1
|
Wang R, Li Y, Jiang Y, Liu X, Feng H, Jiao Z, Li B, Liu C, Shen Y, Chu F, Zhu C, Sun D, Zhang W. Interaction study of the effects of environmental exposure and gene polymorphisms of inflammatory and immune-active factors on chronic obstructive pulmonary disease. Respir Res 2025; 26:5. [PMID: 39780163 PMCID: PMC11707857 DOI: 10.1186/s12931-024-03079-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease, influenced by both environmental and genetic factors. Single nucleotide polymorphism (SNP) in the human genome may influence the risk of developing COPD and the response to treatment. We assessed the effects of gene polymorphism of inflammatory and immune-active factors and gene-environment interaction on risk of COPD in middle-aged and older Chinese individuals. METHODS In this community-based case-control study, 471 patients with COPD and 485 controls aged 40-76 years in Heilongjiang Province, China were included. Face-to-face interviews, lung function tests, and multiplex polymerase chain reaction were used to obtain data. Logistic regression model, generalized multifactor dimensionality reduction and crossover analysis were used to analyse the effects of SNPs, gene-gene interactions, and gene-environment interactions on COPD. RESULTS CRP gene[rs1130864-A allele (OR, 1.77; 95% CI 1.11-2.81); G/A + A/A genotype (OR, 1.75; 95% CI 1.07-2.84)], FCAR gene[rs4806606-G (OR, 0.72; 95% CI 0.53-0.98); rs8112766-G (OR, 0.79; 95% CI 0.64-0.98)] and FCGR2A gene[rs4656308-C (OR, 0.74; 95% CI 0.55-1.00); rs4656309-T (OR, 0.81; 95% CI 0.66-0.99)] are independent influential factors for COPD. Rs1205 [RERI: 0.15 (0.07-1.00)] and rs1130864 [RERI: 2.45 (0.73-4.18)] of CRP gene, rs11084376 [OR: 0.54 (0.29-0.97)] of FCAR gene, rs844 of FCGR2B [SI: 0.30 (0.11-0.77); OR: 0.46 (0.24-0.90)] gene, rs4656308-rs4656309-rs2165088 haplotype [SI: 0.48 (0.26-0.89)] of FCGR2A gene and exposure to smoking index > 200, indoor coal/wood/straw use, and outdoor straw burning play synergistic or antagonistic roles in the development of COPD. CONCLUSIONS Alleles and genotypes of the CRP/FCAR/FCGR2A gene can increase the susceptibility to COPD in the northern Chinese population. For the first time, environmental exposure to the CRP/FCAR/FCGR2A/FCGR2B genes has been shown to have synergistic or antagonistic effects on COPD susceptibility on genotypes or haplotypes.
Collapse
Affiliation(s)
- Rui Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an, 223005, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuanyuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuting Jiang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hongqi Feng
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zhe Jiao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Bingyun Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Chang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuncheng Shen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Fang Chu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Chenpeng Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China.
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China.
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China.
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, 150081, People's Republic of China.
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, 150081, People's Republic of China.
- Joint Key Laboratory of Endemic Diseases (Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
2
|
Dobrowolski C, Lao SM, Kharouf F, Croci PP, Wither J, Gladman DD, Garcia LW, Jauhal A, Touma Z. Lupus nephritis: Biomarkers. Adv Clin Chem 2024; 124:87-122. [PMID: 39818439 DOI: 10.1016/bs.acc.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Lupus nephritis (LN) or renal involvement of systemic lupus erythematosus (SLE), is a common manifestation occurring in at least 50 % of SLE patients. LN remains a significant source of morbidity, often leading to progressive renal dysfunction and is a major cause of death in SLE. Despite these challenges, advances in the understanding of the pathogenesis and genetic underpinnings of LN have led to a commendable expansion in available treatments over the past decade. This chapter provides a foundation for the understanding LN pathogenesis, diagnosis, and epidemiology, and guides the reader through recent advances in biomarkers, genetic susceptibility of this intricate condition.
Collapse
Affiliation(s)
- Chrisanna Dobrowolski
- Division of Rheumatology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, United States
| | - Shu Min Lao
- Division of Rheumatology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, United States
| | - Fadi Kharouf
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Paula Parnizari Croci
- Hospital Manuel Quintela, Facultad de Medicina, Universidad de la República, Uruguay
| | - Joan Wither
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Dafna D Gladman
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Laura Whitall Garcia
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Arenn Jauhal
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Zahi Touma
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada.
| |
Collapse
|
3
|
Knol EF, van Neerven RJJ. IgE versus IgG and IgA: Differential roles of allergen-specific antibodies in sensitization, tolerization, and treatment of allergies. Immunol Rev 2024; 328:314-333. [PMID: 39285523 PMCID: PMC11659938 DOI: 10.1111/imr.13386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
The prevalence of asthma, rhinitis, and food allergies has increased dramatically over the last few decades. This increase originally started in western countries, but is now also evident in many other regions of the world. Given the fact that the increase is so quick, the noted increase cannot be linked to a genetic effect, and many environmental factors have been identified that are associated with increased or reduced prevalence of allergies, like changing dietary habits, increased urbanization, pollution, exposure to microorganisms and LPS, and the farming environment and raw milk consumption. Although the key role of allergen-specific IgE in allergies is well known, the role of allergen-specific IgG and IgA antibodies is less well defined. This review will provide an overview of the functions of allergen-specific IgE in allergy, the role of allergen-specific antibodies (IgG (4) and IgA) in allergen immunotherapy (AIT), the possibility to use allergen-specific antibodies for treatment of ongoing allergies, and the potential role of allergen-specific antibodies in tolerance induction to allergens in a preventive setting. In the last, more speculative, section we will present novel hypotheses on the potential role of allergen-specific non-IgE antibodies in allergies by directing antigen presentation, Th2 development, and innate immune training.
Collapse
Affiliation(s)
- E. F. Knol
- Department of Dermatology/AllergologyUMC UtrechtUtrechtthe Netherlands
| | - R. J. J. van Neerven
- Cell Biology and ImmunologyWageningen University & ResearchWageningenthe Netherlands
| |
Collapse
|
4
|
Miao J, Wang HM, Pan XH, Gong Z, Gao XM, Gong FY. hFcγRIIa: a double-edged sword in osteoclastogenesis and bone balance in transgenic mice. Front Immunol 2024; 15:1425670. [PMID: 39281679 PMCID: PMC11392756 DOI: 10.3389/fimmu.2024.1425670] [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: 04/30/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease accompanied by local and systemic bone loss. FcγRs, especially FcγRIIa (hFcγRIIa), have been implicated in the pathogenesis of RA. However, the contribution of hFcγRIIa to bone loss has not been fully elucidated. In the present study, we demonstrated the double-edged sword role of hFcγRIIa on osteoclast differentiation through investigations involving hFcγRIIa-transgenic (hFcγRIIa-Tg) mice. Our findings reveal that hFcγRIIa-Tg mice, previously shown to exhibit heightened susceptibility to collagen-induced arthritis (CIA), displayed increased osteoporosis during CIA or at advanced ages (40 weeks), accompanied by heightened in vivo osteoclast differentiation. Notably, bone marrow cells from hFcγRIIa-Tg mice exhibited enhanced efficiency in differentiating into osteoclasts and bone resorption in vitro compared to wild-type mice when stimulated with receptor activators of NF-κB ligand (RANKL). Additionally, hFcγRIIa-Tg mice exhibited augmented sensitivity to RANKL-induced bone loss in vivo, highlighting the osteoclast-promoting role of hFcγRIIa. Mechanistically, bone marrow cells from hFcγRIIa-Tg mice displayed heightened Syk self-activation, leading to mTOR-pS6 pathway activation, thereby promoting RANKL-driven osteoclast differentiation. Intriguingly, while hFcγRIIa crosslinking hindered RANKL-induced osteoclast differentiation, it activated the kinase cAbl, subsequently triggering STAT5 activation and inhibiting the expression of osteoclast-associated genes. This study provides novel insights into hFcγRIIa-mediated osteoclast biology, suggesting promising therapeutic targets for managing bone remodeling disorders.
Collapse
Affiliation(s)
| | | | | | | | - Xiao-Ming Gao
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Fang-Yuan Gong
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Gil Gonzalez L, Won KD, Tawhidi Z, Cummins E, Cruz-Leal Y, Tundidor Cabado Y, Sachs UJ, Norris PAA, Shan Y, Bhakta V, Li J, Samudio I, Silva-Moreno B, Cerna-Portillo L, Pavon Oro A, Bergqvist P, Chan P, Moorehead A, Sholzberg M, Sheffield WP, Lazarus AH. Human Fc gamma receptor IIIA blockade inhibits platelet destruction in a humanized murine model of ITP. Blood Adv 2024; 8:1869-1879. [PMID: 38330193 PMCID: PMC11007428 DOI: 10.1182/bloodadvances.2023012155] [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: 11/13/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
ABSTRACT Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on macrophages can trigger phagocytosis of IgG-sensitized platelets, and prior pilot studies observed blockade of FcγRIIIA increased platelet counts in patients with ITP. Unfortunately, although blockade of FcγRIIIA in patients with ITP increased platelet counts, its engagement by the blocking antibody drove serious adverse inflammatory reactions. These adverse events were postulated to originate from the antibody's Fc and/or bivalent nature. The blockade of human FcγRIIIA in vivo with a monovalent construct lacking an active Fc region has not yet been achieved. To effectively block FcγRIIIA in vivo, we developed a high affinity monovalent single-chain variable fragment (scFv) that can bind and block human FcγRIIIA. This scFv (17C02) was expressed in 3 formats: a monovalent fusion protein with albumin, a 1-armed human IgG1 antibody, and a standard bivalent mouse (IgG2a) antibody. Both monovalent formats were effective in preventing phagocytosis of ITP serum-sensitized human platelets. In vivo studies using FcγR-humanized mice demonstrated that both monovalent therapeutics were also able to increase platelet counts. The monovalent albumin fusion protein did not have adverse event activity as assessed by changes in body temperature, whereas the 1-armed antibody induced some changes in body temperature even though the Fc region function was impaired by the Leu234Ala and Leu235Ala mutations. These data demonstrate that monovalent blockade of human FcγRIIIA in vivo can potentially be a therapeutic strategy for patients with ITP.
Collapse
Affiliation(s)
- Lazaro Gil Gonzalez
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Kevin D. Won
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zoya Tawhidi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - Yoelys Cruz-Leal
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Yaima Tundidor Cabado
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Ulrich J. Sachs
- Institute for Clinical Immunology, Transfusion Medicine, and Haemostasis, Justus Liebig University, Giessen, Germany
- Department of Thrombosis and Haemostasis, Giessen University Hospital, Giessen, Germany
| | - Peter A. A. Norris
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Yuexin Shan
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Varsha Bhakta
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Janessa Li
- adMare BioInnovations, Vancouver, BC, Canada
| | | | | | | | - Alequis Pavon Oro
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | | | | | - Amy Moorehead
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Michelle Sholzberg
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - William P. Sheffield
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Alan H. Lazarus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| |
Collapse
|
6
|
McCord K, Wang C, Anhalt M, Poon WW, Gavin AL, Wu P, Macauley MS. Dissecting the Ability of Siglecs To Antagonize Fcγ Receptors. ACS CENTRAL SCIENCE 2024; 10:315-330. [PMID: 38435516 PMCID: PMC10906256 DOI: 10.1021/acscentsci.3c00969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 03/05/2024]
Abstract
Fcγ receptors (FcγRs) play key roles in the effector function of IgG, but their inappropriate activation plays a role in several disease etiologies. Therefore, it is critical to better understand how FcγRs are regulated. Numerous studies suggest that sialic acid-binding immunoglobulin-type lectins (Siglecs), a family of immunomodulatory receptors, modulate FcγR activity; however, it is unclear of the circumstances in which Siglecs can antagonize FcγRs and which Siglecs have this ability. Using liposomes displaying selective ligands to coengage FcγRs with a specific Siglec, we explore the ability of Siglec-3, Siglec-5, Siglec-7, and Siglec-9 to antagonize signaling downstream of FcγRs. We demonstrate that Siglec-3 and Siglec-9 can fully inhibit FcγR activation in U937 cells when coengaged with FcγRs. Cells expressing Siglec mutants reveal differential roles for the immunomodulatory tyrosine-based inhibitory motif (ITIM) and immunomodulatory tyrosine-based switch motif (ITSM) in this inhibition. Imaging flow cytometry enabled visualization of SHP-1 recruitment to Siglec-3 in an ITIM-dependent manner, while SHP-2 recruitment is more ITSM-dependent. Conversely, both cytosolic motifs of Siglec-9 contribute to SHP-1/2 recruitment. Siglec-7 poorly antagonizes FcγR activation for two reasons: masking by cis ligands and differences in its ITIM and ITSM. A chimera of the Siglec-3 extracellular domains and Siglec-5 cytosolic tail strongly inhibits FcγR when coengaged, providing evidence that Siglec-5 is more like Siglec-3 and Siglec-9 in its ability to antagonize FcγRs. Additionally, Siglec-3 and Siglec-9 inhibited FcγRs when coengaged by cells displaying ligands for both the Siglec and FcγRs. These results suggest a role for Siglecs in mediating FcγR inhibition in the context of an immunological synapse, which has important relevance to the effectiveness of immunotherapies.
Collapse
Affiliation(s)
- Kelli
A. McCord
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Chao Wang
- Department
of Molecular Medicine, Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Mirjam Anhalt
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Wayne W. Poon
- Institute
for Memory Impairments and Neurological Disorders, University of California, Irvine, California 92617, United States
| | - Amanda L. Gavin
- Department
of Immunology and Microbiology, Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Peng Wu
- Department
of Molecular Medicine, Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Matthew S. Macauley
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
7
|
Conley HE, He MM, Easterhoff D, Kirshner HF, Cocklin SL, Meyer J, Hoxie T, Berry M, Bradley T, Tolbert WD, Pazgier M, Tomaras GD, Schmitz JE, Moody MA, Wiehe K, Pollara J. Defining genetic diversity of rhesus macaque Fcγ receptors with long-read RNA sequencing. Front Immunol 2024; 14:1306292. [PMID: 38264644 PMCID: PMC10803544 DOI: 10.3389/fimmu.2023.1306292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Fcγ receptors (FcγRs) are membrane-bound glycoproteins that bind to the fragment crystallizable (Fc) constant regions of IgG antibodies. Interactions between IgG immune complexes and FcγRs can initiate signal transduction that mediates important components of the immune response including activation of immune cells for clearance of opsonized pathogens or infected host cells. In humans, many studies have identified associations between FcγR gene polymorphisms and risk of infection, or progression of disease, suggesting a gene-level impact on FcγR-dependent immune responses. Rhesus macaques are an important translational model for most human health interventions, yet little is known about the breadth of rhesus macaque FcγR genetic diversity. This lack of knowledge prevents evaluation of the impact of FcγR polymorphisms on outcomes of preclinical studies performed in rhesus macaques. In this study we used long-read RNA sequencing to define the genetic diversity of FcγRs in 206 Indian-origin Rhesus macaques, Macaca mulatta. We describe the frequency of single nucleotide polymorphisms, insertions, deletions, frame-shift mutations, and isoforms. We also index the identified diversity using predicted and known rhesus macaque FcγR and Fc-FcγR structures. Future studies that define the functional significance of this genetic diversity will facilitate a better understanding of the correlation between human and macaque FcγR biology that is needed for effective translation of studies with antibody-mediated outcomes performed in rhesus macaques.
Collapse
Affiliation(s)
- Haleigh E. Conley
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Max M. He
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - David Easterhoff
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Hélène Fradin Kirshner
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Sarah L. Cocklin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jacob Meyer
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Taylor Hoxie
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Todd Bradley
- Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, MO, United States
| | - William D. Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgia D. Tomaras
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Joern E. Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Michael Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Justin Pollara
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
8
|
Abdeldaim DT, Schindowski K. Fc-Engineered Therapeutic Antibodies: Recent Advances and Future Directions. Pharmaceutics 2023; 15:2402. [PMID: 37896162 PMCID: PMC10610324 DOI: 10.3390/pharmaceutics15102402] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Monoclonal therapeutic antibodies have revolutionized the treatment of cancer and other diseases. Fc engineering aims to enhance the effector functions or half-life of therapeutic antibodies by modifying their Fc regions. Recent advances in the Fc engineering of modern therapeutic antibodies can be considered the next generation of antibody therapy. Various strategies are employed, including altering glycosylation patterns via glycoengineering and introducing mutations to the Fc region, thereby enhancing Fc receptor or complement interactions. Further, Fc engineering strategies enable the generation of bispecific IgG-based heterodimeric antibodies. As Fc engineering techniques continue to evolve, an expanding portfolio of Fc-engineered antibodies is advancing through clinical development, with several already approved for medical use. Despite the plethora of Fc-based mutations that have been analyzed in in vitro and in vivo models, we focus here in this review on the relevant Fc engineering strategies of approved therapeutic antibodies to finetune effector functions, to modify half-life and to stabilize asymmetric bispecific IgGs.
Collapse
Affiliation(s)
- Dalia T. Abdeldaim
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
| |
Collapse
|
9
|
Laassili C, Ben El Hend F, Benzidane R, Oumeslakht L, Aziz AI, El Fatimy R, Bensussan A, Ben Mkaddem S. Fc receptors act as innate immune receptors during infection? Front Immunol 2023; 14:1188497. [PMID: 37564652 PMCID: PMC10410254 DOI: 10.3389/fimmu.2023.1188497] [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: 03/17/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
Innate immunity constitutes the first nonspecific immunological line of defense against infection. In this response, a variety of mechanisms are activated: the complement system, phagocytosis, and the inflammatory response. Then, adaptive immunity is activated. Major opsonization mediators during infections are immunoglobulins (Igs), the function of which is mediated through Fc receptors (FcRs). However, in addition to their role in adaptive immunity, FcRs have been shown to play a role in innate immunity by interacting directly with bacteria in the absence of their natural ligands (Igs). Additionally, it has been hypothesized that during the early phase of bacterial infection, FcRs play a protective role via innate immune functions mediated through direct recognition of bacteria, and as the infection progresses to later phases, FcRs exhibit their established function as receptors in adaptive immunity. This review provides detailed insight into the potential role of FcRs as innate immune mediators of the host defense against bacterial infection independent of opsonins.
Collapse
Affiliation(s)
- Chaimaa Laassili
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Fatiha Ben El Hend
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Riad Benzidane
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Loubna Oumeslakht
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Abdel-Ilah Aziz
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Rachid El Fatimy
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Armand Bensussan
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
- INSERM U976, Université de Paris, Hôpital Saint Louis, Paris, France
- Institut Jean Godinot, Centre de Lutte Contre le Cancer, Reims, France
| | - Sanae Ben Mkaddem
- Faculty of Medical Sciences, Mohammed VI Polytechnic University, Benguerir, Morocco
| |
Collapse
|
10
|
Abyadeh M, Yadav VK, Kaya A. Common molecular signatures between coronavirus infection and Alzheimer's disease reveal targets for drug development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.544970. [PMID: 37398415 PMCID: PMC10312734 DOI: 10.1101/2023.06.14.544970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Cognitive decline has been reported as a common consequence of COVID-19, and studies have suggested a link between COVID-19 infection and Alzheimer's disease (AD). However, the molecular mechanisms underlying this association remain unclear. To shed light on this link, we conducted an integrated genomic analysis using a novel Robust Rank Aggregation method to identify common transcriptional signatures of the frontal cortex, a critical area for cognitive function, between individuals with AD and COVID-19. We then performed various analyses, including the KEGG pathway, GO ontology, protein-protein interaction, hub gene, gene-miRNA, and gene-transcription factor interaction analyses to identify molecular components of biological pathways that are associated with AD in the brain also show similar changes in severe COVID-19. Our findings revealed the molecular mechanisms underpinning the association between COVID-19 infection and AD development and identified several genes, miRNAs, and TFs that may be targeted for therapeutic purposes. However, further research is needed to investigate the diagnostic and therapeutic applications of these findings.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284 USA
| | - Vijay K. Yadav
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284 USA
| |
Collapse
|
11
|
Typiak M, Rękawiecki B, Rębała K, Dubaniewicz A. Comparative Analysis of FCGR Gene Polymorphism in Pulmonary Sarcoidosis and Tuberculosis. Cells 2023; 12:cells12091221. [PMID: 37174624 PMCID: PMC10177102 DOI: 10.3390/cells12091221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
The clinical outcome of sarcoidosis (SA) is very similar to tuberculosis (TB); however, they are treated differently and should not be confused. In search for their biomarkers, we have previously revealed changes in the phagocytic activity of monocytes in sarcoidosis and tuberculosis. On these monocytes we found a higher expression of receptors for the Fc fragment of immunoglobulin G (FcγR) in SA and TB patients vs. healthy controls. FcγRs are responsible for the binding of immune complexes (ICs) to initiate an (auto)immune response and for ICs clearance. Surprisingly, our SA patients had a high blood level of ICs, despite the abundant presence of FcγRs. It pointed to FcγR disfunction, presumably caused by the polymorphism of their (FCGR) genes. Therefore, we present here an analysis of the occurrence of FCGR2A, FCGR2B, FCGR2C, FCGR3A and FCGR3B variants in Caucasian SA and TB patients, and healthy individuals with the use of polymerase chain reaction (PCR) and real-time PCR. The presented data point to a possibility of supporting the differential diagnosis of SA and TB by analyzing FCGR2C, FCGR3A and FCGR3B polymorphism, while for severe stages of SA also by studying FCGR2A variants. Additionally, the genotyping of FCGR2A and FCGR3B might serve as a marker of SA progression.
Collapse
Affiliation(s)
- Marlena Typiak
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | | | - Krzysztof Rębała
- Department of Forensic Medicine, Medical University of Gdansk, 80-204 Gdansk, Poland
| | - Anna Dubaniewicz
- Department of Pulmonology, Medical University of Gdansk, 80-214 Gdansk, Poland
| |
Collapse
|
12
|
Ines M, Myriam M, Aicha BT, Selma B, Leila R, Rawdha T, Olfa S, Yousr G, Mohamed E, Ahmed L, Wafa H, Leila A, Imen S. Impact of FCGR2A R131H, FCGR3A F158V and FCGR3B NA1/NA2 polymorphisms on response to fc-containing TNF inhibitors in Tunisian rheumatoid arthritis patients. Drug Metab Pers Ther 2023:dmdi-2022-0176. [PMID: 36919284 DOI: 10.1515/dmpt-2022-0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/24/2022] [Indexed: 03/16/2023]
Abstract
OBJECTIVES Single nucleotid polymorphisms (SNPs) of Fc-gamma receptors (FcgRs), by inducing a variation of their affinity to the Fc-region of immunoglobulins, might influence the efficacy of Fc-containing biologics prescribed in rheumatoid arthritis (RA). Our aim was to investigate associations of FCGR2A, FCGR3A and FCGR3B SNPs with TNF-inhibitors (TNFi)' response in Tunisian RA patients. METHODS A cross-sectional, observational and analytic multicentric cohort study was conducted in a group of 47 Tunisian RA patients treated with (etanercept [ETA], adalimumab [ADL] and infliximab [IFX]). Treatment outcome was evaluated after 6 months. R131H-FCGR2A, F158V-FCGR3A and NA1/NA2-FCGR3B SNPs were genotyped. RESULTS The analytic study including all types of TNFi showed that FCGR3A-F/F low-affinity receptor was associated with a greater decrease of DAS28, while FCGR3B-NA1/NA1 high-affinity receptor was associated with a lower decrease of DAS28 in ADL group. Furthermore, both of high affinity receptors FCGR3B-NA1/NA1 and FCGR3A-V/V were more prevalent in non-responders to ADL, according to EULAR criteria. CONCLUSIONS Identifying reliable biomarkers of response to biologics in RA is necessary to improve responsiveness, preserve joints' functions and structure, and reduce treatment's cost. Our study showed that FCGR3A and FCGR3B polymorphisms might have an impact on TNFis' response in RA Tunisian patients since bad response was more frequent in homozygous carriers of high affinity alleles FCGR3A-V and FCGR3B-NA1.
Collapse
Affiliation(s)
- Mahmoud Ines
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Moalla Myriam
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Ben Tekaya Aicha
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Bouden Selma
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Rouached Leila
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Tekaya Rawdha
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Saidane Olfa
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Gorji Yousr
- Laboratory of Research in Immunology, Renal Transplantation and Immunopathology (LR03SP01), Charles Nicolle Hospital of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Elleuch Mohamed
- Rheumatology Department, La Rabta Hospital of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Laatar Ahmed
- Rheumatology Department, Monji Slim Hospital of La Marsa, Tunis El Manar University, Tunis, Tunisia
| | - Hamdi Wafa
- Rheumatology Department, Kassab Institute of Orthopedics of La Mannouba, Tunis El Manar University, Tunis, Tunisia
| | - Abdelmoula Leila
- Rheumatology Department, Charles Nicolle Hospital, Tunis El Manar University, Tunis, Tunisia
| | - Sfar Imen
- Laboratory of Research in Immunology, Renal Transplantation and Immunopathology (LR03SP01), Charles Nicolle Hospital of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
13
|
Abyadeh M, Yadav VK, Kaya A. Common Molecular Signatures Between Coronavirus Infection and Alzheimer's Disease Reveal Targets for Drug Development. J Alzheimers Dis 2023; 95:995-1011. [PMID: 37638446 DOI: 10.3233/jad-230684] [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] [Indexed: 08/29/2023]
Abstract
BACKGROUND Cognitive decline is a common consequence of COVID-19, and studies suggest a link between COVID-19 and Alzheimer's disease (AD). However, the molecular mechanisms underlying this association remain unclear. OBJECTIVE To understand the potential molecular mechanisms underlying the association between COVID-19 and AD development, and identify the potential genetic targets for pharmaceutical approaches to reduce the risk or delay the development of COVID-19-related neurological pathologies. METHODS We analyzed transcriptome datasets of 638 brain samples using a novel Robust Rank Aggregation method, followed by functional enrichment, protein-protein, hub genes, gene-miRNA, and gene-transcription factor (TF) interaction analyses to identify molecular markers altered in AD and COVID-19 infected brains. RESULTS Our analyses of frontal cortex from COVID-19 and AD patients identified commonly altered genes, miRNAs and TFs. Functional enrichment and hub gene analysis of these molecular changes revealed commonly altered pathways, including downregulation of the cyclic adenosine monophosphate (cAMP) signaling and taurine and hypotaurine metabolism, alongside upregulation of neuroinflammatory pathways. Furthermore, gene-miRNA and gene-TF network analyses provided potential up- and downstream regulators of identified pathways. CONCLUSION We found that downregulation of cAMP signaling pathway, taurine metabolisms, and upregulation of neuroinflammatory related pathways are commonly altered in AD and COVID-19 pathogenesis, and may make COVID-19 patients more susceptible to cognitive decline and AD. We also identified genetic targets, regulating these pathways that can be targeted pharmaceutically to reduce the risk or delay the development of COVID-19-related neurological pathologies and AD.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Biology, Virginia Common wealth University, Richmond, VA, USA
| | - Vijay K Yadav
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Alaattin Kaya
- Department of Biology, Virginia Common wealth University, Richmond, VA, USA
| |
Collapse
|
14
|
Cai H, Kakiuchi-Kiyota S, Hendricks R, Zhong S, Liu L, Adedeji AO, Chan P, Schutten MM, Kamath AV, Ovacik MA. Nonclinical Pharmacokinetics, Pharmacodynamics, and Translational Model of RO7297089, A Novel Anti-BCMA/CD16A Bispecific Tetravalent Antibody for the Treatment of Multiple Myeloma. AAPS J 2022; 24:100. [PMID: 36127472 DOI: 10.1208/s12248-022-00744-8] [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: 05/27/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023] Open
Abstract
RO7297089, an anti-B-cell maturation antigen (BCMA)/CD16A bispecific tetravalent antibody, is being developed as a multiple myeloma (MM) therapeutic. This study characterized nonclinical pharmacokinetics (PK), pharmacodynamics (PD), soluble BCMA (sBCMA), and soluble CD16 (sCD16) changes following administration of RO7297089 to support clinical trials. Unbound and total RO7297089 concentrations were measured in cynomolgus monkeys. RO7297089 exhibited a bi-phasic systemic concentration-time profile, similar to a typical human immunoglobulin 1 antibody. Target engagement by RO7297089 led to a robust increase (~100-fold) in total systemic sBCMA levels and relatively mild increase (~2-fold) in total sCD16 levels. To describe the relationship of nonclinical PK/PD data, we developed a target-mediated drug disposition (TMDD) model that includes the systemic target engagement of membrane BCMA (mBCMA), sBCMA, membrane CD16 (mCD16), and sCD16. We then used this model to simulate the PK/PD relationship of RO7297089 in MM patients by translating relevant PK parameters and target levels, based on the literature and newly generated data such as baseline sCD16A levels. Our model suggested that the impact of TMDD on RO7297089 exposure may be more significant in MM patients due to significantly higher expression levels of both mBCMA and sBCMA compared to healthy cynomolgus monkeys. Based on model simulations, we propose more frequent dosing of RO7297089 compared to regular monthly frequency in the clinic at the beginning of treatment to ensure sustained target engagement. This study demonstrates a translational research strategy for collecting relevant nonclinical data, establishing a TMDD model, and using simulations from this model to inform clinical dose regimens.
Collapse
Affiliation(s)
- Hao Cai
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Satoko Kakiuchi-Kiyota
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Robert Hendricks
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Shelly Zhong
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Luna Liu
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Adeyemi O Adedeji
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Pamela Chan
- Biochemical Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Melissa M Schutten
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Meric A Ovacik
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
15
|
FCGR3A gene duplication, FcγRIIb-232TT and FcγRIIIb-HNA1a associate with an increased risk of vertical acquisition of HIV-1. PLoS One 2022; 17:e0273933. [PMID: 36084039 PMCID: PMC9462732 DOI: 10.1371/journal.pone.0273933] [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: 03/03/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background Some mother-to-child transmission (MTCT) studies suggest that allelic variations of Fc gamma receptors (FcγR) play a role in infant HIV-1 acquisition, but findings are inconsistent. To address the limitations of previous studies, the present study investigates the association between perinatal HIV-1 transmission and FcγR variability in three cohorts of South African infants born to women living with HIV-1. Methods This nested case-control study combines FCGR genotypic data from three perinatal cohorts at two hospitals in Johannesburg, South Africa. Children with perinatally-acquired HIV-1 (cases, n = 395) were compared to HIV-1-exposed uninfected children (controls, n = 312). All study participants were black South Africans and received nevirapine for prevention of MTCT. Functional variants were genotyped using a multiplex ligation-dependent probe amplification assay, and their representation compared between groups using logistic regression analyses. Results FCGR3A gene duplication associated with HIV-1 acquisition (OR = 10.27; 95% CI 2.00–52.65; P = 0.005) as did the FcγRIIb-232TT genotype even after adjusting for FCGR3A copy number and FCGR3B genotype (AOR = 1.72; 95%CI 1.07–2.76; P = 0.024). The association between FcγRIIb-232TT genotype and HIV-1 acquisition was further strengthened (AOR = 2.28; 95%CI 1.11–4.69; P = 0.024) if adjusted separately for FCGR2C c.134-96C>T. Homozygous FcγRIIIb-HNA1a did not significantly associate with HIV-1 acquisition in a univariate model (OR = 1.42; 95%CI 0.94–2.16; P = 0.098) but attained significance after adjustment for FCGR3A copy number and FCGR2B genotype (AOR = 1.55; 95%CI 1.01–2.38; P = 0.044). Both FcγRIIb-232TT (AOR = 1.83; 95%CI 1.13–2.97; P = 0.014) and homozygous FcγRIIIb-HNA1a (AOR = 1.66; 95%CI 1.07–2.57; P = 0.025) retained significance when birthweight and breastfeeding were added to the model. The common FCGR2A and FCGR3A polymorphisms did not associate with HIV-1 acquisition. Conclusions Collectively, our findings suggest that the FcγRIIb-232TT genotype exerts a controlling influence on infant susceptibility to HIV-1 infection. We also show a role for less studied variants–FCGR3A duplication and homozygous HNA1a. These findings provide additional insight into a role for FcγRs in HIV-1 infection in children.
Collapse
|
16
|
Gstöttner C, Knaupp A, Vidarsson G, Reusch D, Schlothauer T, Wuhrer M, Domínguez-Vega E. Affinity capillary electrophoresis – mass spectrometry permits direct binding assessment of IgG and FcγRIIa in a glycoform-resolved manner. Front Immunol 2022; 13:980291. [PMID: 36159782 PMCID: PMC9494200 DOI: 10.3389/fimmu.2022.980291] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
The impact of antibody glycoforms on FcγRIIa activation and immune responses is poorly understood. Yet, glycoform binding assessment remains one of the major analytical challenges requiring long enrichment or glycoengineering steps. Here, we developed and applied an affinity capillary electrophoresis-mass spectrometry approach to selectively assess the binding of different antibody glycoforms to the FcγIIa receptor without the need of glycoengineering. The approach required only low microgram amounts of antibody and receptor and enables assessing the binding of high and low-abundance glycoforms. The approach indicated clear differences in binging between doubly-, hemi-glycosylated and non-glycosylated antibodies as well as for mutated (Leu234Ala, Leu235Ala – Pro329-Gly (LALA-PG)) IgG1 antibodies silenced for Fcγ binding. The LALA-PG mutated antibody showed no binding to the FcγIIa receptor (excluding potential non-specific binding effects) while the non-glycosylated IgG1 showed a strongly reduced, but still minor binding. The highest binding affinity was for the antibody carrying two complex-type glycans. Man5 glycans resulted in decreased binding compared to complex-type glycans, with the lowest binding for the IgG containing two Man5. For complex-type glycans, galactosylation showed a subtle increase in binding to the FcγIIa receptor, and sialylation showed an increase in binding for lower sialylated species. Fucosylation did not influence binding to the FcγIIa receptor. Finally, the assay was evaluated for the two variants of the FcγRIIa receptor (allotypes H131 and R131) showing highly comparable glycoform selectivity. Overall, the proposed approach allows the direct comparison of binding affinities of different antibody species in mixtures promising a fast establishment of their structure-function relationships.
Collapse
Affiliation(s)
- Christoph Gstöttner
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, Netherlands
| | - Alexander Knaupp
- Pharma Research and Early Development, Roche Innovation Center Munich, Munich, Germany
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Dietmar Reusch
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg, Germany
| | - Tilman Schlothauer
- Pharma Research and Early Development, Roche Innovation Center Munich, Munich, Germany
| | - Manfred Wuhrer
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, Netherlands
| | - Elena Domínguez-Vega
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, Netherlands
- *Correspondence: Elena Domínguez-Vega,
| |
Collapse
|
17
|
Typiak M, Audzeyenka I, Dubaniewicz A. Presence and possible impact of Fcγ receptors on resident kidney cells in health and disease. Immunol Cell Biol 2022; 100:591-604. [DOI: 10.1111/imcb.12570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/13/2022] [Accepted: 06/28/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Marlena Typiak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences Gdansk Poland
- Department of General and Medical Biochemistry, Faculty of Biology University of Gdansk Gdansk Poland
| | - Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences Gdansk Poland
- Department of Molecular Biotechnology, Faculty of Chemistry University of Gdansk Gdansk Poland
| | - Anna Dubaniewicz
- Department of Pulmonology Medical University of Gdansk Gdansk Poland
| |
Collapse
|
18
|
Delpire B, Van Loon E, Naesens M. The Role of Fc Gamma Receptors in Antibody-Mediated Rejection of Kidney Transplants. Transpl Int 2022; 35:10465. [PMID: 35935272 PMCID: PMC9346079 DOI: 10.3389/ti.2022.10465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
For the past decades, complement activation and complement-mediated destruction of allograft cells were considered to play a central role in anti-HLA antibody-mediated rejection (AMR) of kidney transplants. However, also complement-independent mechanisms are relevant in the downstream immune activation induced by donor-specific antibodies, such as Fc-gamma receptor (FcγR)-mediated direct cellular activation. This article reviews the literature regarding FcγR involvement in AMR, and the potential contribution of FcγR gene polymorphisms to the risk for antibody mediated rejection of kidney transplants. There is large heterogeneity between the studies, both in the definition of the clinical phenotypes and in the technical aspects. The study populations were generally quite small, except for two larger study cohorts, which obviates drawing firm conclusions regarding the associations between AMR and specific FcγR polymorphisms. Although FcγR are central in the pathophysiology of AMR, it remains difficult to identify genetic risk factors for AMR in the recipient’s genome, independent of clinical risk factors, independent of the donor-recipient genetic mismatch, and in the presence of powerful immunosuppressive agents. There is a need for larger, multi-center studies with standardised methods and endpoints to identify potentially relevant FcγR gene polymorphisms that represent an increased risk for AMR after kidney transplantation.
Collapse
Affiliation(s)
- Boris Delpire
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Naesens
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| |
Collapse
|
19
|
Abstract
Leukocyte immunoglobulin-like receptor B4 (LILRB4) is an inhibitory receptor in the LILR family mainly expressed on normal and malignant human cells of myeloid origin. By binding to ligands, LILRB4 is activated and subsequently recruits adaptors to cytoplasmic immunoreceptor tyrosine inhibitory motifs to initiate different signaling cascades, thus playing an important role in physiological and pathological conditions, including autoimmune diseases, microbial infections, and cancers. In normal myeloid cells, LILRB4 regulates intrinsic cell activation and differentiation. In disease-associated or malignant myeloid cells, LILRB4 is significantly correlated with disease severity or patient survival and suppresses T cells, thereby participating in the pathogenesis of various diseases. In summary, LILRB4 functions as an immune checkpoint on myeloid cells and may be a promising therapeutic target for various human immune diseases, especially for cancer immunotherapy.
Collapse
|
20
|
Immunogenetics of Lupus Erythematosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:213-257. [DOI: 10.1007/978-3-030-92616-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
Ebonwu J, Lassaunière R, Paximadis M, Goosen M, Strehlau R, Gray GE, Kuhn L, Tiemessen CT. An HIV Vaccine Protective Allele in FCGR2C Associates With Increased Odds of Perinatal HIV Acquisition. Front Immunol 2021; 12:760571. [PMID: 34917081 PMCID: PMC8668943 DOI: 10.3389/fimmu.2021.760571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
In the Thai RV144 HIV-1 vaccine trial, a three-variant haplotype within the Fc gamma receptor 2C gene (FCGR2C) reduced the risk of HIV-1 acquisition. A follow-on trial, HVTN702, of a similar vaccine candidate found no efficacy in South Africa, where the predominant population is polymorphic for only a single variant in the haplotype, c.134-96C>T (rs114945036). To investigate a role for this variant in HIV-1 acquisition in South Africans, we used the model of maternal-infant HIV-1 transmission. A nested case-control study was conducted of infants born to mothers living with HIV-1, comparing children with perinatally-acquired HIV-1 (cases, n = 176) to HIV-1-exposed uninfected children (controls, n = 349). All had received nevirapine for prevention of mother-to-child transmission. The FCGR2C copy number and expression variants (c.-386G>C, c.-120A>T c.169T>C, and c.798+1A>G) were determined using a multiplex ligation-dependent probe amplification assay and the c.134-96C>T genotype with Sanger sequencing. The copy number, genotype and allele carriage were compared between groups using univariate and multivariate logistic regression. The FCGR2C c.134-96C>T genotype distribution and copy number differed significantly between HIV-1 cases and exposed-uninfected controls (P = 0.002, PBonf = 0.032 and P = 0.010, PBonf = > 0.05, respectively). The FCGR2C c.134-96T allele was overrepresented in the cases compared to the controls (58% vs 42%; P = 0.001, PBonf = 0.016). Adjusting for birthweight and FCGR2C copy number, perinatal HIV-1 acquisition was associated with the c.134-96C>T (AOR = 1.89; 95% CI 1.25-2.87; P = 0.003, PBonf = 0.048) and c.169C>T (AOR = 2.39; 95% CI 1.45-3.95; P = 0.001, PBonf = 0.016) minor alleles but not the promoter variant at position c.-386G>C. The c.134-96C>T variant was in strong linkage disequilibrium with the c.169C>T variant, but remained significantly associated with perinatal acquisition when adjusted for c.169C>T in multivariate analysis. In contrast to the protective effect observed in the Thai RV144 trial, we found the FCGR2C variant c.134-96T-allele associated with increased odds of perinatal HIV-1 acquisition in South African children. These findings, taken together with a similar deleterious association found with HIV-1 disease progression in South African adults, highlight the importance of elucidating the functional relevance of this variant in different populations and vaccination/disease contexts.
Collapse
Affiliation(s)
- Joy Ebonwu
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ria Lassaunière
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Maria Paximadis
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Mark Goosen
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Glenda E. Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Centre, College of Physicians and Surgeons, Columbia University, New York, NY, United States
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Caroline T. Tiemessen
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| |
Collapse
|
22
|
Yang G, Guo N, Yin J, Wu J. Elevated soluble CD163 predicts renal function deterioration in lupus nephritis: a cohort study in Eastern China. J Int Med Res 2021; 49:3000605211049963. [PMID: 34755559 PMCID: PMC8586176 DOI: 10.1177/03000605211049963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective This study investigated the association between soluble scavenger receptor differentiation antigen 163 (sCD163) and the severity and prognosis of renal injury in lupus nephritis (LN). Methods Serum sCD163 levels in 121 Eastern Chinese patients with LN who underwent renal biopsy were determined by enzyme-linked immunosorbent assays. Clinical data were collected, and the glomerular filtration rate and disease activity score of lupus were calculated. Pathological classification was performed, and renal pathological scores were assessed by the activity index (AI) and chronic index (CI). Kaplan–Meier survival curves were drawn to evaluate prognosis. Results The pathological classification, AI and CI scores in the high sCD163 group were increased. The sCD163 levels were positively correlated with serum creatinine, blood urea nitrogen, AI scores and CI scores and negatively correlated with the estimated glomerular filtration rate. Kaplan–Meier survival analysis showed that the incidence of renal endpoint events was increased in the high sCD163 group compared with the normal sCD163 group. Conclusion The serum sCD163 level correlates with the severity of LN and is an important indicator of poor renal prognosis in patients with LN.
Collapse
Affiliation(s)
- Guangxia Yang
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Naifeng Guo
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jun Yin
- Department of Nephrology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, China
| | - Jianhua Wu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
23
|
Onyishi CU, May RC. Human immune polymorphisms associated with the risk of cryptococcal disease. Immunology 2021; 165:143-157. [PMID: 34716931 PMCID: PMC9426616 DOI: 10.1111/imm.13425] [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: 06/25/2021] [Revised: 10/07/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that can cause lethal cryptococcal meningitis in immunocompromised individuals such as those with HIV/AIDS. In addition, cryptococcal infections occasionally arise in immunocompetent individuals or those with previously undiagnosed immunodeficiencies. The course of cryptococcosis is highly variable in both patient groups, and there is rapidly growing evidence that genetic polymorphisms may have a significant impact on the trajectory of disease. Here, we review what is currently known about the nature of these polymorphisms and their impact on host response to C. neoformans infection. Thus far, polymorphisms in Fc gamma receptors, mannose‐binding lectin, Dectin‐2, Toll‐like receptors and macrophage colony‐stimulating factor have been associated with susceptibility to cryptococcal disease. Notably, however, in some cases the impact of these polymorphisms depends on the genetic background of the population; for example, the FCGR3A 158 F/V polymorphism was associated with an increased risk of cryptococcal disease in both HIV‐positive and HIV‐negative white populations, but not in Han Chinese patients. In most cases, the precise mechanism by which the identified polymorphisms influence disease progression remains unclear, although impaired fungal recognition and phagocytosis by innate immune cells appears to play a major role. Finally, we highlight outstanding questions in the field and emphasize the need for future research to include more diverse populations in their genetic association studies.
Collapse
Affiliation(s)
- Chinaemerem U Onyishi
- Institute of Microbiology & Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Robin C May
- Institute of Microbiology & Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
24
|
Zuo Y, Deng GM. Fc Gamma Receptors as Regulators of Bone Destruction in Inflammatory Arthritis. Front Immunol 2021; 12:688201. [PMID: 34248975 PMCID: PMC8262610 DOI: 10.3389/fimmu.2021.688201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 12/29/2022] Open
Abstract
Bone erosion is one of the primary features of inflammatory arthritis and is caused by excessive differentiation and activation of osteoclasts. Fc gamma receptors (FcγRs) have been implicated in osteoclastogenesis. Our recent studies demonstrate that joint-deposited lupus IgG inhibited RANKL-induced osteoclastogenesis. FcγRI is required for RANKL-induced osteoclastogenesis and lupus IgG-induced signaling transduction. We reviewed the results of studies that analyzed the association between FcγRs and bone erosion in inflammatory arthritis. The analysis revealed the dual roles of FcγRs in bone destruction in inflammatory arthritis. Thus, IgG/FcγR signaling molecules may serve as potential therapeutic targets against bone erosion.
Collapse
Affiliation(s)
- Yuyue Zuo
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guo-Min Deng
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Long-Term Hypermethylation of FcγR2B in Leukocytes of Patients with Kawasaki Disease. J Clin Med 2021; 10:jcm10112347. [PMID: 34071896 PMCID: PMC8199050 DOI: 10.3390/jcm10112347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/08/2021] [Accepted: 05/23/2021] [Indexed: 12/18/2022] Open
Abstract
The Fc gamma receptor family contains several activating receptors and the only inhibitory receptor, FcγR2B. In this study, we investigated the dynamic methylation change of FcγR2B in different stages of Kawasaki disease (KD). We enrolled a total of 116 participants, which included patients with febrile diseases as controls and KD patients. Whole blood cells of KD patients were collected prior to intravenous immunoglobulin (IVIG) treatment (KD1), three to seven days after IVIG (KD2), three weeks after IVIG treatment (KD3), six months after IVIG (KD4), and one year after IVIG treatment (KD5). In total, 76 KD patients provided samples in every stage. Leukocytes of controls were also recruited. We performed DNA extraction and pyrosequencing. FcγR2B methylation levels were higher in KD3 compared to both the controls and KD1. A significantly higher methylation of FcγR2B was found in KD5 when compared with KD1. FcγR2B methylation levels in the IVIG-resistant group were lower than those in the IVIG-responsive group at KD1-3 (p = 0.004, 0.004, 0.005 respectively). This study is the first to report the dynamic change of FcγR2B methylation and to demonstrate long-term hypermethylation one year after disease onset. Hypomethylation of FcγR2B is associated with IVIG resistance.
Collapse
|
26
|
Lamptey H, Bonney EY, Adu B, Kyei GB. Are Fc Gamma Receptor Polymorphisms Important in HIV-1 Infection Outcomes and Latent Reservoir Size? Front Immunol 2021; 12:656894. [PMID: 34017334 PMCID: PMC8129575 DOI: 10.3389/fimmu.2021.656894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Fc gamma receptors (FcγR) are cell surface glycoproteins which trigger specific effector-cell responses when cross-linked with the Fc portions of immunoglobulin (IgG) antibodies. During HIV-1 infection, the course of disease progression, ART response, and viral reservoir size vary in different individuals. Several factors may account for these differences; however, Fc gamma receptor gene polymorphisms, which influence receptor binding to IgG antibodies, are likely to play a key role. FcγRIIa (CD32) was recently reported as a potential marker for latent HIV reservoir, however, this assertion is still inconclusive. Whether FcγR polymorphisms influence the size of the viral reservoir, remains an important question in HIV cure studies. In addition, potential cure or viral suppression methods such as broadly neutralizing antibody (bNAbs) may depend on FcγRs to control the virus. Here, we discuss the current evidence on the potential role played by FcγR polymorphisms in HIV-1 infection, treatment and vaccine trial outcomes. Importantly, we highlight contrasting findings that may be due to multiple factors and the relatively limited data from African populations. We recommend further studies especially in sub-Saharan Africa to confirm the role of FcγRIIa in the establishment of latent reservoir and to determine their influence in therapies involving bNAbs.
Collapse
Affiliation(s)
- Helena Lamptey
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Evelyn Y. Bonney
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - George B. Kyei
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Department of Medicine, Washington University School of Medicine in St Louis, St. Louis, MO, United States
- Medical and Scientific Research Centre, University of Ghana Medical Centre, University of Ghana, Accra, Ghana
| |
Collapse
|
27
|
Tajbakhsh A, Farahani N, Gheibihayat SM, Mirkhabbaz AM, Savardashtaki A, Hamblin MR, Mirzaei H. Autoantigen-specific immune tolerance in pathological and physiological cell death: Nanotechnology comes into view. Int Immunopharmacol 2020; 90:107177. [PMID: 33249046 DOI: 10.1016/j.intimp.2020.107177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Apoptotic cells are tolerogenic and can present self-antigens in the absence of inflammation, to antigen-presenting cells by the process of efferocytosis, resulting in anergy and depletion of immune effector cells. This tolerance is essential to maintain immune homeostasis and prevent systemic autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus. Consequently, effective efferocytosis can result in the induction of immune tolerance mediated via triggering modulatory lymphocytes and anti-inflammatory responses. Furthermore, several distinct soluble factors, receptors and pathways have been found to be involved in the efferocytosis, which are able to regulate immune tolerance by lessening antigen presentation, inhibition of T-cell proliferation and induction of regulatory T-cells. Some newly developed nanotechnology-based approaches can induce antigen-specific immunological tolerance without any systemic immunosuppression. These strategies have been explored to reverse autoimmune responses induced against various protein antigens in different diseases. In this review, we describe some nanotechnology-based approaches for the maintenance of self-tolerance using the apoptotic cell clearance process (efferocytosis) that may be able to induce immune tolerance and treat autoimmune diseases.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayed Mohammad Gheibihayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R., Iran.
| |
Collapse
|
28
|
Ladel S, Maigler F, Flamm J, Schlossbauer P, Handl A, Hermann R, Herzog H, Hummel T, Mizaikoff B, Schindowski K. Impact of Glycosylation and Species Origin on the Uptake and Permeation of IgGs through the Nasal Airway Mucosa. Pharmaceutics 2020; 12:E1014. [PMID: 33114132 PMCID: PMC7690786 DOI: 10.3390/pharmaceutics12111014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Although we have recently reported the involvement of neonatal Fc receptor (FcRn) in intranasal transport, the transport mechanisms are far from being elucidated. Ex vivo porcine olfactory tissue, primary cells from porcine olfactory epithelium (OEPC) and the human cell line RPMI 2650 were used to evaluate the permeation of porcine and human IgG antibodies through the nasal mucosa. IgGs were used in their wild type and deglycosylated form to investigate the impact of glycosylation. Further, the expression of FcRn and Fc-gamma receptor (FCGR) and their interaction with IgG were analyzed. Comparable permeation rates for human and porcine IgG were observed in OEPC, which display the highest expression of FcRn. Only traces of porcine IgGs could be recovered at the basolateral compartment in ex vivo olfactory tissue, while human IgGs reached far higher levels. Deglycosylated human IgG showed significantly higher permeation in comparison to the wild type in RPMI 2650 and OEPC, but insignificantly elevated in the ex vivo model. An immunoprecipitation with porcine primary cells and tissue identified FCGR2 as a potential interaction partner in the nasal mucosa. Glycosylation sensitive receptors appear to be involved in the uptake, transport, but also degradation of therapeutic IgGs in the airway epithelial layer.
Collapse
Affiliation(s)
- Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Frank Maigler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Alina Handl
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Rebecca Hermann
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Helena Herzog
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Thomas Hummel
- Smell & Taste Clinic, Department of Otorhinolaryngology, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany;
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| |
Collapse
|
29
|
Gut Microbiota and Immune System Interactions. Microorganisms 2020; 8:microorganisms8101587. [PMID: 33076307 PMCID: PMC7602490 DOI: 10.3390/microorganisms8101587] [Citation(s) in RCA: 415] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Dynamic interactions between gut microbiota and a host’s innate and adaptive immune systems play key roles in maintaining intestinal homeostasis and inhibiting inflammation. The gut microbiota metabolizes proteins and complex carbohydrates, synthesize vitamins, and produce an enormous number of metabolic products that can mediate cross-talk between gut epithelial and immune cells. As a defense mechanism, gut epithelial cells produce a mucosal barrier to segregate microbiota from host immune cells and reduce intestinal permeability. An impaired interaction between gut microbiota and the mucosal immune system can lead to an increased abundance of potentially pathogenic gram-negative bacteria and their associated metabolic changes, disrupting the epithelial barrier and increasing susceptibility to infections. Gut dysbiosis, or negative alterations in gut microbial composition, can also dysregulate immune responses, causing inflammation, oxidative stress, and insulin resistance. Over time, chronic dysbiosis and the translocation of bacteria and their metabolic products across the mucosal barrier may increase prevalence of type 2 diabetes, cardiovascular disease, inflammatory bowel disease, autoimmune disease, and a variety of cancers. In this paper, we highlight the pivotal role gut microbiota and their metabolites (short-chain fatty acids (SCFAs)) play in mucosal immunity.
Collapse
|
30
|
Huang L, Yu X, Li L, Liu J, Wu X, Zeng Y, Liao X, Liu W, Zhang F, Zhang X, Chen H, Zheng W. Aberrant FcγRIIb and FcγRIII expression on monocytes from patients with Behçet's disease. Clin Immunol 2020; 219:108549. [PMID: 32739412 DOI: 10.1016/j.clim.2020.108549] [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: 04/21/2020] [Revised: 07/26/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
Behçet's disease (BD) patients have abnormal FcγR polymorphisms, the implication of which remains elusive. We examined FcγRIIb expression on neutrophils, monocytes, B cells, natural killer cells, dendritic cells and T cells, and FcγRI and FcγRIII expression on monocytes in BD patients and healthy controls using flow cytometry. We further stimulated monocytes with IgG and (or) lipopolysaccharide (LPS) and measured IL-6 and TNF-α production by enzyme-linked immunosorbent assay. We found that BD monocytes expressed a lower level of FcγRIIb and a higher level of FcγRIII, which were correlated with erythrocyte sedimentation rate and C-reactive protein and were rescued after treatment. Furthermore, LPS- and IgG-stimulated BD monocytes produced higher levels of IL-6 and TNF-α than HC monocytes. In summary, we found that BD monocytes downregulated FcγRIIb expression and upregulated FcγRIII expression, which were correlated with disease activity and potentially contributed to monocyte hyperactivation in BD.
Collapse
Affiliation(s)
- Linfang Huang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China; Department of Rheumatology and Clinical Immunology, The First People's Hospital of Yueyang, Yueyang, Hunan 414000, China
| | - Xin Yu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Lu Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Jinjing Liu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Xiuhua Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Yan Zeng
- Department of Rheumatology and Clinical Immunology, The First People's Hospital of Yueyang, Yueyang, Hunan 414000, China
| | - Xiangping Liao
- Department of Rheumatology and Clinical Immunology, The First People's Hospital of ChenZhou, ChenZhou, Hunan 423000, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Fengchun Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Xuan Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Hua Chen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China.
| | - Wenjie Zheng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
31
|
Olson WJ, Jakic B, Hermann‐Kleiter N. Regulation of the germinal center response by nuclear receptors and implications for autoimmune diseases. FEBS J 2020; 287:2866-2890. [PMID: 32246891 PMCID: PMC7497069 DOI: 10.1111/febs.15312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/15/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023]
Abstract
The immune system plays an essential role in protecting the host from infectious diseases and cancer. Notably, B and T lymphocytes from the adaptive arm of the immune system can co-operate to form long-lived antibody responses and are therefore the main target in vaccination approaches. Nevertheless, protective immune responses must be tightly regulated to avoid hyper-responsiveness and responses against self that can result in autoimmunity. Nuclear receptors (NRs) are perfectly adapted to rapidly alter transcriptional cellular responses to altered environmental settings. Their functional role is associated with both immune deficiencies and autoimmunity. Despite extensive linking of nuclear receptor function with specific CD4 T helper subsets, research on the functional roles and mechanisms of specific NRs in CD4 follicular T helper cells (Tfh) and germinal center (GC) B cells during the germinal center reaction is just emerging. We review recent advances in our understanding of NR regulation in specific cell types of the GC response and discuss their implications for autoimmune diseases such as systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- William J. Olson
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| | - Bojana Jakic
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
- Department of Immunology, Genetics and PathologyUppsala UniversitySweden
| | - Natascha Hermann‐Kleiter
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| |
Collapse
|
32
|
Marcoux G, Laroche A, Espinoza Romero J, Boilard E. Role of platelets and megakaryocytes in adaptive immunity. Platelets 2020; 32:340-351. [PMID: 32597341 DOI: 10.1080/09537104.2020.1786043] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is comprised of two principal interconnected components called innate and adaptive immunity. While the innate immune system mounts a nonspecific response that provides protection against the spread of foreign pathogens, the adaptive immune system has developed to specifically recognize a given pathogen and lead to immunological memory. Platelets are small fragments produced from megakaryocytes in bone marrow and lungs. They circulate throughout the blood to monitor the integrity of the vasculature and to prevent bleeding. Given their large repertoire of immune receptors and inflammatory molecules, platelets and megakaryocytes can contribute to both innate and adaptive immunity. In adaptive immunity, platelets and megakaryocytes can process and present antigens to lymphocytes. Moreover, platelets, via FcγRIIA, rapidly respond to pathogens in an immune host when antibodies are present. This manuscript reviews the reported contributions of platelets and megakaryocytes with emphasis on antigen presentation and antibody response in adaptive immunity.
Collapse
Affiliation(s)
- Genevieve Marcoux
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Jenifer Espinoza Romero
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Eric Boilard
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| |
Collapse
|
33
|
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by multiple system involvement and positive serum autoantibodies. Lupus nephritis (LN) is the most common and serious complication of SLE, and it is the main cause of death in patients with SLE. Abnormalities in the immune system lead to LN and involve a variety of cells (T cells, B cells, macrophages, NK cells, etc.), cytokines (interleukin, tumor necrosis factor α, etc.) and their related pathways. Previous studies have shown that the interactions of genetic, epigenetic and environmental factors contribute to the pathogenesis and development of LN. In recent years, one genome-wide association study (GWAS) and a number of gene association studies have explored the susceptibility genes of LN, including immunization-, inflammation-, adhesion- and other pathway-related genes. These genes participate in or suggest the pathogenesis and progression of LN. In this review, we summarize the genetic susceptibility of LN and discuss the possible mechanism underlying the susceptibility genes of LN.
Collapse
|
34
|
Manoylov IK, Boneva GV, Doytchinova IA, Mihaylova NM, Tchorbanov AI. Suppression of Disease-Associated B Lymphocytes by GAD65 Epitope-Carrying Protein-Engineered Molecules in a Streptozotocin-Induced Mouse Model of Diabetes. Monoclon Antib Immunodiagn Immunother 2020; 38:201-208. [PMID: 31603741 DOI: 10.1089/mab.2019.0030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Type 1 diabetes mellitus is an autoimmune syndrome defined by the presence of autoreactive T and B cells, which results in destruction of insulin-producing beta cells. Autoantibodies against GAD65 (glutamic acid decarboxylase 65)-a membrane-bound enzyme on pancreatic beta cells, contribute to beta cells' destruction and the loss of pancreatic functions. Mouse FcγRIIb on B lymphocytes possesses an inhibitory effect on the activity of these cells. We hypothesized that it may be possible to suppress GAD65-specific B cells in mice with diabetes using chimeric molecules, containing an anti-FcγRIIb antibody, coupled to peptide B/T epitopes derived from the GAD65 protein. With these engineered chimeras, we expect to selectively co-cross-link the anti-GAD65-specific B cell receptor (BCR) and FcγRIIb, thus delivering a suppressive signal to the targeted B cells. An anti-FcγRIIb monoclonal antibody and two synthetic peptide epitopes derived from the GAD65 molecule were used for chimeras' construction. The suppressive activity of the engineered molecules was tested in vivo in mice with streptozotocin (STZ)-induced type 1 diabetes. These chimeric molecules exclusively bind disease-associated B cells by recognizing their GAD65-specific BCR and selectively deliver a strong inhibitory signal through their surface FcγRIIb receptors. A reduction in the number of anti-GAD65 IgG antibody-secreting plasmocytes and an increased percentage of apoptotic B lymphocytes were observed after treatment with protein-engineered antibodies of mice with STZ-induced type 1 diabetes.
Collapse
Affiliation(s)
- Iliyan Konstantinov Manoylov
- Laboratory of Experimental Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Gabriela Valentinova Boneva
- Laboratory of Experimental Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Nikolina Mihaylova Mihaylova
- Laboratory of Experimental Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Andrey Ivanov Tchorbanov
- Laboratory of Experimental Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
35
|
Li J, Wang L, Tian J, Zhou Z, Li J, Yang H. Nongenetic engineering strategies for regulating receptor oligomerization in living cells. Chem Soc Rev 2020; 49:1545-1568. [DOI: 10.1039/c9cs00473d] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nongenetic strategies for regulating receptor oligomerization in living cells based on DNA, protein, small molecules and physical stimuli.
Collapse
Affiliation(s)
- Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Jinmiao Tian
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Zhilan Zhou
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| |
Collapse
|
36
|
Song K, Liu L, Zhang X, Chen X. An update on genetic susceptibility in lupus nephritis. Clin Immunol 2019; 210:108272. [PMID: 31683055 DOI: 10.1016/j.clim.2019.108272] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/11/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by multiple system involvement and positive serum autoantibodies. Lupus nephritis (LN) is the most common and serious complication of SLE, and it is the main cause of death in patients with SLE. Abnormalities in the immune system lead to LN and involve a variety of cells (T cells, B cells, macrophages, NK cells, etc.), cytokines (interleukin, tumor necrosis factor α, etc.) and their related pathways. Previous studies have shown that the interactions of genetic, epigenetic and environmental factors contribute to the pathogenesis and development of LN. In recent years, one genome-wide association study (GWAS) and a number of gene association studies have explored the susceptibility genes of LN, including immunization-, inflammation-, adhesion- and other pathway-related genes. These genes participate in or suggest the pathogenesis and progression of LN. In this review, we summarize the genetic susceptibility of LN and discuss the possible mechanism underlying the susceptibility genes of LN.
Collapse
Affiliation(s)
- Kangkang Song
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, 28 Fuxing Road, Beijing, China
| | - Lu Liu
- Institute of Dermatology and Department of Dermatology at No.1 Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education (Anhui Medical University), Hefei, Anhui, China
| | - Xuejun Zhang
- Institute of Dermatology and Department of Dermatology at No.1 Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education (Anhui Medical University), Hefei, Anhui, China; Institute of Dermatology and Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, 28 Fuxing Road, Beijing, China.
| |
Collapse
|
37
|
Wang Y, Jönsson F. Expression, Role, and Regulation of Neutrophil Fcγ Receptors. Front Immunol 2019; 10:1958. [PMID: 31507592 PMCID: PMC6718464 DOI: 10.3389/fimmu.2019.01958] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/02/2019] [Indexed: 12/31/2022] Open
Abstract
Neutrophils are best known for their critical role in host defense, for which they utilize multiple innate immune mechanisms, including microbe-associated pattern recognition, phagocytosis, production of reactive oxygen species, and the release of potent proteases, mediators, antimicrobials, and neutrophil extracellular traps. Beyond their well-established contribution to innate immunity, neutrophils were more recently reported to interact with various other cell types, including cells from the adaptive immune system, thereby enabling neutrophils to tune the overall immune response of the host. Neutrophils express different receptors for IgG antibodies (Fcγ receptors), which facilitate the engulfment of IgG-opsonized microbes and trigger cell activation upon cross-linking of several receptors. Indeed, FcγRs (via IgG antibodies) confer neutrophils with a key feature of the adaptive immunity: an antigen-specific cell response. This review summarizes the expression and function of FcγRs on human neutrophils in health and disease and how they are affected by polymorphisms in the FCGR loci. Additionally, we will discuss the role of neutrophils in providing help to marginal zone B cells for the production of antibodies, which in turn may trigger neutrophil effector functions when engaging FcγRs.
Collapse
Affiliation(s)
- Yu Wang
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, Paris, France
- Université Diderot Paris VII, PSL University, Paris, France
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, Paris, France
| |
Collapse
|
38
|
Habib T, Long SA, Samuels PL, Brahmandam A, Tatum M, Funk A, Hocking AM, Cerosaletti K, Mason MT, Whalen E, Rawlings DJ, Greenbaum C, Buckner JH. Dynamic Immune Phenotypes of B and T Helper Cells Mark Distinct Stages of T1D Progression. Diabetes 2019; 68:1240-1250. [PMID: 30894366 PMCID: PMC6610015 DOI: 10.2337/db18-1081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/15/2019] [Indexed: 01/01/2023]
Abstract
Multiple studies of B- and T-cell compartments and their response to stimuli demonstrate alterations in established type 1 diabetes (T1D). Yet it is not known whether these alterations reflect immune mechanisms that initiate islet autoimmunity, promote disease progression, or are secondary to disease. To address these questions, we used samples from the TrialNet Pathway to Prevention study to investigate T-cell responses to interleukin (IL)-2 and regulatory T cell-mediated suppression, the composition of the B-cell compartment, and B-cell responses to B-cell receptor and IL-21 receptor engagement. These studies revealed stage-dependent T- and B-cell functional and immune phenotypes; namely, early features that differentiate autoantibody-positive at-risk first-degree relatives (FDRs) from autoantibody-negative FDRs and persisted through clinical diagnosis; late features that arose at or near T1D diagnosis; and dynamic features that were enhanced early and blunted at later disease stages, indicating evolving responses along the continuum of T1D. We further explored how these specific phenotypes are influenced by therapeutic interventions. Our integrated studies provide unique insights into stable and dynamic stage-specific immune states and define novel immune phenotypes of potential clinical relevance.
Collapse
Affiliation(s)
- Tania Habib
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - S Alice Long
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Peter L Samuels
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Archana Brahmandam
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Megan Tatum
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Andrew Funk
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Anne M Hocking
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Karen Cerosaletti
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Michael T Mason
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Elizabeth Whalen
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
- Departments of Pediatrics and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Carla Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | | |
Collapse
|
39
|
Null donors favor prepared immunologists. Blood 2019; 133:1389-1390. [PMID: 30923104 DOI: 10.1182/blood-2019-01-898775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
40
|
Nagelkerke SQ, Tacke CE, Breunis WB, Tanck MWT, Geissler J, Png E, Hoang LT, van der Heijden J, Naim ANM, Yeung RSM, Levin ML, Wright VJ, Burgner DP, Ponsonby AL, Ellis JA, Cimaz R, Shimizu C, Burns JC, Fijnvandraat K, van der Schoot CE, van den Berg TK, de Boer M, Davila S, Hibberd ML, Kuijpers TW. Extensive Ethnic Variation and Linkage Disequilibrium at the FCGR2/3 Locus: Different Genetic Associations Revealed in Kawasaki Disease. Front Immunol 2019; 10:185. [PMID: 30949161 PMCID: PMC6437109 DOI: 10.3389/fimmu.2019.00185] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/21/2019] [Indexed: 12/23/2022] Open
Abstract
The human Fc-gamma receptors (FcγRs) link adaptive and innate immunity by binding immunoglobulin G (IgG). All human low-affinity FcγRs are encoded by the FCGR2/3 locus containing functional single nucleotide polymorphisms (SNPs) and gene copy number variants. This locus is notoriously difficult to genotype and high-throughput methods commonly used focus on only a few SNPs. We performed multiplex ligation-dependent probe amplification for all relevant genetic variations at the FCGR2/3 locus in >4,000 individuals to define linkage disequilibrium (LD) and allele frequencies in different populations. Strong LD and extensive ethnic variation in allele frequencies was found across the locus. LD was strongest for the FCGR2C-ORF haplotype (rs759550223+rs76277413), which leads to expression of FcγRIIc. In Europeans, the FCGR2C-ORF haplotype showed strong LD with, among others, rs201218628 (FCGR2A-Q27W, r2 = 0.63). LD between these two variants was weaker (r2 = 0.17) in Africans, whereas the FCGR2C-ORF haplotype was nearly absent in Asians (minor allele frequency <0.005%). The FCGR2C-ORF haplotype and rs1801274 (FCGR2A-H131R) were in weak LD (r2 = 0.08) in Europeans. We evaluated the importance of ethnic variation and LD in Kawasaki Disease (KD), an acute vasculitis in children with increased incidence in Asians. An association of rs1801274 with KD was previously shown in ethnically diverse genome-wide association studies. Now, we show in 1,028 European KD patients that the FCGR2C-ORF haplotype, although nearly absent in Asians, was more strongly associated with susceptibility to KD than rs1801274 in Europeans. Our data illustrate the importance of interpreting findings of association studies concerning the FCGR2/3 locus with knowledge of LD and ethnic variation.
Collapse
Affiliation(s)
- Sietse Q Nagelkerke
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carline E Tacke
- Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Willemijn B Breunis
- Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Michael W T Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Judy Geissler
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eileen Png
- Infectious Diseases, Genome Institute of Singapore, Singapore, Singapore
| | - Long T Hoang
- Infectious Diseases, Genome Institute of Singapore, Singapore, Singapore
| | - Joris van der Heijden
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ahmad N M Naim
- Infectious Diseases, Genome Institute of Singapore, Singapore, Singapore
| | - Rae S M Yeung
- Division of Rheumatology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Michael L Levin
- Department of Pediatrics, Imperial College London, London, United Kingdom
| | - Victoria J Wright
- Department of Pediatrics, Imperial College London, London, United Kingdom
| | - David P Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Justine A Ellis
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Health, Centre for Social and Early Emotional Development, Deakin University, Burwood, VIC, Australia
| | - Rolando Cimaz
- Rheumatology Unit, Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Chisato Shimizu
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, United States
| | - Jane C Burns
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, United States
| | - Karin Fijnvandraat
- Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Plasma Proteins, Sanquin Research, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - C Ellen van der Schoot
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Martin de Boer
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Martin L Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore, Singapore.,Department of Pathogen Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Identification of function and potential pathogenic mechanisms of SLE risk genes in dendritic cells. RECENT FINDINGS Functional studies of individual SLE risk factors in dendritic cells were performed, and functional alterations of some risk genes in dendritic cells were observed. Recent studies confirmed the pathogenic function of known risk genes. These findings postulate novel pathogenic mechanisms made by dendritic cells. SLE is a complex disease and its etiology is not clearly understood. Dendritic cells are innate immune cells and critical for determining immune activation and immune tolerance. Genetic studies identified several new candidate genes which predispose to development of autoimmune diseases, but the mechanism of those genes has not been identified. This report updates functional implications or pathways in dendritic cells which are putatively important for the development or propagation of SLE based on genetic and functional studies performed in both human and animal models.
Collapse
Affiliation(s)
- Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Disease, Department of Molecular Medicine, The Feinstein Institute for Medical Research, School of Medicine at Northwell-Hofstra University, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
42
|
Polymorphism of FCGR3A gene in chronic beryllium disease. Genes Immun 2018; 20:493-499. [PMID: 30245507 PMCID: PMC6431584 DOI: 10.1038/s41435-018-0046-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022]
Abstract
Previously we showed that alveolar macrophages(AMs) from patients with chronic beryllium disease(CBD) and beryllium sensitization(BeS) demonstrated significantly greater cell surface CD16(encoded by the FCGR3A gene) than controls. We hypothesized that these differences were related to polymorphisms in the FCGR3A gene. This study was to determine the association between FCGR3A polymorphisms in CBD, BeS versus controls as well as clinical data, providing potential information about disease pathogenesis, risk and activity. 189 CBD/154 BeS /150 controls (92 Be-exposed-non-diseased and 58 healthy controls) were included in this study. Sequence specific primers polymerase chain reaction(PCR-SSP) was used to determine FCGR3A 158V/F polymorphisms. We found significantly higher frequencies of the 158V allele (OR:1.60 (CI: 1.17-2.19),p=0.004) and 158VV homozygotes (OR:2.97 (CI:1.48-5.97) p=0.007) in CBD versus controls. No differences were found in the frequencies of FCGR3A alleles or genotypes between BeS versus controls and CBD versus BeS. Average changes in exercise testing maximum workload(Wlm),maximum oxygen consumption(VO2m) and diffusion capacity of carbon monoxide(DLCO) demonstrated greater decline over time in those CBD cases with the 158VV gene, modelled between 10 and 40 years from first beryllium exposure. The FCGR3A V158F polymorphism is associated with CBD compared to BeS and controls and may impact lung function in CBD.
Collapse
|
43
|
Herrlich A, Kefaloyianni E. iRhoms: A Potential Path to More Specific Therapeutic Targeting of Lupus Nephritis. Am J Kidney Dis 2018; 72:617-619. [PMID: 29887489 DOI: 10.1053/j.ajkd.2018.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Andreas Herrlich
- Washington University School of Medicine in St. Louis, St. Louis, MO.
| | | |
Collapse
|
44
|
Anaya JM, Leon KJ, Rojas M, Rodriguez Y, Pacheco Y, Acosta-Ampudia Y, Monsalve DM, Ramirez-Santana C. Progress towards precision medicine for lupus: the role of genetic biomarkers. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1448266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Kelly J. Leon
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yhojan Rodriguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yovana Pacheco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Carolina Ramirez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
45
|
Qing X, Chinenov Y, Redecha P, Madaio M, Roelofs JJ, Farber G, Issuree PD, Donlin L, Mcllwain DR, Mak TW, Blobel CP, Salmon JE. iRhom2 promotes lupus nephritis through TNF-α and EGFR signaling. J Clin Invest 2018; 128:1397-1412. [PMID: 29369823 DOI: 10.1172/jci97650] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/23/2018] [Indexed: 12/14/2022] Open
Abstract
Lupus nephritis (LN) often results in progressive renal dysfunction. The inactive rhomboid 2 (iRhom2) is a newly identified key regulator of A disintegrin and metalloprotease 17 (ADAM17), whose substrates, such as TNF-α and heparin-binding EGF (HB-EGF), have been implicated in the pathogenesis of chronic kidney diseases. Here, we demonstrate that deficiency of iRhom2 protects the lupus-prone Fcgr2b-/- mice from developing severe kidney damage without altering anti-double-stranded DNA (anti-dsDNA) Ab production by simultaneously blocking HB-EGF/EGFR and TNF-α signaling in the kidney tissues. Unbiased transcriptome profiling of kidneys and kidney macrophages revealed that TNF-α and HB-EGF/EGFR signaling pathways are highly upregulated in Fcgr2b-/- mice, alterations that were diminished in the absence of iRhom2. Pharmacological blockade of either TNF-α or EGFR signaling protected Fcgr2b-/- mice from severe renal damage. Finally, kidneys from LN patients showed increased iRhom2 and HB-EGF expression, with interstitial HB-EGF expression significantly associated with chronicity indices. Our data suggest that activation of iRhom2/ADAM17-dependent TNF-α and EGFR signaling plays a crucial role in mediating irreversible kidney damage in LN, thereby uncovering a target for selective and simultaneous dual inhibition of 2 major pathological pathways in the effector arm of the disease.
Collapse
Affiliation(s)
| | - Yurii Chinenov
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | | | - Michael Madaio
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Joris Jth Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Gregory Farber
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA
| | - Priya D Issuree
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - Laura Donlin
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - David R Mcllwain
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA.,Institute for Advanced Study, Technical University Munich, Munich, Germany.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Jane E Salmon
- Program in Inflammation and Autoimmunity, and.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
46
|
Li W, Sivakumar R, Titov AA, Choi SC, Morel L. Metabolic Factors that Contribute to Lupus Pathogenesis. Crit Rev Immunol 2017; 36:75-98. [PMID: 27480903 DOI: 10.1615/critrevimmunol.2016017164] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which organ damage is mediated by pathogenic autoantibodies directed against nucleic acids and protein complexes. Studies in SLE patients and in mouse models of lupus have implicated virtually every cell type in the immune system in the induction or amplification of the autoimmune response as well as the promotion of an inflammatory environment that aggravates tissue injury. Here, we review the contribution of CD4+ T cells, B cells, and myeloid cells to lupus pathogenesis and then discuss alterations in the metabolism of these cells that may contribute to disease, given the recent advances in the field of immunometabolism.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610; Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology, Beijing Key Laboratory, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Ramya Sivakumar
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Anton A Titov
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
47
|
Typiak M, Rębała K, Haraś A, Skotarczak M, Słomiński JM, Dubaniewicz A. Copy number variation of FCGR genes in etiopathogenesis of sarcoidosis. PLoS One 2017; 12:e0177194. [PMID: 28472129 PMCID: PMC5417662 DOI: 10.1371/journal.pone.0177194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 04/24/2017] [Indexed: 11/19/2022] Open
Abstract
We have previously revealed that, in contrast to polymorphism of FCGR2B and FCGR3B, polymorphism of FCGR2A, FCGR2C and FCGR3A genes, encoding receptors for Fc fragment of immunoglobulin G (Fcγ receptors), play a role in increased level of circulating immune complexes with occurrence of Mycobacterium tuberculosis heat shock proteins in patients with sarcoidosis. However, this immunocomplexemia might also be caused by decreased clearance by immune cells due to a changed copy number of FCGR genes. Thus, the next step of our study was to evaluate copy number variation of FCGR2A, FCGR2B, FCGR2C, FCGR3A and FCGR3B in this disease. The analysis was carried out by real-time quantitative PCR on 104 patients and 110 healthy volunteers. Despite previously detected variation in allele/genotype frequencies of FCGR in sarcoidosis and its particular stages, there was no copy number variation of the tested genes between sarcoidosis or its stages and healthy control, as well as between stages themselves. A relevant increase in copy number of FCGR2C and FCGR3B in Stage IV of sarcoidosis vs. other stages and controls was detected, but this observation was based on a limited number of Stage IV patients. Hence, polymorphism of FCGR genes seems to be more important than their copy number variation in etiopathogenesis of sarcoidosis in patients from the Polish population.
Collapse
Affiliation(s)
- Marlena Typiak
- Department of Pulmonology, Medical University of Gdansk, Gdansk, Poland
| | - Krzysztof Rębała
- Department of Forensic Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Haraś
- Department of Forensic Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Monika Skotarczak
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland
| | | | - Anna Dubaniewicz
- Department of Pulmonology, Medical University of Gdansk, Gdansk, Poland
- * E-mail:
| |
Collapse
|
48
|
Margery-Muir AA, Bundell C, Nelson D, Groth DM, Wetherall JD. Gender balance in patients with systemic lupus erythematosus. Autoimmun Rev 2017; 16:258-268. [DOI: 10.1016/j.autrev.2017.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
|
49
|
Krahn N, Spearman M, Meier M, Dorion-Thibaudeau J, McDougall M, Patel TR, De Crescenzo G, Durocher Y, Stetefeld J, Butler M. Inhibition of glycosylation on a camelid antibody uniquely affects its FcγRI binding activity. Eur J Pharm Sci 2017; 96:428-439. [DOI: 10.1016/j.ejps.2016.09.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/13/2016] [Accepted: 09/30/2016] [Indexed: 11/27/2022]
|
50
|
Grecco M, Santos VCD, Pereira KMC, Andrade LEC, Silva NPD. Fc gamma receptor IIIa polymorphism is not associated with susceptibility to systemic lupus erythematosus in Brazilian patients. REVISTA BRASILEIRA DE REUMATOLOGIA 2016; 56:515-520. [PMID: 27914599 DOI: 10.1016/j.rbre.2016.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 06/15/2016] [Indexed: 10/20/2022] Open
Abstract
We evaluated the possible association between FCGR3A V/F (158) polymorphism and SLE susceptibility and clinical phenotype in 305 sequentially retrieved SLE patients and 300 healthy controls from the southeastern part of Brazil by allele-specific polymerase chain reaction. Our results showed no association between FCGR3A 158V/F alleles and susceptibility to SLE in this series of patients albeit the heterozygous genotype was strongly associated with the disease.
Collapse
Affiliation(s)
- Marcelle Grecco
- Universidade Federal de São Paulo (UNIFESP), Departamento de Medicina, Disciplina de Reumatologia, São Paulo, SP, Brazil
| | - Viviane Cardoso Dos Santos
- Universidade Federal de São Paulo (UNIFESP), Departamento de Medicina, Disciplina de Reumatologia, São Paulo, SP, Brazil
| | - Kaline Medeiros Costa Pereira
- Universidade Federal de São Paulo (UNIFESP), Departamento de Medicina, Disciplina de Reumatologia, São Paulo, SP, Brazil
| | - Luís Eduardo Coelho Andrade
- Universidade Federal de São Paulo (UNIFESP), Departamento de Medicina, Disciplina de Reumatologia, São Paulo, SP, Brazil
| | - Neusa Pereira da Silva
- Universidade Federal de São Paulo (UNIFESP), Departamento de Medicina, Disciplina de Reumatologia, São Paulo, SP, Brazil.
| |
Collapse
|