1
|
Gan SY, Tye GJ, Chew AL, Lai NS. Current development of Fc gamma receptors (FcγRs) in diagnostics: a review. Mol Biol Rep 2024; 51:937. [PMID: 39190190 DOI: 10.1007/s11033-024-09877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
The ability of the immune system to fight against pathogens relies on the intricate collaboration between antibodies and Fc gamma receptors (FcγRs). These receptors are a group of transmembrane glycoprotein molecules, which can specifically detect and bind to the Fc portion of immunoglobulin G (IgG) molecules. They are distributed on a diverse array of immune cells, forming a strong defence system to eliminate invading threats. FcγRs have gained increasing attention as potential biomarkers for various diseases in recent years due to their ability to reflect immune dysregulation and disease pathogenesis. Increasing lines of evidence have shed new light on the remarkable association of FcγRs polymorphisms with the susceptibility of autoimmune diseases such as systemic lupus erythematosus (SLE) and lupus nephritis. Several studies have also reported the application of FcγR as a novel biomarker for the diagnosis of infection and cancer. Due to the surge in interest and concern regarding the potential of FcγRs as promising diagnostic biomarkers, this review, thereby, serves to provide a comprehensive overview of the structural characteristics, functional roles, and expression patterns of FcγRs, with a particular focus on their evolving role as diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Shin Yi Gan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Halaman Bukit Gambir, Gelugor, Penang, 11700, Malaysia
| | - Ai Lan Chew
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Ngit Shin Lai
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia.
| |
Collapse
|
2
|
Uribe-Querol E, Rosales C. Phagocytosis. Methods Mol Biol 2024; 2813:39-64. [PMID: 38888769 DOI: 10.1007/978-1-0716-3890-3_3] [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: 06/20/2024]
Abstract
One hundred years have passed since the death of Élie Metchnikoff (1845-1916). He was the first to observe the uptake of particles by cells and realized the importance of this process, named phagocytosis, for the host response to injury and infection. He also was a strong advocate of the role of phagocytosis in cellular immunity, and with this, he gave us the basis for our modern understanding of inflammation and the innate immune response. Phagocytosis is an elegant but complex process for the ingestion and elimination of pathogens, but it is also important for the elimination of apoptotic cells and hence fundamental for tissue homeostasis. Phagocytosis can be divided into four main steps: (i) recognition of the target particle, (ii) signaling to activate the internalization machinery, (iii) phagosome formation, and (iv) phagolysosome maturation. In this chapter, we present a general view of our current knowledge on phagocytosis performed mainly by professional phagocytes through antibody and complement receptors and discuss aspects that remain incompletely understood.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- Laboratorio de Biología del Desarrollo, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
3
|
Abstract
Although the need for a universal influenza vaccine has long been recognized, only a handful of candidates have been identified so far, with even fewer advancing in the clinical pipeline. The 24–amino acid ectodomain of M2 protein (M2e) has been developed over the past two decades. However, M2e-based vaccine candidates have shortcomings, including the need for several administrations and the lack of sustained antibody titers over time. We report here a vaccine targeting strategy that has the potential to confer sustained and strong protection upon a single shot of a small amount of M2e antigen. The current COVID-19 pandemic has highlighted the importance of developing versatile, powerful platforms for the rapid deployment of vaccines against any incoming threat. Influenza, commonly referred to as “flu,” is a major global public health concern and a huge economic burden to societies. Current influenza vaccines need to be updated annually to match circulating strains, resulting in low take-up rates and poor coverage due to inaccurate prediction. Broadly protective universal flu vaccines that do not need to be updated annually have therefore been pursued. The highly conserved 24–amino acid ectodomain of M2 protein (M2e) is a leading candidate, but its poor immunogenicity has been a major roadblock in its clinical development. Here, we report a targeting strategy that shuttles M2e to a specific dendritic cell subset (cDC1) by engineering a recombinant anti-Clec9A monoclonal antibody fused at each of its heavy chains with three copies of M2e. Single administration in mice of 2 µg of the Clec9A–M2e construct triggered an exceptionally sustained anti-M2e antibody response and resulted in a strong anamnestic protective response upon influenza challenge. Furthermore, and importantly, Clec9A–M2e immunization significantly boosted preexisting anti-M2e titers from prior flu exposure. Thus, the Clec9A-targeting strategy allows antigen and dose sparing, addressing the shortcomings of current M2e vaccine candidates. As the cDC1 subset exists in humans, translation to humans is an exciting and realistic avenue.
Collapse
|
4
|
Blanco-Camarillo C, Alemán OR, Rosales C. Low-Density Neutrophils in Healthy Individuals Display a Mature Primed Phenotype. Front Immunol 2021; 12:672520. [PMID: 34276661 PMCID: PMC8285102 DOI: 10.3389/fimmu.2021.672520] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human peripheral blood, comprising about 70% of all leukocytes. They are regarded as the first line of defense of the innate immune system, but neutrophils have also the ability of regulating the adaptive immune response. Recently, However, multiple phenotypes and functional states of neutrophils have been reported, particularly in inflammation, autoimmunity, and cancer. One possible subtype of neutrophils, the so-called low-density neutrophils (LDN) is found among mononuclear cells (MNC), monocytes and lymphocytes, after separating the leukocytes from blood by density gradient centrifugation. LDN increase in numbers during several pathological conditions. However, LDN present in healthy conditions have not been investigated further. Therefore, in order to confirm the presence of LDN in blood of healthy individuals and to explore some of their cellular functions, neutrophils and MNC were isolated by density gradient centrifugation. Purified neutrophils were further characterized by multicolor flow cytometry (FACS) and then, using the same FACS parameters cells in the MNC fraction were analyzed. Within the MNC, LDN were consistently found. These LDN had a normal mature neutrophil morphology and displayed a CD10+, CD11b+, CD14low, CD15high, CD16bhigh, CD62L+, CD66b+, and CXCR4+ phenotype. These LDN had an enhanced reactive oxygen species (ROS) production and increased phagocytic capacity and were able to produce neutrophil extracellular traps (NET) similarly to neutrophils. These data confirm the presence of a small number of LDN is blood of healthy individuals and suggest that these LDN represent mature cells with a primed phenotype.
Collapse
Affiliation(s)
- Carlos Blanco-Camarillo
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Omar Rafael Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
5
|
Zalfa C, Paust S. Natural Killer Cell Interactions With Myeloid Derived Suppressor Cells in the Tumor Microenvironment and Implications for Cancer Immunotherapy. Front Immunol 2021; 12:633205. [PMID: 34025641 PMCID: PMC8133367 DOI: 10.3389/fimmu.2021.633205] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous environment composed of cancer cells, tumor stroma, a mixture of tissue-resident and infiltrating immune cells, secreted factors, and extracellular matrix proteins. Natural killer (NK) cells play a vital role in fighting tumors, but chronic stimulation and immunosuppression in the TME lead to NK cell exhaustion and limited antitumor functions. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells with potent immunosuppressive activity that gradually accumulate in tumor tissues. MDSCs interact with innate and adaptive immune cells and play a crucial role in negatively regulating the immune response to tumors. This review discusses MDSC-mediated NK cell regulation within the TME, focusing on critical cellular and molecular interactions. We review current strategies that target MDSC-mediated immunosuppression to enhance NK cell cytotoxic antitumor activity. We also speculate on how NK cell-based antitumor immunotherapy could be improved.
Collapse
Affiliation(s)
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
6
|
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
|
7
|
Barb AW. Fc γ receptor compositional heterogeneity: Considerations for immunotherapy development. J Biol Chem 2021; 296:100057. [PMID: 33172893 PMCID: PMC7948983 DOI: 10.1074/jbc.rev120.013168] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
The antibody-binding crystallizable fragment (Fc) γ receptors (FcγRs) are expressed by leukocytes and activate or suppress a cellular response once engaged with an antibody-coated target. Therapeutic mAbs that require FcγR binding for therapeutic efficacy are now frontline treatments for multiple diseases. However, substantially fewer development efforts are focused on the FcγRs, despite accounting for half of the antibody-receptor complex. The recent success of engineered cell-based immunotherapies now provides a mechanism to introduce modified FcγRs into the clinic. FcγRs are highly heterogeneous because of multiple functionally distinct alleles for many genes, the presence of membrane-tethered and soluble forms, and a high degree of post-translational modification, notably asparagine-linked glycans. One significant factor limiting FcγR improvement is the fundamental lack of knowledge regarding endogenous receptor forms present in the human body. This review describes the composition of FcγRs isolated from primary human leukocytes, summarizes recent efforts to engineer FcγRs, and concludes with a description of potential FcγR features to enrich for enhanced function. Further understanding FcγR biology could accelerate the development of new clinical therapies targeting immune-related disease.
Collapse
Affiliation(s)
- Adam W Barb
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
8
|
Tang JJJ, Sung AP, Guglielmo MJ, Navarrete-Galvan L, Redelman D, Smith-Gagen J, Hudig D. Natural Killer (NK) Cell Expression of CD2 as a Predictor of Serial Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC). Antibodies (Basel) 2020; 9:antib9040054. [PMID: 33081115 PMCID: PMC7709134 DOI: 10.3390/antib9040054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities for IgG. CD2, a costimulatory protein, associates with CD16A and can augment CD16A-signaling. Pore-forming perforin is essential for rapid NK-mediated killing. NK cells were monitored for their ADCC serial killing frequency (KF). KF is the average number of target cells killed per cell by a cytotoxic cell population. KF comparisons were made at 1:4 CD16pos NK effector:target ratios. ADCC was toward Daudi cells labeled with 51Cr and obinutuzumab anti-CD20 antibody. CD16A genotypes were determined by DNA sequencing. CD2, CD16A, and perforin expression was monitored by flow cytometry. Serial killing KFs varied two-fold among 24 donors and were independent of CD16A genotypes and perforin levels. However, high percentages of CD2pos of the CD16Apos NK cells and high levels of CD16A were associated with high KFs. ROC analysis indicated that the %CD2pos of CD16Apos NK cells can predict KFs. In conclusion, the extent of serial ADCC varies significantly among donors and appears predictable by the CD2posCD16Apos NK phenotype.
Collapse
Affiliation(s)
- Jennifer J.-J. Tang
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Alexander P. Sung
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Michael J. Guglielmo
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Lydia Navarrete-Galvan
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Doug Redelman
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Dorothy Hudig
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
- Correspondence: ; Tel.: +1-775-784-4430
| |
Collapse
|
9
|
Uribe-Querol E, Rosales C. Phagocytosis: Our Current Understanding of a Universal Biological Process. Front Immunol 2020; 11:1066. [PMID: 32582172 PMCID: PMC7280488 DOI: 10.3389/fimmu.2020.01066] [Citation(s) in RCA: 278] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022] Open
Abstract
Phagocytosis is a cellular process for ingesting and eliminating particles larger than 0.5 μm in diameter, including microorganisms, foreign substances, and apoptotic cells. Phagocytosis is found in many types of cells and it is, in consequence an essential process for tissue homeostasis. However, only specialized cells termed professional phagocytes accomplish phagocytosis with high efficiency. Macrophages, neutrophils, monocytes, dendritic cells, and osteoclasts are among these dedicated cells. These professional phagocytes express several phagocytic receptors that activate signaling pathways resulting in phagocytosis. The process of phagocytosis involves several phases: i) detection of the particle to be ingested, ii) activation of the internalization process, iii) formation of a specialized vacuole called phagosome, and iv) maturation of the phagosome to transform it into a phagolysosome. In this review, we present a general view of our current understanding on cells, phagocytic receptors and phases involved in phagocytosis.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
10
|
Kerntke C, Nimmerjahn F, Biburger M. There Is (Scientific) Strength in Numbers: A Comprehensive Quantitation of Fc Gamma Receptor Numbers on Human and Murine Peripheral Blood Leukocytes. Front Immunol 2020; 11:118. [PMID: 32117269 PMCID: PMC7013094 DOI: 10.3389/fimmu.2020.00118] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Antibodies are essential mediators of immunological defense mechanisms, are clinically used as therapeutic agents, but are also functionally involved in various immune-mediated disorders. Whereas IgG antibodies accomplish some of their biological tasks autonomously, many functions depend on their binding to activating and inhibitory Fcγ receptors (FcγR). From a qualitative point of view expression patterns of FcγR on immunologically relevant cell types are well-characterized both for mice and humans. Surprisingly, however, there is only quite limited information available on actual quantities of FcγR expressed by the different leukocyte populations. In this study we provide a comprehensive data set assessing quantitatively how many individual human and mouse FcγRs are expressed on B cells, NK cells, eosinophils, neutrophils, basophils and both classical, and non-classical monocytes under steady state conditions. Moreover, among human donors we found two groups with different expression levels of the inhibitory FcγRIIb on monocytes which appears to correlate with haplotypes of the activating FcγRIIIa.
Collapse
Affiliation(s)
- Christina Kerntke
- Division of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Biburger
- Division of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
The Other Side of the Coin: Anti-inflammatory Antibody Therapy for Infectious Diseases. Infect Immun 2020; 88:IAI.00844-19. [PMID: 31712268 DOI: 10.1128/iai.00844-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The inflammatory response to the fungus Pneumocystis jirovecii plays a central role in the respiratory failure associated with Pneumocystis pneumonia. To help ameliorate the inflammatory response, corticosteroids are used as an adjuvant to standard antimicrobial therapy. Corticosteroids, however, can have a wide range of effects (including deleterious effects) on the host immune response. To date, pathogen-specific antibody therapy has primarily been developed for both its direct antimicrobial activity (e.g., toxin and viral neutralization) and its ability to enhance the antimicrobial activity of the host immune response via effector cells, like macrophages and neutrophils. In this issue of Infection and Immunity, Hoy et al. (Z. Hoy, T. W. Wright, M. Elliott, J. Malone, et al., Infect Immun 88:e00640-19, 2020, https://doi.org/10.1128/IAI.00640-19) report on a surprising application of Pneumocystis-specific antibody therapy in treating disease by decreasing the inflammatory response. This effect appears to occur as a result of an enhanced phagocytic activity within the lung and an associated alteration in the macrophage phenotype. This study adds insight into our understanding of antibody activity and highlights the possibility of using antibody therapy to limit inflammation for other infectious diseases in which inflammatory damage plays a significant role in disease pathogenesis.
Collapse
|
12
|
FcgRIII Deficiency and FcgRIIb Defeciency Promote Renal Injury in Diabetic Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3514574. [PMID: 31534958 PMCID: PMC6724446 DOI: 10.1155/2019/3514574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/12/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023]
Abstract
The immune system is involved in the development of diabetes complications and IgG Fc gamma receptors (FcgRs) are key immune receptors responsible for the effective control of both humoral and innate immunity. We investigated the effects of members of the FcgR superfamily into both the streptozotocin plus high fat-induced type 2 diabetes and high fat diet (HFD) models. FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice had elevated levels of serum creatinine compared with wildtype (WT) diabetic mice. Renal histology of diabetic FcgRIII knockout and FcgRIIb knockout mice showed mesangial expansion and GBM thickening; the mechanistic study indicated a higher expression of TGF-β1, TNF-α, and p-NFκB-p65 compared with wild type mouse. The HFD mouse with FcgRIII knockout or FcgRIIb knockout had increased biochemical and renal injury factors, but oxLDL deposition was higher than in FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice. In vitro we further examined the mechanism by which the Fc gamma receptor promoted renal injury and transfected glomerular mesangial cells (GMCs) with FcgRI siRNA attenuated the level of TGF-β1, TNF-α expression. In summary, FcgRI knockdown downregulated kidney inflammation and fibrosis and FcgRIIb knockout accelerated inflammation, fibrosis, and the anomalous deposition of oxLDL whereas FcgRIII deficiency failed to protect kidney from diabetic renal injury. These observations suggested that FcgRs might represent a novel target for the therapeutic intervention of diabetic nephropathy.
Collapse
|
13
|
Zhang G, Bogdanova N, Gao T, Sheikh KA. Elimination of activating Fcγ receptors in spontaneous autoimmune peripheral polyneuropathy model protects from neuropathic disease. PLoS One 2019; 14:e0220250. [PMID: 31415574 PMCID: PMC6695161 DOI: 10.1371/journal.pone.0220250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/11/2019] [Indexed: 12/26/2022] Open
Abstract
Spontaneous autoimmune peripheral polyneuropathy (SAPP) is a reproducible mouse model of chronic inflammatory peripheral neuropathy in female non-obese diabetic mice deficient in co-stimulatory molecule, B7-2 (also known as CD86). There is evidence that SAPP is an interferon-γ, CD4+ T-cell-mediated disorder, with autoreactive T-cells and autoantibodies directed against myelin protein zero involved in its immunopathogenesis. Precise mechanisms leading to peripheral nerve system inflammation and nerve injury including demyelination in this model are not well defined. We examined the role of activating Fc-gamma receptors (FcγRs) by genetically ablating Fcγ-common chain (Fcer1g) shared by all activating FcγRs in the pathogenesis of this model. We have generated B7-2/ Fcer1g-double null animals for these studies and found that the neuropathic disease is substantially ameliorated in these animals as assessed by behavior, electrophysiology, immunocytochemistry, and morphometry. Our current studies focused on characterizing systemic and endoneurial inflammation in B7-2-null and B7-2/ Fcer1g-double nulls. We found that accumulation of endoneurial inflammatory cells was significantly attenuated in B7-2/ Fcer1g-double nulls compared to B7-2-single nulls. Whereas, systemically the frequency of CD4+ regulatory T cells and expression of immunosuppressive cytokine, IL-10, were significantly enhanced in B7-2/ Fcer1g-double nulls. Overall, these findings suggest that elimination of activating FcγRs modulate nerve injury by altering endoneurial and systemic inflammation. These observations raise the possibility of targeting activating FcγRs as a treatment strategy in acquired inflammatory demyelinating neuropathies.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Nataliia Bogdanova
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Tong Gao
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kazim A Sheikh
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
14
|
de Aguiar RB, de Moraes JZ. Exploring the Immunological Mechanisms Underlying the Anti-vascular Endothelial Growth Factor Activity in Tumors. Front Immunol 2019; 10:1023. [PMID: 31156623 PMCID: PMC6530399 DOI: 10.3389/fimmu.2019.01023] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Several studies report the key role of the vascular endothelial growth factor (VEGF) signaling on angiogenesis and on tumor growth. This has led to the development of a number of VEGF-targeted agents to treat cancer patients by disrupting the tumor blood vessel supply. Of them, bevacizumab, an FDA-approved humanized monoclonal antibody against VEGF, is the most promising. Although the use of antibodies targeting the VEGF pathway has shown clinical benefits associated with a reduction in the tumor blood vessel density, the inhibition of VEGF-driven vascular effects is only part of the functional mechanism of these therapeutic agents in the tumor ecosystem. Compelling reports have demonstrated that VEGF confers, in addition to the activation of angiogenesis-related processes, immunosuppressive properties in tumors. It is also known that structural remodeling of the tumor blood vessel bed by anti-VEGF approaches affect the influx and activation of immune cells into tumors, which might influence the therapeutic results. Besides that, part of the therapeutic effects of antiangiogenic antibodies, including their role in the tumor vascular network, might be triggered by Fc receptors in an antigen-independent manner. In this mini-review, we explore the role of VEGF inhibitors in the tumor microenvironment with focus on the immune system, discussing around the functional contribution of both bevacizumab's Fab and Fc domains to the therapeutic results and the combination of bevacizumab therapy with other immune-stimulatory settings, including adjuvant-based vaccine approaches.
Collapse
|
15
|
Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies. J Immunol Res 2019; 2019:1356540. [PMID: 31111075 PMCID: PMC6487120 DOI: 10.1155/2019/1356540] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 01/18/2023] Open
Abstract
Infectious diseases caused by pathogens including viruses, bacteria, fungi, and parasites are ranked as the second leading cause of death worldwide by the World Health Organization. Despite tremendous improvements in global public health since 1950, a number of challenges remain to either prevent or eradicate infectious diseases. Many pathogens can cause acute infections that are effectively cleared by the host immunity, but a subcategory of these pathogens called "intracellular pathogens" can establish persistent and sometimes lifelong infections. Several of these intracellular pathogens manage to evade the host immune monitoring and cause disease by replicating inside the host cells. These pathogens have evolved diverse immune escape strategies and overcome immune responses by residing and multiplying inside host immune cells, primarily macrophages. While these intracellular pathogens that cause persistent infections are phylogenetically diverse and engage in diverse immune evasion and persistence strategies, they share common pathogen type-specific mechanisms during host-pathogen interaction inside host cells. Likewise, the host immune system is also equipped with a diverse range of effector functions to fight against the establishment of pathogen persistence and subsequent host damage. This article provides an overview of the immune effector functions used by the host to counter pathogens and various persistence strategies used by intracellular pathogens to counter host immunity, which enables their extended period of colonization in the host. The improved understanding of persistent intracellular pathogen-derived infections will contribute to develop improved disease diagnostics, therapeutics, and prophylactics.
Collapse
|
16
|
Lai JY, Klatt S, Lim TS. Potential application of Leishmania tarentolae as an alternative platform for antibody expression. Crit Rev Biotechnol 2019; 39:380-394. [DOI: 10.1080/07388551.2019.1566206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
| | - Stephan Klatt
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
17
|
Kumar NN, Pizzo ME, Nehra G, Wilken-Resman B, Boroumand S, Thorne RG. Passive Immunotherapies for Central Nervous System Disorders: Current Delivery Challenges and New Approaches. Bioconjug Chem 2018; 29:3937-3966. [PMID: 30265523 PMCID: PMC7234797 DOI: 10.1021/acs.bioconjchem.8b00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Passive immunotherapy, i.e., the administration of exogenous antibodies that recognize a specific target antigen, has gained significant momentum as a potential treatment strategy for several central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and brain cancer, among others. Advances in antibody engineering to create therapeutic antibody fragments or antibody conjugates have introduced new strategies that may also be applied to treat CNS disorders. However, drug delivery to the CNS for antibodies and other macromolecules has thus far proven challenging, due in large part to the blood-brain barrier and blood-cerebrospinal fluid barriers that greatly restrict transport of peripherally administered molecules from the systemic circulation into the CNS. Here, we summarize the various passive immunotherapy approaches under study for the treatment of CNS disorders, with a primary focus on disease-specific and target site-specific challenges to drug delivery and new, cutting edge methods.
Collapse
Affiliation(s)
- Niyanta N. Kumar
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Michelle E. Pizzo
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Geetika Nehra
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Brynna Wilken-Resman
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Sam Boroumand
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Robert G. Thorne
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Neuroscience Training Program & Center for
Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705, United
States
- Cellular and Molecular Pathology Graduate Training Program,
University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
18
|
Di Ceglie I, Ascone G, Cremers NAJ, Sloetjes AW, Walgreen B, Vogl T, Roth J, Verbeek JS, van de Loo FAJ, Koenders MI, van der Kraan PM, Blom AB, van den Bosch MHJ, van Lent PLEM. Fcγ receptor-mediated influx of S100A8/A9-producing neutrophils as inducer of bone erosion during antigen-induced arthritis. Arthritis Res Ther 2018; 20:80. [PMID: 29720243 PMCID: PMC5932875 DOI: 10.1186/s13075-018-1584-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/28/2018] [Indexed: 01/01/2023] Open
Abstract
Background Osteoclast-mediated bone erosion is a central feature of rheumatoid arthritis (RA). Immune complexes, present in a large percentage of patients, bind to Fcγ receptors (FcγRs), thereby modulating the activity of immune cells. In this study, we investigated the contribution of FcγRs, and FcγRIV in particular, during antigen-induced arthritis (AIA). Methods AIA was induced in knee joints of wild-type (WT), FcγRI,II,III−/−, and FcγRI,II,III,IV−/− mice. Bone destruction, numbers of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts, and inflammation were evaluated using histology; expression of the macrophage marker F4/80, neutrophil marker NIMPR14, and alarmin S100A8 was evaluated using immunohistochemistry. The percentage of osteoclast precursors in the bone marrow was determined using flow cytometry. In vitro osteoclastogenesis was evaluated with TRAP staining, and gene expression was assessed using real-time PCR. Results FcγRI,II,III,IV−/− mice showed decreased bone erosion compared with WT mice during AIA, whereas both the humoral and cellular immune responses against methylated bovine serum albumin were not impaired in FcγRI,II,III,IV−/− mice. The percentage of osteoclast precursors in the bone marrow of arthritic mice and their ability to differentiate into osteoclasts in vitro were comparable between FcγRI,II,III,IV−/− and WT mice. In line with these observations, numbers of TRAP+ osteoclasts on the bone surface during AIA were comparable between the two groups. Inflammation, a process that strongly activates osteoclast activity, was reduced in FcγRI,II,III,IV−/− mice, and of note, mainly decreased numbers of neutrophils were present in the joint. In contrast to FcγRI,II,III,IV−/− mice, AIA induction in knee joints of FcγRI,II,III−/− mice resulted in increased bone erosion, inflammation, and numbers of neutrophils, suggesting a crucial role for FcγRIV in the joint pathology by the recruitment of neutrophils. Finally, significant correlations were found between bone erosion and the number of neutrophils present in the joint as well as between bone erosion and the number of S100A8-positive cells, with S100A8 being an alarmin strongly produced by neutrophils that stimulates osteoclast resorbing activity. Conclusions FcγRs play a crucial role in the development of bone erosion during AIA by inducing inflammation. In particular, FcγRIV mediates bone erosion in AIA by inducing the influx of S100A8/A9-producing neutrophils into the arthritic joint. Electronic supplementary material The online version of this article (10.1186/s13075-018-1584-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irene Di Ceglie
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Giuliana Ascone
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Niels A J Cremers
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Annet W Sloetjes
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Birgitte Walgreen
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - J Sjef Verbeek
- Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Fons A J van de Loo
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud university medical center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Besson L, Charrier E, Karlin L, Allatif O, Marçais A, Rouzaire P, Belmont L, Attal M, Lombard C, Salles G, Walzer T, Viel S. One-Year Follow-Up of Natural Killer Cell Activity in Multiple Myeloma Patients Treated With Adjuvant Lenalidomide Therapy. Front Immunol 2018; 9:704. [PMID: 29706958 PMCID: PMC5908898 DOI: 10.3389/fimmu.2018.00704] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/22/2018] [Indexed: 01/22/2023] Open
Abstract
Multiple myeloma (MM) is a proliferation of tumoral plasma B cells that is still incurable. Natural killer (NK) cells can recognize and kill MM cells in vitro and can limit MM growth in vivo. Previous reports have shown that NK cell function is impaired during MM progression and suggested that treatment with immunomodulatory drugs (IMIDs) such as lenalidomide (LEN) could enhance it. However, the effects of IMIDs on NK cells have been tested mostly in vitro or in preclinical models and supporting evidence of their effect in vivo in patients is lacking. Here, we monitored NK cell activity in blood samples from 10 MM patients starting after frontline induction chemotherapy (CTX) consisting either of association of bortezomib–lenalidomide–dexamethasone (Velcade Revlimid Dexamethasone) or autologous stem-cell transplantation (SCT). We also monitored NK cell activity longitudinally each month during 1 year, after maintenance therapy with LEN. Following frontline chemotherapy, peripheral NK cells displayed a very immature phenotype and retained poor reactivity toward target cells ex vivo. Upon maintenance treatment with LEN, we observed a progressive normalization of NK cell maturation, likely caused by discontinuation of chemotherapy. However, LEN treatment neither activated NK cells nor improved their capacity to degranulate or to secrete IFN-γ or MIP1-β following stimulation with MHC-I-deficient or antibody-coated target cells. Upon LEN discontinuation, there was no reduction of NK cell effector function either. These results caution against the use of LEN as single therapy to improve NK cell activity in patients with cancer and call for more preclinical assessments of the potential of IMIDs in NK cell activation.
Collapse
Affiliation(s)
- Laurie Besson
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France.,Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Emily Charrier
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France.,Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Lionel Karlin
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Service d'Hematologie, Pierre-Benite, Universite Claude Bernard Lyon 1, Lyon, France
| | - Omran Allatif
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France
| | - Paul Rouzaire
- Service d'Immunologie, CHU de Clermont-Ferrand, équipe ERTICa EA4677, Université d'Auvergne, Clermont-Ferrand, France
| | - Lucie Belmont
- Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Michel Attal
- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Christine Lombard
- Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Gilles Salles
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Service d'Hematologie, Pierre-Benite, Universite Claude Bernard Lyon 1, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France
| | - Sébastien Viel
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Lyon, France.,INSERM, U1111, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France.,Université Lyon 1, Lyon, France.,CNRS, UMR5308, Lyon, France.,Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| |
Collapse
|
20
|
Wang J, Li Z, Xu L, Yang H, Liu W. Transmembrane domain dependent inhibitory function of FcγRIIB. Protein Cell 2018; 9:1004-1012. [PMID: 29497990 PMCID: PMC6251803 DOI: 10.1007/s13238-018-0509-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/22/2017] [Indexed: 01/26/2023] Open
Abstract
FcγRIIB, the only inhibitory IgG Fc receptor, functions to suppress the hyper-activation of immune cells. Numerous studies have illustrated its inhibitory function through the ITIM motif in the cytoplasmic tail of FcγRIIB. However, later studies revealed that in addition to the ITIM, the transmembrane (TM) domain of FcγRIIB is also indispensable for its inhibitory function. Indeed, recent epidemiological studies revealed that a non-synonymous single nucleotide polymorphism (rs1050501) within the TM domain of FcγRIIB, responsible for the I232T substitution, is associated with the susceptibility to systemic lupus erythematosus (SLE). In this review, we will summarize these epidemiological and functional studies of FcγRIIB-I232T in the past few years, and will further discuss the mechanisms accounting for the functional loss of FcγRIIB-I232T. Our review will help the reader gain a deeper understanding of the importance of the TM domain in mediating the inhibitory function of FcγRIIB and may provide insights to a new therapeutic target for the associated diseases.
Collapse
Affiliation(s)
- Junyi Wang
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zongyu Li
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Liling Xu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, 400 Technology Square, Cambridge, MA, 02139, USA.
| | - Hengwen Yang
- The First Affiliate Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, 510632, China.
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
21
|
Abstract
Germinal centers (GCs) are dynamic microenvironments that form in the secondary lymphoid organs and generate somatically mutated high-affinity antibodies necessary to establish an effective humoral immune response. Tight regulation of GC responses is critical for maintaining self-tolerance. GCs can arise in the absence of purposeful immunization or overt infection (called spontaneous GCs, Spt-GCs). In autoimmune-prone mice and patients with autoimmune disease, aberrant regulation of Spt-GCs is thought to promote the development of somatically mutated pathogenic autoantibodies and the subsequent development of autoimmunity. The mechanisms that control the formation of Spt-GCs and promote systemic autoimmune diseases remain an open question and the focus of ongoing studies. Here, we discuss the most current studies on the role of Spt-GCs in autoimmunity.
Collapse
Affiliation(s)
- Phillip P Domeier
- a Department of Microbiology and Immunology, Penn State College of Medicine , USA
| | - Stephanie L Schell
- a Department of Microbiology and Immunology, Penn State College of Medicine , USA
| | - Ziaur S M Rahman
- a Department of Microbiology and Immunology, Penn State College of Medicine , USA
| |
Collapse
|
22
|
Uribe-Querol E, Rosales C. Control of Phagocytosis by Microbial Pathogens. Front Immunol 2017; 8:1368. [PMID: 29114249 PMCID: PMC5660709 DOI: 10.3389/fimmu.2017.01368] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
Phagocytosis is a fundamental process of cells to capture and ingest foreign particles. Small unicellular organisms such as free-living amoeba use this process to acquire food. In pluricellular organisms, phagocytosis is a universal phenomenon that all cells are able to perform (including epithelial, endothelial, fibroblasts, etc.), but some specialized cells (such as neutrophils and macrophages) perform this very efficiently and were therefore named professional phagocytes by Rabinovitch. Cells use phagocytosis to capture and clear all particles larger than 0.5 µm, including pathogenic microorganisms and cellular debris. Phagocytosis involves a series of steps from recognition of the target particle, ingestion of it in a phagosome (phagocytic vacuole), maturation of this phagosome into a phagolysosome, to the final destruction of the ingested particle in the robust antimicrobial environment of the phagolysosome. For the most part, phagocytosis is an efficient process that eliminates invading pathogens and helps maintaining homeostasis. However, several pathogens have also evolved different strategies to prevent phagocytosis from proceeding in a normal way. These pathogens have a clear advantage to perpetuate the infection and continue their replication. Here, we present an overview of the phagocytic process with emphasis on the antimicrobial elements professional phagocytes use. We also summarize the current knowledge on the microbial strategies different pathogens use to prevent phagocytosis either at the level of ingestion, phagosome formation, and maturation, and even complete escape from phagosomes.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
23
|
Phagocytosis: A Fundamental Process in Immunity. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9042851. [PMID: 28691037 PMCID: PMC5485277 DOI: 10.1155/2017/9042851] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/18/2017] [Indexed: 01/12/2023]
Abstract
One hundred years have passed since the death of Élie Metchnikoff (1845-1916). He was the first to observe the uptake of particles by cells and realized the importance of this process for the host response to injury and infection. He also was a strong advocate of the role of phagocytosis in cellular immunity, and with this he gave us the basis for our modern understanding of inflammation and the innate and acquired immune responses. Phagocytosis is an elegant but complex process for the ingestion and elimination of pathogens, but it is also important for the elimination of apoptotic cells and hence fundamental for tissue homeostasis. Phagocytosis can be divided into four main steps: (i) recognition of the target particle, (ii) signaling to activate the internalization machinery, (iii) phagosome formation, and (iv) phagolysosome maturation. In recent years, the use of new tools of molecular biology and microscopy has provided new insights into the cellular mechanisms of phagocytosis. In this review, we present a general view of our current knowledge on phagocytosis. We emphasize novel molecular findings, particularly on phagosome formation and maturation, and discuss aspects that remain incompletely understood.
Collapse
|
24
|
Pearson RM, Casey LM, Hughes KR, Miller SD, Shea LD. In vivo reprogramming of immune cells: Technologies for induction of antigen-specific tolerance. Adv Drug Deliv Rev 2017; 114:240-255. [PMID: 28414079 PMCID: PMC5582017 DOI: 10.1016/j.addr.2017.04.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/01/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023]
Abstract
Technologies that induce antigen-specific immune tolerance by mimicking naturally occurring mechanisms have the potential to revolutionize the treatment of many immune-mediated pathologies such as autoimmunity, allograft rejection, and allergy. The immune system intrinsically has central and peripheral tolerance pathways for eliminating or modulating antigen-specific responses, which are being exploited through emerging technologies. Antigen-specific tolerogenic responses have been achieved through the functional reprogramming of antigen-presenting cells or lymphocytes. Alternatively, immune privileged sites have been mimicked using biomaterial scaffolds to locally suppress immune responses and promote long-term allograft survival. This review describes natural mechanisms of peripheral tolerance induction and the various technologies being developed to achieve antigen-specific immune tolerance in vivo. As currently approved therapies are non-specific and carry significant associated risks, these therapies offer significant progress towards replacing systemic immune suppression with antigen-specific therapies to curb aberrant immune responses.
Collapse
Affiliation(s)
- Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Liam M Casey
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 E. Chicago Avenue, Chicago, IL 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA; Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA.
| |
Collapse
|
25
|
McMichael EL, Courtney NB, Duggan MC, Wesolowski R, Quiroga D, Kondadasula SV, Atwal LS, Bhave N, Luedke E, Jaime-Ramirez AC, Campbell AR, Mo X, Byrd JC, Carson Iii WE. Activation of the FcgammaReceptorIIIa on human natural killer cells leads to increased expression of functional interleukin-21 receptor. Oncoimmunology 2017. [PMID: 28638738 DOI: 10.1080/2162402x.2017.1312045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Natural killer (NK) cells are innate immune effector cells that play a crucial role in immune surveillance and the destruction of cancer cells. NK cells express a low-affinity receptor for the Fc or constant region of immunoglobulin G (FcγRIIIa) and multiple cytokine receptors that respond to antibody-coated targets and cytokines in the tumor microenvironment. In the present work, microarray gene expression analysis revealed that the IL-21 receptor (IL-21R) was strongly upregulated following FcR stimulation. The IL-21R was found to be upregulated on FcR-stimulated NK cells at the transcript level as determined by reverse transcription polymerase chain reaction (RT-PCR). Immunoblot analysis revealed that protein expression of the IL-21R peaked at 8 h post-stimulation of the FcR. Inhibition of the mitogen-activated protein kinase (MAPK) pathway downstream of the FcR blocked the induction of IL-21R expression. Increased expression of the IL-21R sensitized NK cells to IL-21 stimulation, as treatment of FcR-stimulated NK cells led to significantly increased phosphorylation of STAT1 and STAT3, as measured by intracellular flow cytometry and immunoblot analysis. Following FcR-stimulation, IL-21-activated NK cells were better able to mediate the lysis of trastuzumab-coated human epidermal growth factor receptor 2 (HER2+) SK-BR-3 tumor cells as compared to control-treated cells. Likewise, IL-21-induced NK cell secretion of IFNγ following exposure to antibody-coated tumor cells was enhanced following FcR-stimulation. The analysis of NK cells from patients receiving trastuzumab therapy for HER2+ cancer exhibited increased levels of the IL-21R following the administration of antibody suggesting that the presence of monoclonal antibody-coated tumor cells in vivo can stimulate the increased expression of IL-21R on NK cells.
Collapse
Affiliation(s)
| | | | - Megan C Duggan
- Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, Columbus, OH, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, Department of Internal Medicine, Columbus, OH, USA
| | | | | | - Neela Bhave
- Comprehensive Cancer Center, Columbus, OH, USA
| | - Eric Luedke
- Department of Surgery, Division of Surgical Oncology, Columbus, OH, USA
| | | | - Amanda R Campbell
- Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US.,Medical Scientist Training Program, College of Medicine, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, Columbus, OH, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, Columbus, OH, USA
| | - William E Carson Iii
- Department of Surgery, Division of Surgical Oncology, Columbus, OH, USA.,Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA.,Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US
| |
Collapse
|
26
|
Rosales C. Fcγ Receptor Heterogeneity in Leukocyte Functional Responses. Front Immunol 2017; 8:280. [PMID: 28373871 PMCID: PMC5357773 DOI: 10.3389/fimmu.2017.00280] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/27/2017] [Indexed: 01/12/2023] Open
Abstract
Antibodies participate in defense of the organism from all types of pathogens, including viruses, bacteria, fungi, and protozoa. IgG antibodies recognize their associated antigen via their two Fab portions and are in turn recognized though their Fc portion by specific Fcγ receptors (FcγRs) on the membrane of immune cells. Multiple types and polymorphic variants of FcγR exist. These receptors are expressed in many cells types and are also redundant in inducing cell responses. Crosslinking of FcγR on the surface of leukocytes activates several effector functions aimed toward the destruction of pathogens and the induction of an inflammatory response. In the past few years, new evidence on how the particular IgG subclass and the glycosylation pattern of the antibody modulate the IgG-FcγR interaction has been presented. Despite these advances, our knowledge of what particular effector function is activated in a certain cell and in response to a specific type of FcγR remains very limited today. On one hand, each immune cell could be programmed to perform a particular cell function after FcγR crosslinking. On the other, each FcγR could activate a particular signaling pathway leading to a unique cell response. In this review, I describe the main types of FcγRs and our current view of how particular FcγRs activate various signaling pathways to promote unique leukocyte functions.
Collapse
Affiliation(s)
- Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
27
|
Hierarchical and Redundant Roles of Activating FcγRs in Protection against Influenza Disease by M2e-Specific IgG1 and IgG2a Antibodies. J Virol 2017; 91:JVI.02500-16. [PMID: 28077656 DOI: 10.1128/jvi.02500-16] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/01/2023] Open
Abstract
The ectodomain of matrix protein 2 is a universal influenza A virus vaccine candidate that provides protection through antibody-dependent effector mechanisms. Here we compared the functional engagement of Fcγ receptor (FcγR) family members by two M2e-specific monoclonal antibodies (MAbs), MAb 37 (IgG1) and MAb 65 (IgG2a), which recognize a similar epitope in M2e with similar affinities. The binding of MAb 65 to influenza A virus-infected cells triggered all three activating mouse Fcγ receptors in vitro, whereas MAb 37 activated only FcγRIII. The passive transfer of MAb 37 or MAb 65 in wild-type, Fcer1g-/-, Fcgr3-/-, and Fcgr1-/-Fcgr3-/- BALB/c mice revealed the importance of these receptors for protection against influenza A virus challenge, with a clear requirement of FcγRIII for IgG1 MAb 37 being found. We also report that FcγRIV contributes to protection by M2e-specific IgG2a antibodies.IMPORTANCE There is increased awareness that protection by antibodies directed against viral antigens is also mediated by the Fc domain of these antibodies. These Fc-mediated effector functions are often missed in clinical assays, which are used, for example, to define correlates of protection induced by vaccines. The use of antibodies to prevent and treat infectious diseases is on the rise and has proven to be a promising approach in our battle against newly emerging viral infections. It is now also realized that Fcγ receptors significantly enhance the in vivo protective effect of broadly neutralizing antibodies directed against the conserved parts of the influenza virus hemagglutinin. We show here that two M2e-specific monoclonal antibodies with close to identical antigen-binding specificities and affinities have a very different in vivo protective potential that is controlled by their capacity to interact with activating Fcγ receptors.
Collapse
|
28
|
Kasturirangan S, Rainey GJ, Xu L, Wang X, Portnoff A, Chen T, Fazenbaker C, Zhong H, Bee J, Zeng Z, Jenne C, Wu H, Gao C. Targeted Fcγ Receptor (FcγR)-mediated Clearance by a Biparatopic Bispecific Antibody. J Biol Chem 2017; 292:4361-4370. [PMID: 28100773 PMCID: PMC5354496 DOI: 10.1074/jbc.m116.770628] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/09/2017] [Indexed: 11/14/2022] Open
Abstract
Soluble ligands have commonly been targeted by antibody therapeutics for cancers and other diseases. Although monoclonal antibodies targeting such ligands can block their interactions with their cognate receptors, they can also significantly increase the half-life of their ligands by FcRn-mediated antibody recycling, thereby evading ligand renal clearance and requiring increasingly high antibody doses to neutralize the increasing pool of target. To overcome this issue, we generated a bispecific/biparatopic antibody (BiSAb) that targets two different epitopes on IL-6 to block IL-6-mediated signaling. The BiSAb formed large immune complexes with IL-6 that can bind Fcγ receptors on phagocytic cells and are rapidly internalized. In addition, rapid clearance of the BiSAb·IL-6 complex was observed in mice while the parental antibodies prolonged the serum half-life of IL-6. Intravital imaging of the liver in mice confirmed that the rapid clearance of these large immune complexes was associated with Fcγ receptor-dependent binding to Kupffer cells in the liver. The approach described here provides a general strategy for therapeutic antibodies with the ability to not only neutralize but also actively drive clearance of their soluble antigens.
Collapse
Affiliation(s)
| | - G Jonah Rainey
- From the Departments of Antibody Discovery and Protein Engineering
| | - Linda Xu
- From the Departments of Antibody Discovery and Protein Engineering
| | - Xinwei Wang
- From the Departments of Antibody Discovery and Protein Engineering
| | - Alyse Portnoff
- From the Departments of Antibody Discovery and Protein Engineering
| | | | | | | | - Jared Bee
- Analytical Biotechnology, Medimmune LLC, Gaithersburg, Maryland 20878 and
| | - Zhutian Zeng
- the Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Craig Jenne
- the Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Herren Wu
- From the Departments of Antibody Discovery and Protein Engineering
| | - Changshou Gao
- From the Departments of Antibody Discovery and Protein Engineering,
| |
Collapse
|
29
|
Burton OT, Stranks AJ, Tamayo JM, Koleoglou KJ, Schwartz LB, Oettgen HC. A humanized mouse model of anaphylactic peanut allergy. J Allergy Clin Immunol 2017; 139:314-322.e9. [PMID: 27417025 PMCID: PMC5145786 DOI: 10.1016/j.jaci.2016.04.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 03/08/2016] [Accepted: 04/12/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Food allergy is a growing health problem with very limited treatment options. Investigation of the immunologic pathways underlying allergic sensitization to foods in humans has been greatly constrained by the limited availability of intestinal tissue and gut-resident immune cells. Although mouse models have offered insights into pathways of food sensitization, differences between rodent and human immune physiology limit the extension of these findings to our understanding of human disease. OBJECTIVE We sought to develop a strategy for the generation of mice with humanized adaptive immune systems, complete with tissue engraftment by human mast cells that are competent to mount specific IgE-mediated responses and drive systemic anaphylaxis on ingestion challenge. METHODS Nonobese diabetic severe combined immunodeficient mice lacking the cytokine receptor common gamma chain (γc-/-) and carrying a human stem cell factor transgene were engrafted with human hematopoietic stem cells. The impact of peanut (PN) feeding and IgE neutralization on the development of immune responses, mast cell homeostasis, and anaphylactic food allergy was assessed in these animals. RESULTS Humanized nonobese diabetic severe combined immunodeficient common gamma chain-deficient stem cell factor (huNSG) mice exhibited robust engraftment with functional human T and B lymphocytes and human mast cells were found in significant numbers in their tissues, including the intestinal mucosa. Following gavage feeding with PN, they mounted specific antibody responses, including PN-specific IgE. When enterally challenged with PN, they exhibited mast-cell-mediated systemic anaphylaxis, as indicated by hypothermia and increases in plasma tryptase levels. Anti-IgE (omalizumab) treatment ablated this anaphylactic response. CONCLUSIONS huNSG mice provide a novel tool for studying food allergy and IgE-mediated anaphylaxis.
Collapse
Affiliation(s)
- Oliver T Burton
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Amanda J Stranks
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Jaciel M Tamayo
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Kyle J Koleoglou
- Department of Medicine, Boston Children's Hospital, Boston, Mass
| | - Lawrence B Schwartz
- Division of Rheumatology, Allergy and Immunology and the Department of Internal Medicine, Virginia Commonwealth University, Richmond, Va
| | - Hans C Oettgen
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| |
Collapse
|
30
|
Jacobsen FW, Stevenson R, Li C, Salimi-Moosavi H, Liu L, Wen J, Luo Q, Daris K, Buck L, Miller S, Ho SY, Wang W, Chen Q, Walker K, Wypych J, Narhi L, Gunasekaran K. Engineering an IgG Scaffold Lacking Effector Function with Optimized Developability. J Biol Chem 2016; 292:1865-1875. [PMID: 27994062 DOI: 10.1074/jbc.m116.748525] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/11/2016] [Indexed: 01/12/2023] Open
Abstract
IgG isotypes can differentially bind to Fcγ receptors and complement, making the selection of which isotype to pursue for development of a particular therapeutic antibody important in determining the safety and efficacy of the drug. IgG2 and IgG4 isotypes have significantly lower binding affinity to Fcγ receptors. Recent evidence suggests that the IgG2 isotype is not completely devoid of effector function, whereas the IgG4 isotype can undergo in vivo Fab arm exchange leading to bispecific antibody and off-target effects. Here an attempt was made to engineer an IgG1-based scaffold lacking effector function but with stability equivalent to that of the parent IgG1. Care was taken to ensure that both stability and lack of effector function was achieved with a minimum number of mutations. Among the Asn297 mutants that result in lack of glycosylation and thus loss of effector function, we demonstrate that the N297G variant has better stability and developability compared with the N297Q or N297A variants. To further improve the stability of N297G, we introduced a novel engineered disulfide bond at a solvent inaccessible location in the CH2 domain. The resulting scaffold has stability greater than or equivalent to that of the parental IgG1 scaffold. Extensive biophysical analyses and pharmacokinetic (PK) studies in mouse, rat, and monkey further confirmed the developability of this unique scaffold, and suggest that it could be used for all Fc containing therapeutics (e.g. antibodies, bispecific antibodies, and Fc fusions) requiring lack of effector function or elimination of binding to Fcγ receptors.
Collapse
Affiliation(s)
- Frederick W Jacobsen
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320.
| | - Riki Stevenson
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Cynthia Li
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Hossein Salimi-Moosavi
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Ling Liu
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Jie Wen
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Quanzhou Luo
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Kristine Daris
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Lynette Buck
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Sterling Miller
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Shu-Yin Ho
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Wei Wang
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Qing Chen
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Kenneth Walker
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Jette Wypych
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Linda Narhi
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320
| | - Kannan Gunasekaran
- From the Biologics Optimization-Therapeutic Discovery, Clinical Immunology, and Process Development, Amgen Inc., Thousand Oaks, California 91320.
| |
Collapse
|
31
|
Xu L, Xia M, Guo J, Sun X, Li H, Xu C, Gu X, Zhang H, Yi J, Fang Y, Xie H, Wang J, Shen Z, Xue B, Sun Y, Meckel T, Chen YH, Hu Z, Li Z, Xu C, Gong H, Liu W. Impairment on the lateral mobility induced by structural changes underlies the functional deficiency of the lupus-associated polymorphism FcγRIIB-T232. J Exp Med 2016; 213:2707-2727. [PMID: 27799621 PMCID: PMC5110019 DOI: 10.1084/jem.20160528] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/08/2016] [Accepted: 09/28/2016] [Indexed: 12/18/2022] Open
Abstract
Xu et al. show that the lupus-associated polymorphism FcγRIIB-T232 has structural changes of the TM domain that reduces lateral mobility and inhibitory functions. FcγRIIB functions to suppress the activation of immune cells. A single-nucleotide polymorphism in the transmembrane (TM) domain of FcγRIIB, FcγRIIB-T232, is associated with lupus. In this study, we investigated the pathogenic mechanism of FcγRIIB-T232 at both functional and structural levels. Our results showed that FcγRIIB-T232 exhibited significantly reduced lateral mobility compared with FcγRIIB-I232 and was significantly less enriched into the microclusters of immune complexes (ICs) after stimulation. However, if sufficient responding time is given for FcγRIIB-T232 to diffuse and interact with the ICs, FcγRIIB-T232 can restore its inhibitory function. Moreover, substituting the FcγRIIB-T232 TM domain with that of a fast floating CD86 molecule restored both the rapid mobility and the inhibitory function, which further corroborated the importance of fast mobility for FcγRIIB to function. Mechanistically, the crippled lateral mobility of FcγRIIB-T232 can be explained by the structural changes of the TM domain. Both atomistic simulations and nuclear magnetic resonance measurement indicated that the TM helix of FcγRIIB-T232 exhibited a more inclined orientation than that of FcγRIIB-I232, thus resulting in a longer region embedded in the membrane. Therefore, we conclude that the single-residue polymorphism T232 enforces the inclination of the TM domain and thereby reduces the lateral mobility and inhibitory functions of FcγRIIB.
Collapse
Affiliation(s)
- Liling Xu
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Mengdie Xia
- Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jun Guo
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing 100044, China
| | - Hua Li
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenguang Xu
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaomei Gu
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haowen Zhang
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junyang Yi
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yan Fang
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hengyi Xie
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Wang
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhixun Shen
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Boxin Xue
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yujie Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Tobias Meckel
- Membrane Dynamics, Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Ying-Hua Chen
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhibin Hu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing 100044, China
| | - Chenqi Xu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China .,School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Haipeng Gong
- Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wanli Liu
- Ministry of Education Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
32
|
Dézsi L, Horváth Z, Vécsei L. Intravenous immunoglobulin: pharmacological properties and use in polyneuropathies. Expert Opin Drug Metab Toxicol 2016; 12:1343-1358. [PMID: 27428464 DOI: 10.1080/17425255.2016.1214715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Intravenous immunoglobulin (IVIg) is increasingly used for the treatment of autoimmune and systemic inflammatory diseases with both licensed and off-label indications. The mechanism of action is complex and not fully understood, involving the neutralization of pathological antibodies, Fc receptor blockade, complement inhibition, immunoregulation of dendritic cells, B cells and T cells and the modulation of apoptosis. Areas covered: First, this review describes the pharmacological properties of IVIg, including the composition, mechanism of action, and adverse events. The second part gives an overview of some of the immune-mediated polyneuropathies, with special focus on the pathomechanism and clinical trials assessing the efficacy of IVIg. A literature search on PubMed was performed using the terms IVIg, IVIg preparations, side effects, mechanism of action, clinical trials, GBS, CIDP. Expert opinion: Challenges associated with IVIg therapy and the treatment possibilities for immune-mediated polyneuropathies are discussed. The availability of IVIg is limited, the expenses are high, and, in several diseases, a chronic therapy is necessary to maintain the immunomodulatory effect. The better understanding of the mechanism of action of IVIg could open the possibility of the development of disease-specific, targeted immune therapies.
Collapse
Affiliation(s)
- Livia Dézsi
- a Department of Neurology , University of Szeged , Szeged , Hungary
| | - Zoltán Horváth
- a Department of Neurology , University of Szeged , Szeged , Hungary
| | - László Vécsei
- a Department of Neurology , University of Szeged , Szeged , Hungary.,b MTA-SZTE Neuroscience Research Group , Szeged , Hungary
| |
Collapse
|
33
|
Mula RVR, Machiah D, Holland L, Wang X, Parihar H, Sharma AC, Selvaraj P, Shashidharamurthy R. Immune Complex-Induced, Nitric Oxide-Mediated Vascular Endothelial Cell Death by Phagocytes Is Prevented with Decoy FcγReceptors. PLoS One 2016; 11:e0153620. [PMID: 27101012 PMCID: PMC4839578 DOI: 10.1371/journal.pone.0153620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/31/2016] [Indexed: 01/05/2023] Open
Abstract
Autoimmune vasculitis is an endothelial inflammatory disease that results from the deposition of immune-complexes (ICs) in blood vessels. The interaction between Fcgamma receptors (FcγRs) expressed on inflammatory cells with ICs is known to cause blood vessel damage. Hence, blocking the interaction of ICs and inflammatory cells is essential to prevent the IC-mediated blood vessel damage. Thus we tested if uncoupling the interaction of FcγRs and ICs prevents endothelium damage. Herein, we demonstrate that dimeric FcγR-Igs prevented nitric oxide (NO) mediated apoptosis of human umbilical vein endothelial cells (HUVECs) in an in vitro vasculitis model. Dimeric FcγR-Igs significantly inhibited the IC-induced upregulation of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) release by murine monocytic cell line. However, FcγR-Igs did not affect the exogenously added NO-induced upregulation of pro-apoptotic genes such as Bax (15 fold), Bak (35 fold), cytochrome-C (11 fold) and caspase-3 (30 fold) in HUVECs. In conclusion, these data suggest that IC-induced NO could be one of the major inflammatory mediator promoting blood vessel inflammation and endothelial cell death during IC-mediated vasculitis which can be effectively blocked by dimeric decoy FcγRs.
Collapse
Affiliation(s)
- Ramanjaneya V. R. Mula
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
| | - Deepa Machiah
- Department of Molecular Pathology Laboratory, Yerkes National Primate Research Centre, Atlanta, Georgia, United States of America
| | - Lauren Holland
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
| | - Xinyu Wang
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
| | - Harish Parihar
- Department of Pharmacy Practice, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
| | - Avadhesh C. Sharma
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Rangaiah Shashidharamurthy
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine - School of Pharmacy, Suwanee, Georgia, United States of America
- * E-mail:
| |
Collapse
|
34
|
Hickey MJ, Valenzuela NM, Reed EF. Alloantibody Generation and Effector Function Following Sensitization to Human Leukocyte Antigen. Front Immunol 2016; 7:30. [PMID: 26870045 PMCID: PMC4740371 DOI: 10.3389/fimmu.2016.00030] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/20/2016] [Indexed: 02/06/2023] Open
Abstract
Allorecognition is the activation of the adaptive immune system to foreign human leukocyte antigen (HLA) resulting in the generation of alloantibodies. Due to a high polymorphism, foreign HLA is recognized by the immune system following transplant, transfusion, or pregnancy resulting in the formation of the germinal center and the generation of long-lived alloantibody-producing memory B cells. Alloantibodies recognize antigenic epitopes displayed by the HLA molecule on the transplanted allograft and contribute to graft damage through multiple mechanisms, including (1) activation of the complement cascade resulting in the formation of the MAC complex and inflammatory anaphylatoxins, (2) transduction of intracellular signals leading to cytoskeletal rearrangement, growth, and proliferation of graft vasculature, and (3) immune cell infiltration into the allograft via FcγR interactions with the FC portion of the antibody. This review focuses on the generation of HLA alloantibody, routes of sensitization, alloantibody specificity, and mechanisms of antibody-mediated graft damage.
Collapse
Affiliation(s)
- Michelle J Hickey
- Department of Pathology and Laboratory Medicine, UCLA Immunogenetics Center, University of California Los Angeles , Los Angeles, CA , USA
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, UCLA Immunogenetics Center, University of California Los Angeles , Los Angeles, CA , USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, UCLA Immunogenetics Center, University of California Los Angeles , Los Angeles, CA , USA
| |
Collapse
|
35
|
Dashivets T, Thomann M, Rueger P, Knaupp A, Buchner J, Schlothauer T. Multi-Angle Effector Function Analysis of Human Monoclonal IgG Glycovariants. PLoS One 2015; 10:e0143520. [PMID: 26657484 PMCID: PMC4676693 DOI: 10.1371/journal.pone.0143520] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/05/2015] [Indexed: 01/26/2023] Open
Abstract
Therapeutic performance of recombinant antibodies relies on two independent mechanisms: antigen recognition and Fc-mediated antibody effector functions. Interaction of Fc-fragment with different FcR triggers antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity and determines longevity of the antibody in serum. In context of therapeutic antibodies FcγRs play the most important role. It has been demonstrated that the Fc-attached sugar moiety is essential for IgG effector functionality, dictates its affinity to individual FcγRs and determines binding to different receptor classes: activating or inhibitory. In this study, we systematically analyze effector functions of monoclonal IgG1 and its eight enzymatically engineered glycosylation variants. The analysis of interaction of glycovariants with FcRs was performed for single, as well as for antigen-bound antibodies and IgGs in a form of immune complex. In addition to functional properties we addressed impact of glycosylation on the structural properties of the tested glycovariants. We demonstrate a clear impact of glycosylation pattern on antibody stability and interaction with different FcγRs. Consistent with previous reports, deglycosylated antibodies failed to bind all Fcγ-receptors, with the exception of high affinity FcγRI. The FcγRII and FcγRIIIa binding activity of IgG1 was observed to depend on the galactosylation level, and hypergalactosylated antibodies demonstrated increased receptor interaction. Sialylation did not decrease the FcγR binding of the tested IgGs; in contrast, sialylation of antibodies improved binding to FcγRIIa and IIb. We demonstrate that glycosylation influences to some extent IgG1 interaction with FcRn. However, independent of glycosylation pattern the interaction of IgG1 with a soluble monomeric target surprisingly resulted in an impaired receptor binding. Here, we demonstrate, that immune complexes (IC), induced by multimeric ligand, compensated for the decreased affinity of target bound antibody towards FcRs, showing the importance of the IC-formation for the FcR- mediated effector functions.
Collapse
Affiliation(s)
- Tetyana Dashivets
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg, Germany
- Center for Integrated Protein Science Munich, Department Chemie, Technische Universität München, 85748, Garching, Germany
| | - Marco Thomann
- Pharma Biotech Development Penzberg, Roche Diagnostics GmbH, Penzberg, Germany
| | - Petra Rueger
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg, Germany
| | - Alexander Knaupp
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science Munich, Department Chemie, Technische Universität München, 85748, Garching, Germany
| | - Tilman Schlothauer
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg, Germany
- * E-mail:
| |
Collapse
|
36
|
Hargreaves CE, Iriyama C, Rose-Zerilli MJJ, Nagelkerke SQ, Hussain K, Ganderton R, Lee C, Machado LR, Hollox EJ, Parker H, Latham KV, Kuijpers TW, Potter KN, Coupland SE, Davies A, Stackpole M, Oates M, Pettitt AR, Glennie MJ, Cragg MS, Strefford JC. Evaluation of High-Throughput Genomic Assays for the Fc Gamma Receptor Locus. PLoS One 2015; 10:e0142379. [PMID: 26545243 PMCID: PMC4636148 DOI: 10.1371/journal.pone.0142379] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/21/2015] [Indexed: 11/18/2022] Open
Abstract
Cancer immunotherapy has been revolutionised by the use monoclonal antibodies (mAb) that function through their interaction with Fc gamma receptors (FcγRs). The low-affinity FcγR genes are highly homologous, map to a complex locus at 1p23 and harbour single nucleotide polymorphisms (SNPs) and copy number variation (CNV) that can impact on receptor function and response to therapeutic mAbs. This complexity can hinder accurate characterisation of the locus. We therefore evaluated and optimised a suite of assays for the genomic analysis of the FcγR locus amenable to peripheral blood mononuclear cells and formalin-fixed paraffin-embedded (FFPE) material that can be employed in a high-throughput manner. Assessment of TaqMan genotyping for FCGR2A-131H/R, FCGR3A-158F/V and FCGR2B-232I/T SNPs demonstrated the need for additional methods to discriminate genotypes for the FCGR3A-158F/V and FCGR2B-232I/T SNPs due to sequence homology and CNV in the region. A multiplex ligation-dependent probe amplification assay provided high quality SNP and CNV data in PBMC cases, but there was greater data variability in FFPE material in a manner that was predicted by the BIOMED-2 multiplex PCR protocol. In conclusion, we have evaluated a suite of assays for the genomic analysis of the FcγR locus that are scalable for application in large clinical trials of mAb therapy. These assays will ultimately help establish the importance of FcγR genetics in predicting response to antibody therapeutics.
Collapse
Affiliation(s)
- Chantal E. Hargreaves
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Chisako Iriyama
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Sietse Q. Nagelkerke
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX, Amsterdam, The Netherlands
| | - Khiyam Hussain
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Rosalind Ganderton
- Molecular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, United Kingdom
| | - Charlotte Lee
- Molecular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, United Kingdom
| | - Lee R. Machado
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, United Kingdom
- School of Health, University of Northampton, Northampton, NN2 7AL, United Kingdom
| | - Edward J. Hollox
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Helen Parker
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Kate V. Latham
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Taco W. Kuijpers
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX, Amsterdam, The Netherlands
- Department of Pediatric Hematology, Immunology and Infectious Disease, Emma Children’s Hospital, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Kathleen N. Potter
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Sarah E. Coupland
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, United Kingdom
| | - Andrew Davies
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Michael Stackpole
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, United Kingdom
| | - Melanie Oates
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, United Kingdom
| | - Andrew R. Pettitt
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, United Kingdom
| | - Martin J. Glennie
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Mark S. Cragg
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Jonathan C. Strefford
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
Tzeng SJ, Li WY, Wang HY. FcγRIIB mediates antigen-independent inhibition on human B lymphocytes through Btk and p38 MAPK. J Biomed Sci 2015; 22:87. [PMID: 26475492 PMCID: PMC4609082 DOI: 10.1186/s12929-015-0200-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022] Open
Abstract
Background The inhibitory Fc receptor, FcγRIIB, has emerged as a key negative regulator of B cell activation and as such is predicted to play an essential role in controlling antibody-mediated autoimmune diseases in humans. Recent studies have shown that crosslinking the FcγRIIB independently of the B-cell receptor (BCR) results in apoptosis in both mouse and chicken B cells. However, the human B cell subpopulations that are susceptible to BCR-independent, FcγRIIB-mediated regulation are not known. How FcγRIIB mediates this inhibition to affect B cell homeostasis is also not determined. Results We isolated naïve B cells, memory B cells and plasma cells (PCs) from peripheral blood of healthy donors and used differentiated PCs in culture to examine the effects on them by FcγRIIB crosslinking. We showed that human PCs, memory and naïve B cells all expressed FcγRIIB with expression on PCs being the highest in circulation. Moreover, they were sensitive to direct inhibition by FcγRIIB through Btk and p38 MAPK. Similarly, PCs resulting from the antigen-independent differentiation of memory B cells in vitro were inhibited by FcγRIIB cross-linking but memory B cell activation itself, as measured by proliferation, was unaffected. In contrast, both the proliferation and differentiation of naïve B cells to PCs were blocked by FcγRIIB crosslinking. Conclusion These results suggest a mechanism to control antibody levels involving the differential expression of FcγRIIB on B cell subpopulations, in which the FcγRIIB functions independently of the BCR to eliminate antibody-secreting effector cells and inhibit naïve B cell proliferation without compromising the long-lived antigen-specific memory B cells. Importantly, FcγRIIB requires Btk and p38 MAPK to mediate antigen-independent inhibition in human B cells. Taken together, our data underscore the importance of antigen-independent inhibition by FcγRIIB in the prevention from antibody-mediated autoimmune diseases and in the regulation of B cell homeostasis.
Collapse
Affiliation(s)
- Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Room 1118, No.1, Section 1, Ren-Ai Road, Taipei, 10051, Taiwan.
| | - Wan-Yu Li
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Room 1118, No.1, Section 1, Ren-Ai Road, Taipei, 10051, Taiwan
| | - Hui-Ying Wang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Room 1118, No.1, Section 1, Ren-Ai Road, Taipei, 10051, Taiwan
| |
Collapse
|
38
|
Yan C, Wang X, Liu Y, Abdulnour RE, Wu M, Gao H. Protective Role of Rho Guanosine Diphosphate Dissociation Inhibitor, Ly-GDI, in Pulmonary Alveolitis. PLoS One 2015; 10:e0140804. [PMID: 26469087 PMCID: PMC4607448 DOI: 10.1371/journal.pone.0140804] [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: 08/05/2015] [Accepted: 09/30/2015] [Indexed: 11/18/2022] Open
Abstract
Growing evidences indicate that Ly-GDI, an inhibitory protein of Rho GTPases, plays an essential role in regulating actin cytoskeletal alteration which is indispensible for the process such as phagocytosis. However, the role of Ly-GDI in inflammation remains largely unknown. In the current study, we found that Ly-GDI expression was significantly decreased in the IgG immune complex-injured lungs. To determine if Ly-GDI might regulate the lung inflammatory response, we constructed adenovirus vectors that could mediate ectopic expression of Ly-GDI (Adeno-Ly-GDI). In vivo mouse lung expression of Ly-GDI resulted in a significant attenuation of IgG immune complex-induced lung injury, which was due to the decreased pulmonary permeability and lung inflammatory cells, especially neutrophil accumulation. Upon IgG immune complex deposition, mice with Ly-GDI over-expression in the lungs produced significant less inflammatory mediators (TNF-α, IL-6, MCP-1, and MIP-1α) in bronchoalveolar lavage fluid when compared control mice receiving airway injection of Adeno-GFP. Mechanically, IgG immune complex-induced NF-κB activity was markedly suppressed by Ly-GDI in both alveolar macrophages and lungs as measured by luciferase assay and electrophoretic mobility shift assay. These findings suggest that Ly-GDI is a critical regulator of inflammatory injury after deposition of IgG immune complexes and that it negatively regulates the lung NF-κB activity.
Collapse
Affiliation(s)
- Chunguang Yan
- Department of Anesthesiology, Perioperative&Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Ximo Wang
- Department of Surgery, Tianjin Nankai Hospital, Tianjin, China
| | - Yanlan Liu
- Department of Anesthesiology, Perioperative&Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Raja-Elie Abdulnour
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Min Wu
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Hongwei Gao
- Department of Anesthesiology, Perioperative&Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Surgery, Tianjin Nankai Hospital, Tianjin, China
- * E-mail:
| |
Collapse
|
39
|
Asthana P, Vong JSL, Kumar G, Chang RCC, Zhang G, Sheikh KA, Ma CHE. Dissecting the Role of Anti-ganglioside Antibodies in Guillain-Barré Syndrome: an Animal Model Approach. Mol Neurobiol 2015; 53:4981-91. [PMID: 26374552 DOI: 10.1007/s12035-015-9430-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/07/2015] [Indexed: 12/19/2022]
Abstract
Guillain-Barré syndrome (GBS) is an autoimmune polyneuropathy disease affecting the peripheral nervous system (PNS). Most of the GBS patients experienced neurological symptoms such as paresthesia, weakness, pain, and areflexia. There are also combinations of non-neurological symptoms which include upper respiratory tract infection and diarrhea. One of the major causes of GBS is due largely to the autoantibodies against gangliosides located on the peripheral nerves. Gangliosides are sialic acid-bearing glycosphingolipids consisting of a ceramide lipid anchor with one or more sialic acids attached to a neutral sugar backbone. Molecular mimicry between the outer components of oligosaccharide of gangliosides on nerve membrane and lipo-oligosaccharide of microbes is thought to trigger the autoimmunity. Intra-peritoneal implantation of monoclonal ganglioside antibodies secreting hybridoma into animals induced peripheral neuropathy. Recent studies demonstrated that injection of synthesized anti-ganglioside antibodies raised by hybridoma cells into mice initiates immune response against peripheral nerves, and eventually failure in peripheral nerve regeneration. Accumulating evidences indicate that the conjugation of anti-ganglioside monoclonal antibodies to activating FcγRIII present on the circulating macrophages inhibits axonal regeneration. The activation of RhoA signaling pathways is also involved in neurite outgrowth inhibition. However, the link between these two molecular events remains unresolved and requires further investigation. Development of anti-ganglioside antagonists can serve as targeted therapy for the treatment of GBS and will open a new approach of drug development with maximum efficacy and specificity.
Collapse
Affiliation(s)
- Pallavi Asthana
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China
| | - Joaquim Si Long Vong
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China
| | - Gajendra Kumar
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong, People's Republic of China
| | - Gang Zhang
- Department of Neurology, University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Kazim A Sheikh
- Department of Neurology, University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Chi Him Eddie Ma
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China. .,Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China. .,State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Hong Kong, People's Republic of China.
| |
Collapse
|
40
|
Burton DR, Mascola JR. Antibody responses to envelope glycoproteins in HIV-1 infection. Nat Immunol 2015; 16:571-6. [PMID: 25988889 DOI: 10.1038/ni.3158] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/26/2015] [Indexed: 02/08/2023]
Abstract
Antibody responses to the HIV-1 envelope glycoproteins can be classified into three groups. Binding but non-neutralizing responses are directed to epitopes that are expressed on isolated envelope glycoproteins but not on the native envelope trimer found on the surface of virions and responsible for mediating the entry of virus into target cells. Strain-specific responses and broadly neutralizing responses, in contrast, target epitopes that are expressed on the native trimer, as revealed by recently resolved structures. The past few years have seen the isolation of many broadly neutralizing antibodies of remarkable potency that have shown prophylactic and therapeutic activities in animal models. These antibodies are helping to guide rational vaccine design and therapeutic strategies for HIV-1.
Collapse
Affiliation(s)
- Dennis R Burton
- 1] Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, USA. [2] International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA. [3] Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA. [4] Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Boston, Massachusetts, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
41
|
Anti-Ganglioside Antibodies Induce Nodal and Axonal Injury via Fcγ Receptor-Mediated Inflammation. J Neurosci 2015; 35:6770-85. [PMID: 25926454 DOI: 10.1523/jneurosci.4926-14.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is a postinfectious autoimmune neuropathy and anti-ganglioside antibodies (Abs) are strongly associated with this disorder. Several studies have implied that specific anti-ganglioside Abs induce neuropathy in patients with axonal forms of GBS. To study the mechanisms of anti-ganglioside Abs-induced neuropathy, we established a new passive transfer mouse model by L5 spinal nerve transection (L5SNT; modified Chung's model) and systemic administration of anti-ganglioside Abs. L5SNT causes degeneration of a small proportion of fibers that constitute sciatic nerve and its branches, but importantly breaks the blood-nerve barrier, which allows access to circulating Abs and inflammatory cells. Our studies indicate that, in this mouse model, anti-ganglioside Abs induce sequential nodal and axonal injury of intact myelinated nerve fibers, recapitulating pathologic features of human disease. Notably, our results showed that immune complex formation and the activating Fc gamma receptors (FcγRs) were involved in the anti-ganglioside Abs-mediated nodal and axonal injury in this model. These studies provide new evidence that the activating FcγRs-mediated inflammation plays a critical role in anti-ganglioside Abs-induced neuropathy (injury to intact nerve fibers) in GBS.
Collapse
|
42
|
Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS. FcRn: The Architect Behind the Immune and Nonimmune Functions of IgG and Albumin. THE JOURNAL OF IMMUNOLOGY 2015; 194:4595-603. [PMID: 25934922 DOI: 10.4049/jimmunol.1403014] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neonatal FcR (FcRn) belongs to the extensive and functionally divergent family of MHC molecules. Contrary to classical MHC family members, FcRn possesses little diversity and is unable to present Ags. Instead, through its capacity to bind IgG and albumin with high affinity at low pH, it regulates the serum half-lives of both of these proteins. In addition, FcRn plays an important role in immunity at mucosal and systemic sites through its ability to affect the lifespan of IgG, as well as its participation in innate and adaptive immune responses. Although the details of its biology are still emerging, the ability of FcRn to rescue albumin and IgG from early degradation represents an attractive approach to alter the plasma half-life of pharmaceuticals. We review some of the most novel aspects of FcRn biology, immune as well as nonimmune, and provide some examples of FcRn-based therapies.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Timo Rath
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115; Division of Gastroenterology, Department of Medicine, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg, 91054 Erlangen, Germany
| | - Wayne I Lencer
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA 02115; Harvard Digestive Diseases Center, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Kristi Baker
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Richard S Blumberg
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115; Harvard Digestive Diseases Center, Boston, MA 02115; and
| |
Collapse
|
43
|
DiLillo DJ, Ravetch JV. Differential Fc-Receptor Engagement Drives an Anti-tumor Vaccinal Effect. Cell 2015; 161:1035-1045. [PMID: 25976835 DOI: 10.1016/j.cell.2015.04.016] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/04/2015] [Accepted: 03/22/2015] [Indexed: 01/21/2023]
Abstract
Passively administered anti-tumor monoclonal antibodies (mAbs) rapidly kill tumor targets via FcγR-mediated cytotoxicity (ADCC), a short-term process. However, anti-tumor mAb treatment can also induce a vaccinal effect, in which mAb-mediated tumor death induces a long-term anti-tumor cellular immune response. To determine how such responses are generated, we utilized a murine model of an anti-tumor vaccinal effect against a model neoantigen. We demonstrate that FcγR expression by CD11c(+) antigen-presenting cells is required to generate anti-tumor T cell responses upon ADCC-mediated tumor clearance. Using FcγR-humanized mice, we demonstrate that anti-tumor human (h)IgG1 must engage hFcγRIIIA on macrophages to mediate ADCC, but also engage hFcγRIIA, the sole hFcγR expressed by human dendritic cells (DCs), to generate a potent vaccinal effect. Thus, while next-generation anti-tumor antibodies with enhanced binding to only hFcγRIIIA are now in clinical use, ideal anti-tumor antibodies must be optimized for both cytotoxic effects as well as hFcγRIIA engagement on DCs to stimulate long-term anti-tumor cellular immunity.
Collapse
Affiliation(s)
- David J DiLillo
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
44
|
Hanson QM, Barb AW. A perspective on the structure and receptor binding properties of immunoglobulin G Fc. Biochemistry 2015; 54:2931-42. [PMID: 25926001 DOI: 10.1021/acs.biochem.5b00299] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recombinant antibodies spurred a revolution in medicine that saw the introduction of powerful therapeutics for treating a wide range of diseases, from cancers to autoimmune disorders and transplant rejection, with more applications looming on the horizon. Many of these therapeutic monoclonal antibodies (mAbs) are based on human immunoglobulin G1 (IgG1) or contain at least a portion of the molecule. Most mAbs require interactions with cell surface receptors for efficacy, including the Fc γ receptors. High-resolution structural models of antibodies and antibody fragments have been available for nearly 40 years; however, a thorough description of the structural features that determine the affinity with which antibodies interact with human receptors has not been published. In this review, we will cover the relevant history of IgG-related literature and how recent developments have changed our view of critical antibody-cell interactions at the atomic level with a nod to outstanding questions in the field and future prospects.
Collapse
Affiliation(s)
- Quinlin M Hanson
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| |
Collapse
|
45
|
Inui M, Tazawa K, Kishi Y, Takai T. Platelets convert peripheral blood circulating monocytes to regulatory cells via immunoglobulin G and activating-type Fcγ receptors. BMC Immunol 2015; 16:20. [PMID: 25896516 PMCID: PMC4407389 DOI: 10.1186/s12865-015-0086-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/16/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Monocytes and macrophages produce interleukin (IL)-10, an immunoregulatory cytokine and a potent therapeutic tool for immune disorders. Augmentation of IL-10 production with a concomitant reduction of proinflammatory cytokines in macrophages in vitro is attained by doubly stimulating the cells with a toll-like receptor ligand and immunoglobulin (Ig)G immune complexes, a response known as that of regulatory (or alternatively activated/M2) macrophages. However, it has not been explored sufficiently how such a regulatory response could be exploited for anti-inflammation. Our objective is to find a potential way or condition for augmenting IL-10 by monocytes/macrophages in vivo and in vitro. RESULTS We show that platelets, when they are opsonized with IgG, can convert human peripheral blood circulating monocytes to IL-10-producing regulatory monocytes in vitro and also in a murine in vivo model. Co-culturing of platelets and monocytes in the presence of anti-integrin IgG and a bacterial lipopolysaccharide augmented IL-10 production via a direct interaction between platelets and monocytes. This novel way of enhancing IL-10 was mediated by activating-type Fc receptors for IgG. CONCLUSION These findings indicate that the IgG-bound platelet-induced conversion of monocytes to regulatory cells might provide a novel strategy for controlling inflammation.
Collapse
Affiliation(s)
- Masanori Inui
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Kino Tazawa
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Yoshiro Kishi
- Medical & Biological Laboratories, Co., Ltd., 4-5-3, Sakae, Naka-ku, Nagoya, 460-0008, Japan.
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| |
Collapse
|
46
|
Itraconazole, a commonly used antifungal, inhibits Fcγ receptor-mediated phagocytosis: alteration of Fcγ receptor glycosylation and gene expression. Shock 2015; 42:52-9. [PMID: 24667630 DOI: 10.1097/shk.0000000000000169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Itraconazole (ICZ) is commonly used for the treatment of fungal infections, particularly in immunocompromised patients. In addition, ICZ has been recently found to have antiangiogenic effects and is currently being tested as a new chemotherapeutic agent in several cancer clinical trials. We have previously shown that ICZ impaired complex N-linked glycosylation processing, leading to the accumulation of high-mannose glycoproteins on the surface of macrophages (Møs). This investigation was directed at determining the effects of ICZ on phagocytosis as a major function of Møs. We found a significant decrease in the phagocytosis of opsonized bacterial particles in ICZ-treated murine Møs in comparison with nontreated Møs. Furthermore, the impairment of phagocytosis was associated with a decrease in cell surface expression of Fcγ receptors (FcγRs) as well as alteration of their glycosylation pattern. Concomitantly, a reduction in all three isoforms of the FcγR family (i.e., Fcgr1, Fcgr2, and Fcgr3) mRNA levels was observed after incubation with ICZ. The effect of ICZ on phagocytosis and FcγR expression was reversed by addition of low-density lipoprotein. These studies indicate that ICZ treatment certainly has a dramatic effect on macrophage function, which could result in a potential impairment of the immune system';s ability to respond to pathogens and may lead to an elevated incidence of infections.
Collapse
|
47
|
Corrales-Aguilar E, Hoffmann K, Hengel H. CMV-encoded Fcγ receptors: modulators at the interface of innate and adaptive immunity. Semin Immunopathol 2014; 36:627-40. [PMID: 25288477 DOI: 10.1007/s00281-014-0448-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022]
Abstract
The constant region of IgG antibodies mediates antiviral activities upon engaging host Fcγ receptors (FcγRs) expressed by a variety of immune cells, such as antibody-dependent cellullar cytotoxcity (ADCC) executed by natural killer (NK)cells. Human cytomegalovirus (HCMV) is unique among viruses by encoding also an array of several Fcγ-binding glycoproteins with cell surface disposition and concomitant incorporation into the virion. Evidence is increasing that the virus-encoded Fcγ receptors differ in their Fcγ binding mode but effectively operate as adversaries of host FcγRs since they are able to prevent IgG-mediated triggering of activating host FcγRs, i.e., FcγRI, FcγRIIA, and FcγRIIIA. Here we discuss virus-encoded FcγRs as the first known HCMV inhibitors of IgG-mediated immunity which could account for the limited efficacy of HCMV hyperimmune globulin in clinical settings. A better understanding of their molecular mode of action opens up new perspectives for improving IgG therapies against HCMV disease.
Collapse
Affiliation(s)
- Eugenia Corrales-Aguilar
- Virology-CIET, Faculty of Microbiology, University of Costa Rica, 11501-2060, San José, Costa Rica
| | | | | |
Collapse
|
48
|
Pincetic A, Bournazos S, DiLillo DJ, Maamary J, Wang TT, Dahan R, Fiebiger BM, Ravetch JV. Type I and type II Fc receptors regulate innate and adaptive immunity. Nat Immunol 2014; 15:707-16. [PMID: 25045879 DOI: 10.1038/ni.2939] [Citation(s) in RCA: 376] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022]
Abstract
Antibodies produced in response to a foreign antigen are characterized by polyclonality, not only in the diverse epitopes to which their variable domains bind but also in the various effector molecules to which their constant regions (Fc domains) engage. Thus, the antibody's Fc domain mediates diverse effector activities by engaging two distinct classes of Fc receptors (type I and type II) on the basis of the two dominant conformational states that the Fc domain may adopt. These conformational states are regulated by the differences among antibody subclasses in their amino acid sequence and by the complex, biantennary Fc-associated N-linked glycan. Here we discuss the diverse downstream proinflammatory, anti-inflammatory and immunomodulatory consequences of the engagement of type I and type II Fc receptors in the context of infectious, autoimmune, and neoplastic disorders.
Collapse
Affiliation(s)
- Andrew Pincetic
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - Stylianos Bournazos
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - David J DiLillo
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - Jad Maamary
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | - Taia T Wang
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | - Rony Dahan
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | | | - Jeffrey V Ravetch
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
49
|
Antonelli LRV, Leoratti FMS, Costa PAC, Rocha BC, Diniz SQ, Tada MS, Pereira DB, Teixeira-Carvalho A, Golenbock DT, Gonçalves R, Gazzinelli RT. The CD14+CD16+ inflammatory monocyte subset displays increased mitochondrial activity and effector function during acute Plasmodium vivax malaria. PLoS Pathog 2014; 10:e1004393. [PMID: 25233271 PMCID: PMC4169496 DOI: 10.1371/journal.ppat.1004393] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 08/11/2014] [Indexed: 01/22/2023] Open
Abstract
Infection with Plasmodium vivax results in strong activation of monocytes, which are important components of both the systemic inflammatory response and parasite control. The overall goal of this study was to define the role of monocytes during P. vivax malaria. Here, we demonstrate that P. vivax-infected patients display significant increase in circulating monocytes, which were defined as CD14(+)CD16- (classical), CD14(+)CD16(+) (inflammatory), and CD14loCD16(+) (patrolling) cells. While the classical and inflammatory monocytes were found to be the primary source of pro-inflammatory cytokines, the CD16(+) cells, in particular the CD14(+)CD16(+) monocytes, expressed the highest levels of activation markers, which included chemokine receptors and adhesion molecules. Morphologically, CD14(+) were distinguished from CD14lo monocytes by displaying larger and more active mitochondria. CD14(+)CD16(+) monocytes were more efficient in phagocytizing P. vivax-infected reticulocytes, which induced them to produce high levels of intracellular TNF-α and reactive oxygen species. Importantly, antibodies specific for ICAM-1, PECAM-1 or LFA-1 efficiently blocked the phagocytosis of infected reticulocytes by monocytes. Hence, our results provide key information on the mechanism by which CD14(+)CD16(+) cells control parasite burden, supporting the hypothesis that they play a role in resistance to P. vivax infection.
Collapse
Affiliation(s)
- Lis R. V. Antonelli
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| | - Fabiana M. S. Leoratti
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro A. C. Costa
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno C. Rocha
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Suelen Q. Diniz
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro S. Tada
- Centro de Pesquisas em Medicina Tropical de Rondônia, Porto Velho, Rondônia, Brazil
| | - Dhelio B. Pereira
- Centro de Pesquisas em Medicina Tropical de Rondônia, Porto Velho, Rondônia, Brazil
| | - Andrea Teixeira-Carvalho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Douglas T. Golenbock
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ricardo Gonçalves
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo T. Gazzinelli
- Laboratório de Immunopatologia, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
50
|
Ahmed AA, Giddens J, Pincetic A, Lomino JV, Ravetch JV, Wang LX, Bjorkman PJ. Structural characterization of anti-inflammatory immunoglobulin G Fc proteins. J Mol Biol 2014; 426:3166-3179. [PMID: 25036289 PMCID: PMC4159253 DOI: 10.1016/j.jmb.2014.07.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/13/2014] [Accepted: 07/07/2014] [Indexed: 12/13/2022]
Abstract
Immunoglobulin G (IgG) is a central mediator of host defense due to its ability to recognize and eliminate pathogens. The recognition and effector responses are encoded on distinct regions of IgGs. The diversity of the antigen recognition Fab domains accounts for IgG's ability to bind with high specificity to essentially any antigen. Recent studies have indicated that the Fc effector domain also displays considerable heterogeneity, accounting for its complex effector functions of inflammation, modulation, and immune suppression. Therapeutic anti-tumor antibodies, for example, require the pro-inflammatory properties of the IgG Fc to eliminate tumor cells, while the anti-inflammatory activity of intravenous IgG requires specific Fc glycans for activity. In particular, the anti-inflammatory activity of intravenous IgG is ascribed to a small population of IgGs in which the Asn297-linked complex N-glycans attached to each Fc CH2 domain include terminal α2,6-linked sialic acids. We used chemoenzymatic glycoengineering to prepare fully disialylated IgG Fc and solved its crystal structure. Comparison of the structures of asialylated Fc, sialylated Fc, and F241A Fc, a mutant that displays increased glycan sialylation, suggests that increased conformational flexibility of the CH2 domain is associated with the switch from pro-inflammatory to anti-inflammatory activity of the Fc.
Collapse
Affiliation(s)
- Alysia A Ahmed
- Division of Biology and Biological Engineering 114-96, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - John Giddens
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Andrew Pincetic
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| | - Joseph V Lomino
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| | - Lai-Xi Wang
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering 114-96, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|