STM heterogeneity might aid the prediction of clinical outcomes and inform novel treatments for RA.
Collapse
Affiliation(s)
- Mariola Kurowska-Stolarska
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Stefano Alivernini
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Rome, Italy.
| |
Collapse
|
13
|
Small AG, Perveen K, Putty T, Patel N, Quinn P, Wechalekar MD, Hii CS, Quach A, Ferrante A. Neutrophils Require Activation to Express Functional Cell-Surface Complement Receptor Immunoglobulin. Front Immunol 2022; 13:840510. [PMID: 35317169 PMCID: PMC8934411 DOI: 10.3389/fimmu.2022.840510] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
The phagocytosis-promoting complement receptor, Complement Receptor Immunoglobulin (CRIg), is exclusively expressed on macrophages. It has been demonstrated that expression in macrophages could be modulated by inflammatory mediators, including cytokines. This raised the possibility that a major phagocyte, the neutrophil, may also express CRIg following activation with inflammatory mediators. Here we show that resting peripheral blood neutrophil lysates subjected to protein analysis by Western blot revealed a 35 kDa CRIg isoform, consistent with the expression of CRIg mRNA by RT-PCR. By flow cytometry, CRIg was detected intracellularly and in very minor amounts on the cell surface. Interestingly, expression on the cell surface was significantly increased to functional levels after activation with inflammatory mediators/neutrophil activators; N-Formylmethionine-leucyl-phenylalanine, tumor necrosis factor (TNF), Granulocyte-Macrophage Colony stimulating Factor (GM-CSF), bacterial lipopolysaccharide, leukotriene B4 and phorbol myristate acetate. The increase in expression required p38 MAP kinase and protein kinase C activation, as well as intracellular calcium. Neutrophils which were defective in actin microfilament reorganization due to a mutation in ARPC1B or inhibition of its upstream regulator, Rac2 lose their ability to upregulate CRIg expression. Inhibition of another small GTPase, Rab27a, with pharmacological inhibitors prevented the increase in CRIg expression, suggesting a requirement for the actin cytoskeleton and exocytosis. Engagement of CRIg on TNF-primed neutrophils with an anti-CRIg monoclonal antibody increased the release of superoxide and promoted the activation of p38 but not ERK1/ERK2 or JNK MAP kinases. The TNF-induced increase in killing of Staphylococcus aureus was blocked by the anti-CRIg antibody. Adding to the anti-microbial role of CRIg, it was found that GM-CSF priming lead to the release of neutrophil extracellular traps. Interestingly in contrast to the above mediators the anti-inflammatory cytokine IL-10 caused a decrease in basal expression and GM-CSF induced increase in CRIg expression. The data demonstrate that neutrophils also express CRIg which is regulated by inflammatory mediators and cytokines. The findings show that the neutrophil antimicrobial function involving CRIg requires priming as a means of arming the cell strategically with microbial invasion of tissues and the bloodstream.
Collapse
Affiliation(s)
- Annabelle G. Small
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Rheumatology Department, College of Medicine and Public Health, Flinders Medical Centre, Flinders University, Bedfort Park, SA, Australia
| | - Khalida Perveen
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Trishni Putty
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Nikita Patel
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Patrick Quinn
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Department of Allergy and Immunology, Women’s and Children’s Health Network, North Adelaide, SA, Australia
| | - Mihir D. Wechalekar
- Rheumatology Department, College of Medicine and Public Health, Flinders Medical Centre, Flinders University, Bedfort Park, SA, Australia
| | - Charles S. Hii
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alex Quach
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Antonio Ferrante
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Antonio Ferrante,
| |
Collapse
|
14
|
Reißing J, Lutz P, Frissen M, Ibidapo-Obe O, Reuken PA, Wirtz TH, Stengel S, Quickert S, Rooney M, Große K, Zimmermann HW, Trautwein C, Stallmach A, Bruns T. Immunomodulatory receptor VSIG4 is released during spontaneous bacterial peritonitis and predicts short-term mortality. JHEP Rep 2022; 4:100391. [PMID: 34917912 PMCID: PMC8666561 DOI: 10.1016/j.jhepr.2021.100391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/01/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND & AIMS V-set Ig-domain-containing 4 (VSIG4) is an immunomodulatory macrophage complement receptor modulating innate and adaptive immunity and affecting the resolution of bacterial infections. Given its expression on peritoneal macrophages (PMs), we hypothesised a prognostic role of peritoneal VSIG4 concentrations in patients with spontaneous bacterial peritonitis (SBP). METHODS We isolated PMs from patients with cirrhosis and analysed VSIG4 expression and release by flow cytometry, quantitative real-time PCR, ELISA, and confocal microscopy. We measured soluble VSIG4 concentrations in ascites from 120 patients with SBP and 40 patients without SBP and investigated the association of soluble VSIG4 in ascites with 90-day survival after SBP using Kaplan-Meier statistics, Cox regression, and competing-risks regression analysis. RESULTS VSIG4 expression was high on resting, large PMs, which co-expressed CD206, CD163, and tyrosine-protein kinase Mer (MERTK). VSIG4 gene expression in PMs decreased in patients with SBP and normalised after resolution. During SBP, VSIG4hi PMs were depleted (25% vs. 57%; p <0.001) and soluble VSIG4 in ascites were higher in patients with SBP than in patients without (0.73 vs. 0.35 μg/ml; p <0.0001). PM activation by Toll-like receptor (TLR) agonists or infection with live bacteria in vitro resulted in a loss of surface VSIG4 and the release of soluble VSIG4. Mechanistically, shedding of VSIG4 from PMs was protease-dependent and susceptible to microtubule transport inhibition. Soluble VSIG4 in ascites exceeded serum concentrations and correlated with serum creatinine, model for end-stage liver disease score and C-reactive protein during SBP. Concentrations of 1.0206 μg/ml or higher indicated increased 90-day mortality (hazard ratio 1.70; 95% CI 1.01-2.86; p = 0.046). CONCLUSIONS VSIG4 is released from activated PMs into ascites during SBP. Higher peritoneal VSIG4 levels indicate patients with organ failure and poor prognosis. LAY SUMMARY Patients with liver cirrhosis who develop ascites have an increased risk of infection and mortality. Our study shows that in patients with infected ascites, the complement receptor VSIG4 is released by resident macrophages into the abdominal fluid where it can be measured. Patients with elevated levels of this protein in ascites are at high risk of dying within 90 days.
Collapse
Key Words
- AF, ascitic fluid
- BSA, bovine serum albumin
- Bacterial infection
- Biomarker
- C3, complement component 3
- CCR2, C-C chemokine receptor type 2
- EEA1, early endosome antigen 1
- FCS, foetal calf serum
- FMO, fluorescence minus one
- HLA-DR, human leucocyte antigen-DR isotype
- IMC, isotype-matched control
- INR, international normalised ratio
- LAMP2, lysosome-associated membrane protein 2
- LPS, lipopolysaccharide
- MACS, magnet-activated cell sorting
- MELD, model for end-stage liver disease
- MERTK, tyrosine-protein kinase Mer
- MFI, median fluorescence intensity
- MMP, matrix metalloproteinase
- MOI, multiplicity of infection
- MPLA, monophosphoryl lipid A
- PAMP, pathogen-associated molecular pattern
- PD-L1, programmed cell death 1 ligand 1
- PFA, paraformaldehyde
- PM, peritoneal macrophage
- Prognostic factor
- Risk of death
- SBP, spontaneous bacterial peritonitis
- TAPI-2, tumour necrosis factor protease inhibitor 2
- TLR, Toll-like receptor
- TNF, tumour necrosis factor
- VSIG4, V-set Ig-domain-containing 4
- qRT-PCR, quantitative real-time PCR
- sVSIG4, soluble V-set Ig-domain-containing 4
Collapse
Affiliation(s)
- Johanna Reißing
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lutz
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research, University of Bonn, Bonn, Germany
| | - Mick Frissen
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Oluwatomi Ibidapo-Obe
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp A. Reuken
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Theresa H. Wirtz
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sven Stengel
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Stefanie Quickert
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Michael Rooney
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Karsten Große
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Henning W. Zimmermann
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
15
|
Duan Y, Chu H, Brandl K, Jiang L, Zeng S, Meshgin N, Papachristoforou E, Argemi J, Mendes BG, Wang Y, Su H, Sun W, Llorente C, Hendrikx T, Liu X, Hosseini M, Kisseleva T, Brenner DA, Bataller R, Ramachandran P, Karin M, Fu W, Schnabl B. CRIg on liver macrophages clears pathobionts and protects against alcoholic liver disease. Nat Commun 2021; 12:7172. [PMID: 34887405 PMCID: PMC8660815 DOI: 10.1038/s41467-021-27385-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Complement receptor of immunoglobulin superfamily (CRIg) is expressed on liver macrophages and directly binds complement component C3b or Gram-positive bacteria to mediate phagocytosis. CRIg plays important roles in several immune-mediated diseases, but it is not clear how its pathogen recognition and phagocytic functions maintain homeostasis and prevent disease. We previously associated cytolysin-positive Enterococcus faecalis with severity of alcohol-related liver disease. Here, we demonstrate that CRIg is reduced in liver tissues from patients with alcohol-related liver disease. CRIg-deficient mice developed more severe ethanol-induced liver disease than wild-type mice; disease severity was reduced with loss of toll-like receptor 2. CRIg-deficient mice were less efficient than wild-type mice at clearing Gram-positive bacteria such as Enterococcus faecalis that had translocated from gut to liver. Administration of the soluble extracellular domain CRIg-Ig protein protected mice from ethanol-induced steatohepatitis. Our findings indicate that ethanol impairs hepatic clearance of translocated pathobionts, via decreased hepatic CRIg, which facilitates progression of liver disease.
Collapse
Affiliation(s)
- Yi Duan
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| | - Huikuan Chu
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.33199.310000 0004 0368 7223Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022 Wuhan, China
| | - Katharina Brandl
- grid.266100.30000 0001 2107 4242Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA USA
| | - Lu Jiang
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| | - Suling Zeng
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| | - Nairika Meshgin
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Eleni Papachristoforou
- grid.511172.10000 0004 0613 128XUniversity of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK
| | - Josepmaria Argemi
- grid.412689.00000 0001 0650 7433Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh Liver Research Center, Pittsburgh, PA USA ,grid.5924.a0000000419370271Hepatology Program, Centro de Investigacion Medica Aplicada (CIMA), Liver Unit, Clinica Universidad de Navarra (CUN), Instituto de Investigacion de Navarra (IdisNA), University of Navarra, Pamplona, Spain
| | - Beatriz G. Mendes
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Yanhan Wang
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| | - Hua Su
- grid.266100.30000 0001 2107 4242Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA USA
| | - Weizhong Sun
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Cristina Llorente
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Tim Hendrikx
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Xiao Liu
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Surgery, University of California San Diego, La Jolla, CA USA
| | - Mojgan Hosseini
- grid.266100.30000 0001 2107 4242Department of Pathology, University of California San Diego, La Jolla, CA USA
| | - Tatiana Kisseleva
- grid.266100.30000 0001 2107 4242Department of Surgery, University of California San Diego, La Jolla, CA USA
| | - David A. Brenner
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Ramon Bataller
- grid.412689.00000 0001 0650 7433Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh Liver Research Center, Pittsburgh, PA USA
| | - Prakash Ramachandran
- grid.511172.10000 0004 0613 128XUniversity of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK
| | - Michael Karin
- grid.266100.30000 0001 2107 4242Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA USA
| | - Wenxian Fu
- grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California San Diego, La Jolla, CA USA ,grid.418158.10000 0004 0534 4718Department of Cancer Immunology, Genentech, South San Francisco, CA USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA. .,Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
16
|
He Z, Li J, Gong S, Xing L, Sun Y, Wang J, Li T, Ning N, Zhang L, Yu W, Luo D, Wang H. B7 Family Molecule VSIG4 Regulates Intestinal Anti- Enterohemorrhagic Escherichia coli Immunity by Altering Gut Flora Diversity. Microorganisms 2021; 9:microorganisms9081769. [PMID: 34442848 PMCID: PMC8398431 DOI: 10.3390/microorganisms9081769] [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: 06/19/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
As an essential member of the B7 family, V-set and immunoglobulin domain-containing 4 (VSIG4) is expressed explicitly in tissue-resident macrophages (TRMs) and plays an essential role in maintaining the homeostasis of the environmental immune system. Here, we demonstrate that gene-targeted VSIG4-deficient mice infected with Enterohemorrhagic Escherichia coli (EHEC) display reduced bacterial burden. To reveal the role of VSIG4 in the fight against EHEC infection, we collected mice feces and used high-throughput 16S rRNA gene amplicons to detect changes in the flora. A total of 657330 sequences were sequenced on the PacBio platform, with an average length of 1498 bp. We found that VSIG4 deficiency could alter the gut microbiota by increasing diversity and shifting community composition. In particular, G_Akkermansia and G_Oscillo spiraceae increased significantly. These findings expand upon a prior observation that VSIG4 deficiency reduced EHEC colonization by changing the gut microbiota diversity and shifting community composition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Deyan Luo
- Correspondence: (D.L.); (H.W.); Tel.: +86-10-66948549 (H.W.)
| | - Hui Wang
- Correspondence: (D.L.); (H.W.); Tel.: +86-10-66948549 (H.W.)
| |
Collapse
|
17
|
Halting targeted and collateral damage to red blood cells by the complement system. Semin Immunopathol 2021; 43:799-816. [PMID: 34191092 PMCID: PMC8243056 DOI: 10.1007/s00281-021-00859-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/18/2021] [Indexed: 12/12/2022]
Abstract
The complement system is an important defense mechanism against pathogens; however, in certain pathologies, the system also attacks human cells, such as red blood cells (RBCs). In paroxysmal nocturnal hemoglobinuria (PNH), RBCs lack certain complement regulators which sensitize them to complement-mediated lysis, while in autoimmune hemolytic anemia (AIHA), antibodies against RBCs may initiate complement-mediated hemolysis. In recent years, complement inhibition has improved treatment prospects for these patients, with eculizumab now the standard of care for PNH patients. Current complement inhibitors are however not sufficient for all patients, and they come with high costs, patient burden, and increased infection risk. This review gives an overview of the underlying pathophysiology of complement-mediated hemolysis in PNH and AIHA, the role of therapeutic complement inhibition nowadays, and the high number of complement inhibitors currently under investigation, as for almost every complement protein, an inhibitor is being developed. The focus lies with novel therapeutics that inhibit complement activity specifically in the pathway that causes pathology or those that reduce costs or patient burden through novel administration routes.
Collapse
|
18
|
Garred P, Tenner AJ, Mollnes TE. Therapeutic Targeting of the Complement System: From Rare Diseases to Pandemics. Pharmacol Rev 2021; 73:792-827. [PMID: 33687995 PMCID: PMC7956994 DOI: 10.1124/pharmrev.120.000072] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.
Collapse
Affiliation(s)
- Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Andrea J Tenner
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Tom E Mollnes
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| |
Collapse
|
19
|
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: 1.0] [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
|
20
|
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: 18.7] [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
|
21
|
de Boer ECW, van Mourik AG, Jongerius I. Therapeutic Lessons to be Learned From the Role of Complement Regulators as Double-Edged Sword in Health and Disease. Front Immunol 2020; 11:578069. [PMID: 33362763 PMCID: PMC7758290 DOI: 10.3389/fimmu.2020.578069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
The complement system is an important part of the innate immune system, providing a strong defense against pathogens and removing apoptotic cells and immune complexes. Due to its strength, it is important that healthy human cells are protected against damage induced by the complement system. To be protected from complement, each cell type relies on a specific combination of both soluble and membrane-bound regulators. Their importance is indicated by the amount of pathologies associated with abnormalities in these complement regulators. Here, we will discuss the current knowledge on complement regulatory protein polymorphisms and expression levels together with their link to disease. These diseases often result in red blood cell destruction or occur in the eye, kidney or brain, which are tissues known for aberrant complement activity or regulation. In addition, complement regulators have also been associated with different types of cancer, although their mechanisms here have not been elucidated yet. In most of these pathologies, treatments are limited and do not prevent the complement system from attacking host cells, but rather fight the consequences of the complement-mediated damage, using for example blood transfusions in anemic patients. Currently only few drugs targeting the complement system are used in the clinic. With further demand for therapeutics rising linked to the wide range of complement-mediated disease we should broaden our horizon towards treatments that can actually protect the host cells against complement. Here, we will discuss the latest insights on how complement regulators can benefit therapeutics. Such therapeutics are currently being developed extensively, and can be categorized into full-length complement regulators, engineered complement system regulators and antibodies targeting complement regulators. In conclusion, this review provides an overview of the complement regulatory proteins and their links to disease, together with their potential in the development of novel therapeutics.
Collapse
Affiliation(s)
- Esther C W de Boer
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Anouk G van Mourik
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Ilse Jongerius
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
22
|
Hall BM, Gleiberman AS, Strom E, Krasnov PA, Frescas D, Vujcic S, Leontieva OV, Antoch MP, Kogan V, Koman IE, Zhu Y, Tchkonia T, Kirkland JL, Chernova OB, Gudkov AV. Immune checkpoint protein VSIG4 as a biomarker of aging in murine adipose tissue. Aging Cell 2020; 19:e13219. [PMID: 32856419 PMCID: PMC7576241 DOI: 10.1111/acel.13219] [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: 01/11/2020] [Revised: 05/26/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is recognized as a major source of systemic inflammation with age, driving age-related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA-seq analysis of ATMs from young (8-week-old) and healthy aged (80-week-old) mice. One of the genes identified, V-set immunoglobulin-domain-containing 4 (VSIG4/CRIg), encodes a Mφ-associated complement receptor and B7 family-related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%-52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi-parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor-adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4-positive Mφ contribute to immunosenescence and/or systemic age-related deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga V. Leontieva
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Marina P. Antoch
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Valeria Kogan
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Igor E. Koman
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Yi Zhu
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | | | | | - Andrei V. Gudkov
- Everon Biosciences IncBuffaloNYUSA
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
- Genome Protection IncBuffaloNYUSA
| |
Collapse
|
23
|
Lukácsi S, Mácsik-Valent B, Nagy-Baló Z, Kovács KG, Kliment K, Bajtay Z, Erdei A. Utilization of complement receptors in immune cell-microbe interaction. FEBS Lett 2020; 594:2695-2713. [PMID: 31989596 DOI: 10.1002/1873-3468.13743] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
The complement system is a major humoral component of immunity and is essential for the fast elimination of pathogens invading the body. In addition to its indispensable role in innate immunity, the complement system is also involved in pathogen clearance during the effector phase of adaptive immunity. The fastest way of killing the invader is lysis by the membrane attack complex, which is formed by the terminal components of the complement cascade. Not all pathogens are lysed however and, if opsonized by a variety of molecules, they undergo phagocytosis and disposal inside immune cells. The most important complement-derived opsonins are C1q, the first component of the classical pathway, MBL, the initiator of the lectin pathway and C3-derived activation fragments, including C3b, iC3b and C3d, which all serve as ligands for their corresponding receptors. In this review, we discuss how complement receptors are utilized by various immune cells to tackle invading microbes, or by pathogens to evade host response.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Nagy-Baló
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Kristóf G Kovács
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
24
|
Cheng TH, Yoon SH, Lee P, Dimaculangan D, Vikram Maheshwari A, Zhang M. Knee synovial fluid complement C3-β chain levels correlate with clinical symptoms of knee osteoarthritis. Int J Rheum Dis 2020; 23:569-575. [PMID: 31989759 DOI: 10.1111/1756-185x.13794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/17/2019] [Accepted: 12/29/2019] [Indexed: 12/20/2022]
Abstract
AIM Early research found innate immune factor complement C3 in the synovial fluid (SF) and activated in serum of osteoarthritis (OA) patients. Whether synovial C3 comes from circulation, or is produced locally, is still unknown. It is also unclear whether synovial and circulating C3 is responsible to OA symptoms. A native C3 molecule consists of two chains, C3-α and C3-β. Small fragments breaking down from C3-α chain in serum and SF were reported to be related to OA severity. Little is known if C3-β chain is involved in the pathogenesis. METHOD In this study, we evaluated these important areas by biochemical analyses of C3-α and C3-β chains in both the SF and plasma of OA patients. RESULTS Our results showed that C3-α and C3-β levels in SF did not correlate with those in plasma, suggesting that synovial C3 is independently and locally produced, rather than being "leaked" from circulation. Synovial C3-β but not C3-α levels correlated with pain, other OA symptoms, function in daily living, and sports/recreational activities. Plasma C3-β levels only marginally correlated with pain, and plasma C3-α levels did not correlate with any of these OA symptoms. CONCLUSION We present first-hand evidence that the clinical symptoms of OA are mainly associated with C3 in the local SF rather than systemic circulation, suggesting local factors in the etiopathogenesis. Future local targeted therapies for pain management may be more effective and safer.
Collapse
Affiliation(s)
- Tzu Hsuan Cheng
- Department of Anesthesiology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Seung Ho Yoon
- Department of Anesthesiology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Philip Lee
- Department of Anesthesiology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Dennis Dimaculangan
- Department of Anesthesiology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | | | - Ming Zhang
- Department of Anesthesiology, SUNY Downstate Medical Center, Brooklyn, NY, USA.,Department of Orthopedics, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
25
|
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: 2.0] [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
|
26
|
Autoantibodies as Diagnostic Markers and Mediator of Joint Inflammation in Arthritis. Mediators Inflamm 2019; 2019:6363086. [PMID: 31772505 PMCID: PMC6854956 DOI: 10.1155/2019/6363086] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis is a systemic, polygenic, and multifactorial syndrome characterized by erosive polyarthritis, damage to joint architecture, and presence of autoantibodies against several self-structures in the serum and synovial fluid. These autoantibodies (anticitrullinated protein/peptide antibodies (ACPAs), rheumatoid factors (RF), anticollagen type II antibodies, antiglucose-6 phosphate isomerase antibodies, anticarbamylated protein antibodies, and antiacetylated protein antibodies) have different characteristics, diagnostic/prognostic value, and pathological significance in RA patients. Some of these antibodies are present in the patients' serum several years before the onset of clinical disease. Various genetic and environmental factors are associated with autoantibody production against different autoantigenic targets. Both the activating and inhibitory FcγRs and the activation of different complement cascades contribute to the downstream effector functions in the antibody-mediated disease pathology. Interplay between several molecules (cytokines, chemokines, proteases, and inflammatory mediators) culminates in causing damage to the articular cartilage and bones. In addition, autoantibodies are proven to be useful disease markers for RA, and different diagnostic tools are being developed for early diagnosis of the clinical disease. Recently, a direct link was proposed between the presence of autoantibodies and bone erosion as well as in the induction of pain. In this review, the diagnostic value of autoantibodies, their synthesis and function as a mediator of joint inflammation, and the significance of IgG-Fc glycosylation are discussed.
Collapse
|
27
|
Battin C, De Sousa Linhares A, Paster W, Isenman DE, Wahrmann M, Leitner J, Zlabinger GJ, Steinberger P, Hofer J. Neuropilin-1 Acts as a Receptor for Complement Split Products. Front Immunol 2019; 10:2209. [PMID: 31572401 PMCID: PMC6753332 DOI: 10.3389/fimmu.2019.02209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/02/2019] [Indexed: 01/07/2023] Open
Abstract
Complement split products (CSPs), such as the fragments C4d and C3d, which are generated as a consequence of complement regulatory processes, are established markers for disease activity in autoimmunity or antibody-mediated graft rejection. Since immunoglobulin-like transcript 4 (ILT4) was previously shown to interact with soluble CSPs, but not with CSPs covalently-bound to target surfaces following classical complement activation, the present study aimed to identify novel cellular receptors interacting with covalently-deposited CSPs. By applying an unbiased screening approach using a cDNA mammalian expression library generated from human monocyte-derived dendritic cells and probed with recombinant human C4d, we identified neuropilin-1 (NRP1) as a novel receptor for C4d, C3d, and iC3b. NRP1, a highly conserved type 1 transmembrane protein, plays important roles in the development of the nervous and cardiovascular system as well as in tumorigenesis through interaction with its established binding partners, such as vascular endothelial growth factor (VEGF) and semaphorin 3A (Sema3A). NRP1 is also expressed on immune cells and serves as a marker for murine Tregs. Although NRP1 contains domains homologous to ones found in some complement proteins, it has not been linked to the complement system. We demonstrate that binding of C4d to NRP1 expressing cells was dose-dependent and saturable, and had a KD value of 0.71 μM. Importantly, and in contrast to ILT4, NRP1 interacted with CSPs that were covalently bound to target surfaces in the course of complement activation, therefore representing a classical complement receptor. The binding site of CSPs was mapped to the b1 domain of the coagulation factor V/VIII homology domain of NRP1. Taken together, our results demonstrate a novel role for NRP1 as a receptor for CSPs deposited on surfaces during complement activation. Further work is required to elucidate the functional consequences of the NRP1-CSP interactions in immunity.
Collapse
Affiliation(s)
- Claire Battin
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika De Sousa Linhares
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Paster
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.,Department of Clinical Cell Biology and FACS Core Unit, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - David E Isenman
- Departments of Biochemistry and Immunology, University of Toronto, Toronto, ON, Canada
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Division of Clinical and Experimental Immunology, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes Hofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| |
Collapse
|
28
|
Lofchy LA, Vu VP, Banda NK, Ramirez JR, Smith WJ, Gifford G, Gaikwad H, Scheinman RI, Simberg D. Evaluation of Targeting Efficiency of Joints with Anticollagen II Antibodies. Mol Pharm 2019; 16:2445-2451. [PMID: 31091104 DOI: 10.1021/acs.molpharmaceut.9b00059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diseases of the joints affect over 10% of the world's population, resulting in significant morbidity. There is an unmet need in strategies for specific delivery of therapeutics to the joints. Collagen type II is synthesized by chondrocytes and is mainly restricted to the cartilage and tendons. Arthrogen-CIA is a commercially available anticollagen II antibody cocktail that reacts with 5 different epitopes on human, bovine, and mouse collagen II. Arthrogen has been used for induction of experimental rheumatoid arthritis (RA) in mice because of high complement activation on the cartilage surface. Native collagen II might serve as a useful target for potential delivery of therapeutics to the joint. To evaluate the efficiency and specificity of targeting collagen II, Arthrogen was labeled with near-infrared (NIR) dye IRDye 800 or IRDye 680. Using ex vivo NIR imaging, we demonstrate that Arthrogen efficiently and specifically accumulated in the limb joints regardless of the label dye or injection route (intravenous and subcutaneous). After subcutaneous injection, the mean fluorescence of the hind limb joints was 19 times higher than that of the heart, 8.7 times higher than that of the liver, and 3.7 times higher than that of the kidney. Control mouse IgG did not show appreciable accumulation. Microscopically, the antibody accumulated on the cartilage surface of joints and on endosteal surfaces. A monoclonal antibody against a single epitope of collagen II showed similar binding affinity and elimination half-life, but about three times lower targeting efficiency than Arthrogen in vitro and ex vivo, and about two times lower targeting efficiency in vivo. We suggest that an antibody against multiple epitopes of collagen II could be developed into a highly effective and specific targeting strategy for diseases of the joints or spine.
Collapse
Affiliation(s)
| | | | - Nirmal K Banda
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Joseline Ramos Ramirez
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | | | | | | | | | | |
Collapse
|
29
|
Renevey A, Riniker S. Benchmarking Hybrid Atomistic/Coarse-Grained Schemes for Proteins with an Atomistic Water Layer. J Phys Chem B 2019; 123:3033-3042. [DOI: 10.1021/acs.jpcb.8b12149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Annick Renevey
- Laboratory of Physical Chemistry, ETH Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Sereina Riniker
- Laboratory of Physical Chemistry, ETH Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| |
Collapse
|
30
|
Abstract
Immune responses are controlled by the optimal balance between protective immunity and immune tolerance. T-cell receptor (TCR) signals are modulated by co-signaling molecules, which are divided into co-stimulatory and co-inhibitory molecules. By expression at the appropriate time and location, co-signaling molecules positively and negatively control T-cell differentiation and function. For example, ligation of the CD28 on T cells provides a critical secondary signal along with TCR ligation for naive T-cell activation. In contrast, co-inhibitory signaling by the CD28-B7 family is important to regulate immune homeostasis and host defense, as these signals limit the strength and duration of immune responses to prevent autoimmunity. At the same time, microorganisms or tumor cells can use these pathways to establish an immunosuppressive environment to inhibit the immune responses against themselves. Understanding these co-inhibitory pathways will support the development of new immunotherapy for the treatment of tumors and autoimmune and infectious diseases. Here, we introduce diverse molecules belonging to the members of the CD28-B7 family.
Collapse
|
31
|
Dong X, Zheng Z, Zhai Y, Zheng Y, Ding J, Jiang J, Zhu P. ACPA mediates the interplay between innate and adaptive immunity in rheumatoid arthritis. Autoimmun Rev 2018; 17:845-853. [DOI: 10.1016/j.autrev.2018.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 01/17/2023]
|
32
|
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: 1.5] [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
|
33
|
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: 2.2] [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
|
34
|
Mödinger Y, Löffler B, Huber-Lang M, Ignatius A. Complement involvement in bone homeostasis and bone disorders. Semin Immunol 2018; 37:53-65. [DOI: 10.1016/j.smim.2018.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
|
35
|
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: 4.2] [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
|
36
|
Li J, Diao B, Guo S, Huang X, Yang C, Feng Z, Yan W, Ning Q, Zheng L, Chen Y, Wu Y. VSIG4 inhibits proinflammatory macrophage activation by reprogramming mitochondrial pyruvate metabolism. Nat Commun 2017; 8:1322. [PMID: 29109438 PMCID: PMC5673889 DOI: 10.1038/s41467-017-01327-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/11/2017] [Indexed: 01/24/2023] Open
Abstract
Exacerbation of macrophage-mediated inflammation contributes to pathogenesis of various inflammatory diseases, but the immunometabolic programs underlying regulation of macrophage activation are unclear. Here we show that V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein that is expressed by resting macrophages, inhibits macrophage activation in response to lipopolysaccharide. Vsig4 -/- mice are susceptible to high-fat diet-caused obesity and murine hepatitis virus strain-3 (MHV-3)-induced fulminant hepatitis due to excessive macrophage-dependent inflammation. VSIG4 activates the PI3K/Akt-STAT3 pathway, leading to pyruvate dehydrogenase kinase-2 (PDK2) upregulation and subsequent phosphorylation of pyruvate dehydrogenase, which results in reduction in pyruvate/acetyl-CoA conversion, mitochondrial reactive oxygen species secretion, and macrophage inhibition. Conversely, interruption of Vsig4 or Pdk2 promotes inflammation. Forced expression of Vsig4 in mice ameliorates MHV-3-induced viral fulminant hepatitis. These data show that VSIG4 negatively regulates macrophage activation by reprogramming mitochondrial pyruvate metabolism.
Collapse
Affiliation(s)
- Jialin Li
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Bo Diao
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Sheng Guo
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Chengying Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Zeqing Feng
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China
| | - Weiming Yan
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Ning
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lixin Zheng
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, MD, 20892, USA
| | - Yongwen Chen
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
37
|
Trouw LA, Pickering MC, Blom AM. The complement system as a potential therapeutic target in rheumatic disease. Nat Rev Rheumatol 2017; 13:538-547. [DOI: 10.1038/nrrheum.2017.125] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Abstract
Myeloid cells make extensive use of the complement system in the context of recruitment, phagocytosis, and other effector functions. There are several types of complement receptors on myeloid cells, including G protein-coupled receptors for localizing the source of complement activation, and three sets of type I transmembrane proteins that link complement to phagocytosis: complement receptor 1, having an extracellular domain with tandem complement regulatory repeats; complement receptors 3 and 4, which are integrin family receptors comprising heterodimers of type I transmembrane subunits; and VSIG4, a member of the Ig superfamily. This review will focus on the role of the different classes of complement receptors and how their activities are integrated in the setting of immune tolerance and inflammatory responses.
Collapse
|
39
|
Wen Y, Ouyang Z, Schoonooghe S, Luo S, De Baetselier P, Lu W, Muyldermans S, Raes G, Zheng F. Structural evaluation of a nanobody targeting complement receptor Vsig4 and its cross reactivity. Immunobiology 2016; 222:807-813. [PMID: 27889311 DOI: 10.1016/j.imbio.2016.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/10/2016] [Accepted: 11/12/2016] [Indexed: 11/18/2022]
Abstract
Vsig4 is a recently identified immune regulatory protein related to the B7 family with dual functionality: a negative regulator of T cell activation and a receptor for the complement components C3b and C3c. Here we present a structural evaluation of a nanobody, Nb119, against the extracellular IgV domain protein of both mouse and human recombinant Vsig4, which have a high degree of sequence identity. Although mouse and human Vsig4 bind to Nb119 with a 250 times difference in dissociation constants, the interaction results in a highly identical assembly with a RMSD of 0.4Å. The molecular determinants for Vsig4 recognition and cross reactivity unveiled by the atomic structure of Nb119 in complex with mVsig4 and hVsig4 afford new insights useful for the further optimization of the nanobody for potential use in humans. Additionally, structural analysis of the Vsig4-Nb119 complexes indicates that Nb119 occupies the interface on Vsig4 recognized by the macroglobulin-like domains MG4 and MG5 of C3b. Thus an affinity-improved Nb119 may have the potential to influence the activation of both T cells and complement.
Collapse
Affiliation(s)
- Yurong Wen
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhenlin Ouyang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Steve Schoonooghe
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Inflammation Research Center, Ghent, Belgium
| | - Siyu Luo
- Department of Biochemistry and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Health Science Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Patrick De Baetselier
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Inflammation Research Center, Ghent, Belgium
| | - Wuyuan Lu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Serge Muyldermans
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Inflammation Research Center, Ghent, Belgium
| | - Geert Raes
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Inflammation Research Center, Ghent, Belgium
| | - Fang Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Health Science Center, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
40
|
Kim KH, Choi BK, Kim YH, Han C, Oh HS, Lee DG, Kwon BS. Extracellular stimulation of VSIG4/complement receptor Ig suppresses intracellular bacterial infection by inducing autophagy. Autophagy 2016; 12:1647-59. [PMID: 27440002 DOI: 10.1080/15548627.2016.1196314] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
VSIG4/CRIg (V-set and immunoglobulin domain containing 4) is a transmembrane receptor of the immunoglobulin superfamily that is expressed specifically on macrophages and mature dendritic cells. VSIG4 signaling accelerates phagocytosis of C3-opsonized bacteria, thereby efficiently clearing pathogens within macrophages. We found that VSIG4 signaling triggered by C3-opsonized Listeria (opLM) or by agonistic anti-VSIG4 monoclonal antibody (mAb) induced macrophages to form autophagosomes. VSIG4-induced autophagosomes were selectively colocalized with the intracellular LM while starvation-induced autophagosomes were not. Consistent with these results, the frequency of autophagosomes induced by infection with opLM was lower in VSIG4-deficient bone marrow-derived macrophages (BMDMs) than in WT BMDMs. Furthermore, when VSIG4 molecules were overexpressed in HeLa cells, which are non-macrophage cells, VSIG4 triggering led to efficient uptake of LM, autophagosome formation, and killing of the infected LM. These findings suggest that VSIG4 signaling not only promotes rapid phagocytosis and killing of C3-opsonized intracellular bacteria, as previously reported, but also induces autophagosome formation, eliminating the LM that have escaped from phagosomes. We conclude that VSIG4 signaling provides an anti-immune evasion mechanism that prevents the outgrowth of intracellular bacteria in macrophages.
Collapse
Affiliation(s)
- Kwang H Kim
- a Eutilex , The Catholic University School of Medicine Seoul , Korea
| | - Beom K Choi
- b Cancer Immunology Branch , Division of Cancer Biology, National Cancer Center , Goyang , Korea
| | - Young H Kim
- c Immune Cell Production Unit , Program for Immunotherapeutic Research, National Cancer Center , Goyang , Korea
| | - Chungyong Han
- b Cancer Immunology Branch , Division of Cancer Biology, National Cancer Center , Goyang , Korea
| | - Ho S Oh
- b Cancer Immunology Branch , Division of Cancer Biology, National Cancer Center , Goyang , Korea
| | - Don G Lee
- b Cancer Immunology Branch , Division of Cancer Biology, National Cancer Center , Goyang , Korea
| | - Byoung S Kwon
- a Eutilex , The Catholic University School of Medicine Seoul , Korea.,b Cancer Immunology Branch , Division of Cancer Biology, National Cancer Center , Goyang , Korea.,d Department of Medicine , Tulane University Health Sciences Center , New Orleans , LA , USA
| |
Collapse
|
41
|
Irvine KM, Banh X, Gadd VL, Wojcik KK, Ariffin JK, Jose S, Lukowski S, Baillie GJ, Sweet MJ, Powell EE. CRIg-expressing peritoneal macrophages are associated with disease severity in patients with cirrhosis and ascites. JCI Insight 2016; 1:e86914. [PMID: 27699269 DOI: 10.1172/jci.insight.86914] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Infections are an important cause of morbidity and mortality in patients with decompensated cirrhosis and ascites. Hypothesizing that innate immune dysfunction contributes to susceptibility to infection, we assessed ascitic fluid macrophage phenotype and function. The expression of complement receptor of the immunoglobulin superfamily (CRIg) and CCR2 defined two phenotypically and functionally distinct peritoneal macrophage subpopulations. The proportion of CRIghi macrophages differed between patients and in the same patient over time, and a high proportion of CRIghi macrophages was associated with reduced disease severity (model for end-stage liver disease) score. As compared with CRIglo macrophages, CRIghi macrophages were highly phagocytic and displayed enhanced antimicrobial effector activity. Transcriptional profiling by RNA sequencing and comparison with human macrophage and murine peritoneal macrophage expression signatures highlighted similarities among CRIghi cells, human macrophages, and mouse F4/80hi resident peritoneal macrophages and among CRIglo macrophages, human monocytes, and mouse F4/80lo monocyte-derived peritoneal macrophages. These data suggest that CRIghi and CRIglo macrophages may represent a tissue-resident population and a monocyte-derived population, respectively. In conclusion, ascites fluid macrophage subset distribution and phagocytic capacity is highly variable among patients with chronic liver disease. Regulating the numbers and/or functions of these macrophage populations could provide therapeutic opportunities in cirrhotic patients.
Collapse
Affiliation(s)
| | | | | | | | - Juliana K Ariffin
- Institute for Molecular Bioscience (IMB), and.,IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), and.,IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth E Powell
- School of Medicine.,Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
42
|
Lieberman LA, Mizui M, Nalbandian A, Bossé R, Crispín JC, Tsokos GC. Complement receptor of the immunoglobulin superfamily reduces murine lupus nephritis and cutaneous disease. Clin Immunol 2015; 160:286-91. [PMID: 25988858 DOI: 10.1016/j.clim.2015.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/28/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
Complement activation takes place in autoimmune diseases and accounts for tissue inflammation. Previously, complement inhibition has been considered for the treatment of SLE. Complement receptor of the immunoglobulin superfamily (CRIg) is a selective inhibitor of the alternative pathway of complement and a soluble form reverses established inflammation and bone destruction in experimental autoimmune arthritis. We asked whether specific inhibition of the alternative pathway could inhibit autoimmunity and/or organ damage in lupus-prone mice. Accordingly, we treated lupus-prone MRL/lpr mice with a soluble form of CRIg (CRIg-Fc) and we found that it significantly diminished skin lesions, proteinuria and pyuria, and kidney pathology. Interestingly, serum levels of anti-DNA antibodies were not affected despite the fact that serum complement 3 (C3) levels increased significantly. Immunofluorescent staining of kidney tissues revealed a reduction in staining intensity for C3, IgG, and the macrophage marker Mac-2. Thus our data show that inhibition of the alternative pathway of complement controls skin and kidney inflammation even in the absence of an effect on the production of autoantibodies. We propose that CRIg should be considered for clinical trials in patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- Linda A Lieberman
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masayuki Mizui
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Angèle Nalbandian
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Robin Bossé
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - José C Crispín
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - George C Tsokos
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
43
|
Radichev IA, Maneva-Radicheva LV, Amatya C, Parker C, Ellefson J, Wasserfall C, Atkinson M, Burn P, Savinov AY. Nardilysin-dependent proteolysis of cell-associated VTCN1 (B7-H4) marks type 1 diabetes development. Diabetes 2014; 63:3470-82. [PMID: 24848066 PMCID: PMC4171653 DOI: 10.2337/db14-0213] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T-cell responses directed against insulin-secreting pancreatic β-cells are the key events highlighting type 1 diabetes (T1D). Therefore, a defective control of T-cell activation is thought to underlie T1D development. Recent studies implicated a B7-like negative costimulatory protein, V-set domain-containing T-cell activation inhibitor-1 (VTCN1), as a molecule capable of inhibiting T-cell activation and, potentially, an important constituent in experimental models of T1D. Here, we unravel a general deficiency within the VTCN1 pathway that is shared between diabetes-prone mice and a subset of T1D patients. Gradual loss of membrane-tethered VTCN1 from antigen-presenting cells combined with an increased release of soluble VTCN1 (sVTCN1) occurs in parallel to natural T1D development, potentiating hyperproliferation of diabetogenic T cells. Mechanistically, we demonstrate that the loss of membrane-tethered VTCN1 is linked to proteolytic cleavage mediated by the metalloproteinase nardilysin. The cleaved sVTCN1 fragment was detected at high levels in the peripheral blood of 53% T1D patients compared with only 9% of the healthy subjects. Elevated blood sVTCN1 levels appeared early in the disease progression and correlated with the aggressive pace of disease, highlighting the potential use of sVTCN1 as a new T1D biomarker, and identifying nardilysin as a potential therapeutic target.
Collapse
Affiliation(s)
- Ilian A Radichev
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Lilia V Maneva-Radicheva
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Christina Amatya
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Camille Parker
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Jacob Ellefson
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Clive Wasserfall
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL
| | - Mark Atkinson
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL
| | - Paul Burn
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| | - Alexei Y Savinov
- Sanford Project/Children's Health Research Center at Sanford Research, Sioux Falls, SD Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD
| |
Collapse
|
44
|
Qiao Q, Teng X, Wang N, Lu R, Guo L, Zhang X, Du Y, Wang W, Chen S, Wu Q, He G, Wang Y, Hu W. A novel CRIg-targeted complement inhibitor protects cells from complement damage. FASEB J 2014; 28:4986-99. [PMID: 25114177 DOI: 10.1096/fj.14-258046] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The inappropriate activation of complement may contribute to various immune diseases. The alternative pathway (AP) predominates during complement activation regardless of the initiating pathways. Hence, the main AP regulator factor H (FH) holds great potential as an attractive therapeutic intervention. In addition, complement receptor of the immunoglobulin superfamily (CRIg) has been demonstrated to inhibit AP and, more notably, still specifically binds to C3b/iC3b. We thus developed novel CRIg-targeted complement inhibitors by connecting the functional domains of CRIg and FH, which we termed CRIg-FH and CRIg-L-FH. CRIg-L-FH, slightly more potent than CRIg-FH, considerably inhibited both AP- and also classical pathway (CP)-mediated hemolysis and successfully eliminated the deposition of C3b/iC3b. Kinetic analysis further revealed that the binding affinity constant (KD) of CRIg/FH was in the micromolar range, consistent with its long-lasting binding to complement-attacked cells. CRIg-L-FH efficiently protected aberrant erythrocytes of patients with paroxysmal nocturnal hemoglobinuria (PNH) from AP- and CP-mediated complement damage (IC50 was 22.43 and 64.69 nM, respectively). Moreover, CRIg-L-FH was found to inhibit complement activation induced by the anti-Thy1 antibody in a mesangioproliferative glomerulonephritis (MPGN) rat model. Hence, CRIg-L-FH protects glomerular mesangial cells (GMCs) from complement-mediated injury and proliferative lesions. These findings strongly suggest that CRIg/FH is a potential therapeutic drug candidate for a range of complement-mediated diseases.
Collapse
Affiliation(s)
- Qian Qiao
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Xiaoyan Teng
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Na Wang
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Renquan Lu
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Lin Guo
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Xin Zhang
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Yiqun Du
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Wenjuan Wang
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Suning Chen
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Qian Wu
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Guangsheng He
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Yingwei Wang
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Weiguo Hu
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, China;
| |
Collapse
|
45
|
Zhong C, Wang J, Li B, Xiang H, Ultsch M, Coons M, Wong T, Chiang NY, Clark S, Clark R, Quintana L, Gribling P, Suto E, Barck K, Corpuz R, Yao J, Takkar R, Lee WP, Damico-Beyer LA, Carano RD, Adams C, Kelley RF, Wang W, Ferrara N. Development and Preclinical Characterization of a Humanized Antibody Targeting CXCL12. Clin Cancer Res 2013; 19:4433-45. [DOI: 10.1158/1078-0432.ccr-13-0943] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
46
|
Kim KH, Choi BK, Song KM, Cha KW, Kim YH, Lee H, Han IS, Kwon BS. CRIg signals induce anti-intracellular bacterial phagosome activity in a chloride intracellular channel 3-dependent manner. Eur J Immunol 2013; 43:667-78. [PMID: 23280470 DOI: 10.1002/eji.201242997] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/14/2012] [Accepted: 12/20/2012] [Indexed: 11/10/2022]
Abstract
Macrophages provide a first line of defense against bacterial infection by engulfing and killing invading bacteria, but intracellular bacteria such as Listeria monocytogenes (LM) can survive in macrophages by various mechanisms of evasion. Complement receptor of the immunoglobulin (CRIg), a C3b receptor, binds to C3b on opsonized bacteria and facilitates clearance of the bacteria by promoting their uptake. We found that CRIg signaling induced by agonistic anti-CRIg mAb enhanced the killing of intracellular LM by macrophages, and that this occurred in LM-containing phagosomes. Chloride intra-cellular channel 3 CLIC3, an intracellular chloride channel protein, was essential for CRIg-mediated LM killing by directly interacting with the cytoplasmic domain of CRIg, and the two proteins colocalized on the membranes of LM-containing vacuoles. CLIC3(-/-) mice were as susceptible to LM as CRIg(-/-) mice. These findings identify a mechanism embedded in the process by which macrophages take up opsonized bacteria that prevents the bacteria from evading cell-mediated killing.
Collapse
Affiliation(s)
- Kwang H Kim
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhao M, Shi Y, Zhu X, Chen K, Pan D, Wei C. Molecular Characterization and Expression Analysis of VSIG4 from the Asian Yellow Pond Turtle, Mauremys mutica. Biochem Genet 2012; 50:946-58. [DOI: 10.1007/s10528-012-9534-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 06/13/2012] [Indexed: 10/27/2022]
|
48
|
The expression and anatomical distribution of BTLA and its ligand HVEM in rheumatoid synovium. Inflammation 2012; 35:1102-12. [PMID: 22179929 DOI: 10.1007/s10753-011-9417-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Co-inhibitory signaling from B and T lymphocyte attenuator (BTLA) can suppress lymphocyte activation and maintain peripheral tolerance. However, the expression and anatomical distribution of BTLA and its ligand, herpesvirus entry mediator (HVEM), in rheumatoid arthritis (RA) synovium have not been reported. In this study, we analyzed the expression of HVEM and BTLA in RA synovium by immunohistochemistry, and our results showed that both factors were observed in all four cases of RA samples. At the cellular level, both HVEM and BTLA were found on the cell membrane and in the cytoplasm. Fluorescence dual staining demonstrated that HVEM was chiefly on CD3(+) T cells, CD68(+) macrophages, and to a lesser extent was found on CD31(+) endothelial cells. Similarly, the expression of BTLA was observed on infiltrated CD3(+) T cells and CD68(+) macrophages. The co-expression of HVEM and BTLA with some members of the B7 family in these sections was also analyzed, and the results showed that HVEM antigen was also found on B7-H3(+) capillaries, while it was absent on B7-H1(+), B7-DC(+), B7-H4(+), and Z39Ig(+) cells. Interestingly, BTLA was observed on B7-H1(+), B7-H4(+), and HVEM(+) cells in the synovium. The characteristic expression and distribution of BTLA/HVEM in the synovium indicated that their signaling probably affects the pathogenesis of RA, and a clear understanding of their functional roles may further elucidate the pathogenesis of this disease.
Collapse
|
49
|
Chiang EY, Kolumam G, McCutcheon KM, Young J, Lin Z, Balazs M, Grogan JL. In vivo depletion of lymphotoxin-alpha expressing lymphocytes inhibits xenogeneic graft-versus-host-disease. PLoS One 2012; 7:e33106. [PMID: 22427961 PMCID: PMC3299734 DOI: 10.1371/journal.pone.0033106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/03/2012] [Indexed: 12/23/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a major barrier to successful allogeneic hematopoietic cell transplantation and is largely mediated by activated donor lymphocytes. Lymphotoxin (LT)-α is expressed by subsets of activated T and B cells, and studies in preclinical models demonstrated that targeted depletion of these cells with a mouse anti-LT-α monoclonal antibody (mAb) was efficacious in inhibiting inflammation and autoimmune disease. Here we demonstrate that LT-α is also upregulated on activated human donor lymphocytes in a xenogeneic model of GVHD and targeted depletion of these donor cells ameliorated GVHD. A depleting humanized anti-LT-α mAb, designated MLTA3698A, was generated that specifically binds to LT-α in both the soluble and membrane-bound forms, and elicits antibody-dependent cellular cytotoxicity (ADCC) activity in vitro. Using a human peripheral blood mononuclear cell transplanted SCID (Hu-SCID) mouse model of GVHD, the anti-human LT-α mAb specifically depleted activated LT-expressing human donor T and B cells, resulting in prolonged survival of the mice. A mutation in the Fc region, rendering the mAb incapable of mediating ADCC, abolished all in vitro and in vivo effects. These data support a role for using a depleting anti-LT-α antibody in treating immune diseases such as GVHD and autoimmune diseases.
Collapse
Affiliation(s)
- Eugene Y. Chiang
- Department of Immunology, Genentech Inc., South San Francisco, California, United States of America
| | - Ganesh Kolumam
- Department of Tumor Biology and Angiogenesis, Genentech Inc., South San Francisco, California, United States of America
| | - Krista M. McCutcheon
- Department of Antibody Engineering, Genentech Inc., South San Francisco, California, United States of America
| | - Judy Young
- Department of Assay and Automation Technology, Genentech Inc., South San Francisco, California, United States of America
| | - Zhonghua Lin
- Department of Immunology, Genentech Inc., South San Francisco, California, United States of America
| | - Mercedesz Balazs
- Department of Immunology, Genentech Inc., South San Francisco, California, United States of America
| | - Jane L. Grogan
- Department of Immunology, Genentech Inc., South San Francisco, California, United States of America
| |
Collapse
|
50
|
Early window of diabetes determinism in NOD mice, dependent on the complement receptor CRIg, identified by noninvasive imaging. Nat Immunol 2012; 13:361-8. [PMID: 22366893 PMCID: PMC3309063 DOI: 10.1038/ni.2233] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 01/11/2012] [Indexed: 12/13/2022]
Abstract
All juvenile NOD mice exhibit insulitis, but there is substantial variation in their progression to diabetes. We demonstrate that a patient-validated magnetic-resonance-imaging (MRI) strategy to non-invasively visualize local effects of pancreatic-islet inflammation can predict diabetes onset in NOD mice. MRI signals acquired during a narrow early time-window allowed pre-sorting into disease-progressors and -nonprogressors and an estimate of time-to-diabetes. We exploited this capability to identify novel elements correlated with disease protection, including CRIg (complement receptor of the immunoglobulin superfamily), which marked a subset of macrophages associated with diabetes resistance. Administration of CRIg-Fc depressed MRI signals and diabetes incidence. In addition to identifying regulators of disease progression, this study shows that diabetes is set at an early age in NOD mice.
Collapse
|