1
|
Wang X, Campbell B, Bodogai M, McDevitt RA, Patrikeev A, Gusev F, Ragonnaud E, Kumaraswami K, Shirenova S, Vardy K, Alameh MG, Weissman D, Ishikawa-Ankerhold H, Okun E, Rogaev E, Biragyn A. CD8 + T cells exacerbate AD-like symptoms in mouse model of amyloidosis. Brain Behav Immun 2024; 122:444-455. [PMID: 39191349 PMCID: PMC11409913 DOI: 10.1016/j.bbi.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Alzheimer's disease (AD) is linked to toxic Aβ plaques in the brain and activation of innate responses. Recent findings however suggest that the disease may also depend on the adaptive immunity, as B cells exacerbate and CD8+ T cells limit AD-like pathology in mouse models of amyloidosis. Here, by artificially blocking or augmenting CD8+ T cells in the brain of 5xFAD mice, we provide evidence that AD-like pathology is promoted by pathogenic, proinflammatory cytokines and exhaustion markers expressing CXCR6+ CD39+CD73+/- CD8+ TRM-like cells. The CD8+ T cells appear to act by targeting disease associated microglia (DAM), as we find them in tight complexes with microglia around Aβ plaques in the brain of mice and humans with AD. We also report that these CD8+ T cells are induced by B cells in the periphery, further underscoring the pathogenic importance of the adaptive immunity in AD. We propose that CD8+ T cells and B cells should be considered as therapeutic targets for control of AD, as their ablation at the onset of AD is sufficient to decrease CD8+ T cells in the brain and block the amyloidosis-linked neurodegeneration.
Collapse
Affiliation(s)
- Xin Wang
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Britney Campbell
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, Baltimore, MD, USA
| | - Anton Patrikeev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fedor Gusev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Konda Kumaraswami
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Sophie Shirenova
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | - Karin Vardy
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | | | - Drew Weissman
- Institute of RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Hellen Ishikawa-Ankerhold
- Department of Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | - Evgeny Rogaev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA.
| |
Collapse
|
2
|
Chan C, Cabanes NC, Jansen JHM, Guillaume J, Nederend M, Passchier EM, Gómez-Mellado VE, Peipp M, Boes M, van Tetering G, Leusen JHW. The relevance of tumor target expression levels on IgA-mediated cytotoxicity in cancer immunotherapy. Cancer Immunol Immunother 2024; 73:238. [PMID: 39358557 PMCID: PMC11447191 DOI: 10.1007/s00262-024-03824-0] [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: 03/25/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024]
Abstract
Recent advances in cancer immunotherapy, particularly the success of immune checkpoint inhibitors, have reignited interest in targeted monoclonal antibodies for immunotherapy. Antibody therapies aim to minimize on-target, off-tumor toxicity by targeting antigens overexpressed on tumor cells but not on healthy cells. Despite considerable efforts, some therapeutic antibodies have been linked to dose-limiting side effects. Our hypothesis suggests that the efficacy of IgG leads to a lower target expression threshold for tumor cell killing, contributing to these side effects. Earlier, therapeutic IgG antibodies were reformatted into the IgA isotype. Unlike IgG, which primarily engages Fc gamma receptors (FcγR) to induce antibody-dependent cellular cytotoxicity (ADCC) by NK cells and antibody-dependent cellular phagocytosis (ADCP) by monocytes/macrophages, IgA antibodies activate neutrophils through the Fc alpha receptor I (CD89, FcαRI). In previous studies, it appeared that IgA may require a higher target expression threshold for effective killing, and we aimed to investigate this in our current study. Moreover, we investigated how blocking the myeloid checkpoint CD47/SIRPα axis affect the target expression threshold. Using a tetracycline-inducible expression system, we regulated target expression in different cell lines. Our findings from ADCC assays indicate that IgA-mediated PMN ADCC requires a higher antigen expression level than IgG-mediated PBMC ADCC. Furthermore, blocking CD47 enhanced IgA-mediated ADCC, lowering the antigen threshold. Validated in two in vivo models, our results show that IgA significantly reduces tumor growth in high-antigen-expressing tumors without affecting low-antigen-expressing healthy tissues. This suggests IgA-based immunotherapy could potentially minimize on-target, off-tumor side effects, improving treatment efficacy and patient safety.
Collapse
Affiliation(s)
- Chilam Chan
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Núria Casalé Cabanes
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - J H Marco Jansen
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Joël Guillaume
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Maaike Nederend
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Elsemieke M Passchier
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | | | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Campus Kiel, 24105, Kiel, Germany
| | - Marianne Boes
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Pediatrics Department, University Medical Center, Utrecht, The Netherlands
| | - Geert van Tetering
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Chan V, Camardi C, Zhang K, Orofiamma LA, Anderson KE, Hoque J, Bone LN, Awadeh Y, Lee DKC, Fu NJ, Chow JTS, Salmena L, Stephens LR, Hawkins PT, Antonescu CN, Botelho RJ. The LCLAT1/LYCAT acyltransferase is required for EGF-mediated phosphatidylinositol-3,4,5-trisphosphate generation and Akt signaling. Mol Biol Cell 2024; 35:ar118. [PMID: 39024272 PMCID: PMC11449395 DOI: 10.1091/mbc.e23-09-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
Receptor tyrosine kinases such as EGF receptor (EGFR) stimulate phosphoinositide 3 kinases to convert phosphatidylinositol-4,5-bisphosophate [PtdIns(4,5)P2] into phosphatidylinositol-3,4,5-trisphosphate [PtdIns(3,4,5)P3]. PtdIns(3,4,5)P3 then remodels actin and gene expression, and boosts cell survival and proliferation. PtdIns(3,4,5)P3 partly achieves these functions by triggering activation of the kinase Akt, which phosphorylates targets like Tsc2 and GSK3β. Consequently, unchecked upregulation of PtdIns(3,4,5)P3-Akt signaling promotes tumor progression. Interestingly, 50-70% of PtdIns and PtdInsPs have stearate and arachidonate at sn-1 and sn-2 positions of glycerol, respectively, forming a species known as 38:4-PtdIns/PtdInsPs. LCLAT1 and MBOAT7 acyltransferases partly enrich PtdIns in this acyl format. We previously showed that disruption of LCLAT1 lowered PtdIns(4,5)P2 levels and perturbed endocytosis and endocytic trafficking. However, the role of LCLAT1 in receptor tyrosine kinase and PtdIns(3,4,5)P3 signaling was not explored. Here, we show that LCLAT1 silencing in MDA-MB-231 and ARPE-19 cells abated the levels of PtdIns(3,4,5)P3 in response to EGF signaling. Importantly, LCLAT1-silenced cells were also impaired for EGF-driven and insulin-driven Akt activation and downstream signaling. Thus, our work provides first evidence that the LCLAT1 acyltransferase is required for receptor tyrosine kinase signaling.
Collapse
Affiliation(s)
- Victoria Chan
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Cristina Camardi
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Kai Zhang
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Laura A. Orofiamma
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Karen E. Anderson
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Jafarul Hoque
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Leslie N. Bone
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Yasmin Awadeh
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Daniel K. C. Lee
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Norman J. Fu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Jonathan T. S. Chow
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Len R. Stephens
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Phillip T. Hawkins
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Costin N. Antonescu
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| |
Collapse
|
4
|
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
|
5
|
L’Estrange-Stranieri E, Gottschalk TA, Wright MD, Hibbs ML. The dualistic role of Lyn tyrosine kinase in immune cell signaling: implications for systemic lupus erythematosus. Front Immunol 2024; 15:1395427. [PMID: 39007135 PMCID: PMC11239442 DOI: 10.3389/fimmu.2024.1395427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.
Collapse
Affiliation(s)
- Elan L’Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Timothy A. Gottschalk
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mark D. Wright
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Oh J, Kirsh C, Hsin JP, Radecki KC, Zampieri A, Manry D, Ando Y, Miller S, Chan J, McLeod E, Cunningham KM, Wong LM, Xu H, Kamb A. NOT gated T cells that selectively target EGFR and other widely expressed tumor antigens. iScience 2024; 27:109913. [PMID: 38799557 PMCID: PMC11126980 DOI: 10.1016/j.isci.2024.109913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 01/04/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Here, we show that a NOT gated cell therapy (Tmod) can exploit antigens such as epidermal growth factor receptor (EGFR) and human leukocyte antigen-E (HLA-E) which are widely expressed on cancer cells. Noncancerous cells-despite high expression of these antigens-are protected from cytotoxicity by the action of an inhibitory receptor ("blocker") via a mechanism that involves blocker modulation of CAR surface expression. The blocker is triggered by the product of a polymorphic HLA allele (e.g., HLA-A∗02) deleted in a significant subset of solid tumors via loss of heterozygosity. Moreover, Tmod constructs that target mouse homologs of EGFR or HLA-E for activation, and a mouse-equivalent of HLA-A∗02 for inhibition, protect mice from toxicity caused by the CAR alone. The blocker also controls graft vs. host response in allogeneic T cells in vitro, consistent with the use of Tmod cells for off-the-shelf therapy without additional gene-editing.
Collapse
Affiliation(s)
- Julyun Oh
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Charles Kirsh
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Jing-Ping Hsin
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Kelly C. Radecki
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | | | - Diane Manry
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Yuta Ando
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Sara Miller
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Jamie Chan
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Ethan McLeod
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | | | - Lu Min Wong
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Han Xu
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| | - Alexander Kamb
- A2 Biotherapeutics; 30301 Agoura Rd., Agoura Hills 91301, CA, USA
| |
Collapse
|
7
|
Merchand-Reyes G, Bull MF, Santhanam R, Valencia-Pena ML, Murugesan RA, Chordia A, Mo XM, Robledo-Avila FH, Ruiz-Rosado JDD, Carson WE, Byrd JC, Woyach JA, Tridandapani S, Butchar JP. NOD2 activation enhances macrophage Fcγ receptor function and may increase the efficacy of antibody therapy. Front Immunol 2024; 15:1409333. [PMID: 38919608 PMCID: PMC11196781 DOI: 10.3389/fimmu.2024.1409333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction Therapeutic antibodies have become a major strategy to treat oncologic diseases. For chronic lymphocytic leukemia, antibodies against CD20 are used to target and elicit cytotoxic responses against malignant B cells. However, efficacy is often compromised due to a suppressive microenvironment that interferes with cellular immune responses. To overcome this suppression, agonists of pattern recognition receptors have been studied which promote direct cytotoxicity or elicit anti-tumoral immune responses. NOD2 is an intracellular pattern recognition receptor that participates in the detection of peptidoglycan, a key component of bacterial cell walls. This detection then mediates the activation of multiple signaling pathways in myeloid cells. Although several NOD2 agonists are being used worldwide, the potential benefit of these agents in the context of antibody therapy has not been explored. Methods Primary cells from healthy-donor volunteers (PBMCs, monocytes) or CLL patients (monocytes) were treated with versus without the NOD2 agonist L18-MDP, then antibody-mediated responses were assessed. In vivo, the Eµ-TCL1 mouse model of CLL was used to test the effects of L18-MDP treatment alone and in combination with anti-CD20 antibody. Results Treatment of peripheral blood mononuclear cells with L18-MDP led to activation of monocytes from both healthy donors and CLL patients. In addition, there was an upregulation of activating FcγR in monocytes and a subsequent increase in antibody-mediated phagocytosis. This effect required the NF-κB and p38 signaling pathways. Treatment with L18-MDP plus anti-CD20 antibody in the Eµ-TCL model of CLL led to a significant reduction of CLL load, as well as to phenotypic changes in splenic monocytes and macrophages. Conclusions Taken together, these results suggest that NOD2 agonists help overturn the suppression of myeloid cells, and may improve the efficacy of antibody therapy for CLL.
Collapse
MESH Headings
- Nod2 Signaling Adaptor Protein/agonists
- Nod2 Signaling Adaptor Protein/metabolism
- Nod2 Signaling Adaptor Protein/immunology
- Animals
- Humans
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Mice
- Macrophages/immunology
- Macrophages/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Acetylmuramyl-Alanyl-Isoglutamine/pharmacology
- Female
- Mice, Inbred C57BL
- Signal Transduction
- Phagocytosis
- Rituximab/pharmacology
- Rituximab/therapeutic use
Collapse
Affiliation(s)
- Giovanna Merchand-Reyes
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Mikayla F. Bull
- College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ramasamy Santhanam
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Maria L. Valencia-Pena
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | | | - Aadesh Chordia
- College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Xiaokui-Molly Mo
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Frank H. Robledo-Avila
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States
| | - Juan De Dios Ruiz-Rosado
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Division of Pediatric Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - John C. Byrd
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Jennifer A. Woyach
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Susheela Tridandapani
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jonathan P. Butchar
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
8
|
Niemand RR, Stafford JL. Counteracting immunotyrosine-based signaling motifs augment zebrafish leukocyte immune-type receptor-mediated phagocytic activity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 153:105121. [PMID: 38135021 DOI: 10.1016/j.dci.2023.105121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Leukocyte immune-type receptors (LITRs) represent a polymorphic and polygenic family of immunoregulatory proteins originally discovered in channel catfish (Ictalurus punctatus; IpLITRs). Belonging to the immunoglobulin superfamily (IgSF), IpLITRs are generally classified as stimulatory or inhibitory types based on their utilization of various intracellular tyrosine-based signaling motifs. While research has shown that IpLITRs can activate as well as abrogate different immune cell effector responses including phagocytosis, recent identification of LITRs within the zebrafish genome (Danio rerio; DrLITRs) revealed the existence of fish LITR-types uniquely containing counteracting stimulatory and inhibitory cytoplasmic tail (CYT) region motifs (i.e., an immunoreceptor tyrosine-based activation motif; ITAM, and immunoreceptor tyrosine-based inhibitory motif; ITIM) within the same receptor. This arrangement is unusual as these motifs typically exist on separate stimulatory (i.e., ITAM-containing) or inhibitory (i.e., ITIM-containing) immunoregulatory receptors that then co-engage to fine-tune cellular signaling and effector responses. Using a flow cytometric-based phagocytosis assay, we show here that engagement of DrLITR 1.2-expressing cells with antibody coated 4.5 μm beads causes a robust ITAM-dependent phagocytic response and reveal that its tandem ITIM motif surprisingly enhances the DrLITR 1.2-induced phagocytic activity while simultaneously decreasing the receptors ability to bind the beads. Confocal microscopy studies also revealed that the ITIM-associated inhibitory signaling molecule SHP-2 is localized to the phagocytic synapse during the phagocytic response. Overall, these results provide the first functional characterization of teleost immune receptors containing a tandem ITAM and ITIM and allow for the proposal of an intracytoplasmic tail signaling model for ITIM-mediated enhancement of ITAM-dependent cellular activation.
Collapse
Affiliation(s)
- Rikus R Niemand
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
9
|
Hoeger S, Drake LA, Drake JR. Proximity-Based Labeling Identifies MHC Class II and CD37 as B Cell Receptor-Proximal Proteins with Immunological Functions. Immunohorizons 2024; 8:326-338. [PMID: 38625120 PMCID: PMC11066716 DOI: 10.4049/immunohorizons.2400014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024] Open
Abstract
The BCR allows for Ag-driven B cell activation and subsequent Ag endocytosis, processing, and presentation to recruit T cell help. Core drivers of BCR signaling and endocytosis are motifs within the receptor's cytoplasmic tail (primarily CD79). However, BCR function can be tuned by other proximal cellular elements, such as CD20 and membrane lipid microdomains. To identify additional proteins that could modulate BCR function, we used a proximity-based biotinylation technique paired with mass spectrometry to identify molecular neighbors of the murine IgM BCR. Those neighbors include MHC class II molecules, integrins, various transporters, and membrane microdomain proteins. Class II molecules, some of which are invariant chain-associated nascent class II, are a readily detected BCR neighbor. This finding is consistent with reports of BCR-class II association within intracellular compartments. The BCR is also in close proximity to multiple proteins involved in the formation of membrane microdomains, including CD37, raftlin, and Ig superfamily member 8. Known defects in T cell-dependent humoral immunity in CD37 knockout mice suggest a role for CD37 in BCR function. In line with this notion, CRISPR-based knockout of CD37 expression in a B cell line heightens BCR signaling, slows BCR endocytosis, and tempers formation of peptide-class II complexes. These results indicate that BCR molecular neighbors can impact membrane-mediated BCR functions. Overall, a proximity-based labeling technique allowed for identification of multiple previously unknown BCR molecular neighbors, including the tetraspanin protein CD37, which can modulate BCR function.
Collapse
Affiliation(s)
- Sean Hoeger
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY
| | - Lisa A. Drake
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY
| | - James R. Drake
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY
| |
Collapse
|
10
|
Stone CA, Spiller BW, Smith SA. Engineering therapeutic monoclonal antibodies. J Allergy Clin Immunol 2024; 153:539-548. [PMID: 37995859 PMCID: PMC11437839 DOI: 10.1016/j.jaci.2023.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/05/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
The use of human antibodies as biologic therapeutics has revolutionized patient care throughout fields of medicine. As our understanding of the many roles antibodies play within our natural immune responses continues to advance, so will the number of therapeutic indications for which an mAb will be developed. The great breadth of function, long half-life, and modular structure allow for nearly limitless therapeutic possibilities. Human antibodies can be rationally engineered to enhance their desired immune functions and eliminate those that may result in unwanted effects. Antibody therapeutics now often start with fully human variable regions, either acquired from genetically engineered humanized mice or from the actual human B cells. These variable genes can be further engineered by widely used methods for optimization of their specificity through affinity maturation, random mutagenesis, targeted mutagenesis, and use of in silico approaches. Antibody isotype selection and deliberate mutations are also used to improve efficacy and tolerability by purposeful fine-tuning of their immune effector functions. Finally, improvements directed at binding to the neonatal Fc receptor can endow therapeutic antibodies with unbelievable extensions in their circulating half-life. The future of engineered antibody therapeutics is bright, with the global mAb market projected to exhibit compound annual growth, forecasted to reach a revenue of nearly half a trillion dollars in 2030.
Collapse
Affiliation(s)
- Cosby A Stone
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Benjamin W Spiller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tenn; Department of Pharmacology, Vanderbilt University, Nashville, Tenn
| | - Scott A Smith
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tenn.
| |
Collapse
|
11
|
Bowman KA, Kaplonek P, McNamara RP. Understanding Fc function for rational vaccine design against pathogens. mBio 2024; 15:e0303623. [PMID: 38112418 PMCID: PMC10790774 DOI: 10.1128/mbio.03036-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Antibodies represent the primary correlate of immunity following most clinically approved vaccines. However, their mechanisms of action vary from pathogen to pathogen, ranging from neutralization, to opsonophagocytosis, to cytotoxicity. Antibody functions are regulated both by antigen specificity (Fab domain) and by the interaction of their Fc domain with distinct types of Fc receptors (FcRs) present in immune cells. Increasing evidence highlights the critical nature of Fc:FcR interactions in controlling pathogen spread and limiting the disease state. Moreover, variation in Fc-receptor engagement during the course of infection has been demonstrated across a range of pathogens, and this can be further influenced by prior exposure(s)/immunizations, age, pregnancy, and underlying health conditions. Fc:FcR functional variation occurs at the level of antibody isotype and subclass selection as well as post-translational modification of antibodies that shape Fc:FcR-interactions. These factors collectively support a model whereby the immune system actively harnesses and directs Fc:FcR interactions to fight disease. By defining the precise humoral mechanisms that control infections, as well as understanding how these functions can be actively tuned, it may be possible to open new paths for improving existing or novel vaccines.
Collapse
Affiliation(s)
- Kathryn A. Bowman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Paulina Kaplonek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
Alemán OR, Rosales C. Human neutrophil Fc gamma receptors: different buttons for different responses. J Leukoc Biol 2023; 114:571-584. [PMID: 37437115 DOI: 10.1093/jleuko/qiad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Neutrophils are fundamental cells in host defense. These leukocytes are quickly recruited from the blood to sites of infection or tissue damage. At these sites, neutrophils initiate several innate immune responses, including phagocytosis, production of reactive oxygen species, degranulation to release proteases and other antimicrobial compounds, production of inflammatory mediators, and formation of neutrophil extracellular traps. In addition to their role in innate immunity, neutrophils are now recognized as cells that also regulate adaptive immunity, via interaction with dendritic cells and lymphocytes. Neutrophils also respond to adaptive immunity by interacting with antibody molecules. Indeed, antibody molecules allow neutrophils to have antigen-specific responses. Neutrophils express different receptors for antibodies. The receptors for immunoglobulin G molecules are known as Fcγ receptors. Upon Fcγ receptor aggregation on the cell membrane, these receptors trigger distinct signal transduction cascades that activate particular cellular responses. In this review, we describe the major Fcγ receptors expressed on human neutrophils and discuss how each Fcγ receptor activates a choice of signaling pathways to stimulate particular neutrophil responses.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apdo. Postal 70228, Ciudad Universitaria, Ciudad de México 04510, México
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apdo. Postal 70228, Ciudad Universitaria, Ciudad de México 04510, México
| |
Collapse
|
13
|
Khalaji A, Yancheshmeh FB, Farham F, Khorram A, Sheshbolouki S, Zokaei M, Vatankhah F, Soleymani-Goloujeh M. Don't eat me/eat me signals as a novel strategy in cancer immunotherapy. Heliyon 2023; 9:e20507. [PMID: 37822610 PMCID: PMC10562801 DOI: 10.1016/j.heliyon.2023.e20507] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 09/04/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023] Open
Abstract
Cancer stands as one of the prominent global causes of death, with its incidence burden continuously increasing, leading to a substantial rise in mortality rates. Cancer treatment has seen the development of various strategies, each carrying its drawbacks that can negatively impact the quality of life for cancer patients. The challenge remains significant within the medical field to establish a definitive cancer treatment that minimizes complications and limitations. In the forthcoming years, exploring new strategies to surmount the failures in cancer treatment appears to be an unavoidable pursuit. Among these strategies, immunology-based ones hold substantial promise in combatting cancer and immune-related disorders. A particular subset of this approach identifies "eat me" and "Don't eat me" signals in cancer cells, contrasting them with their counterparts in non-cancerous cells. This distinction could potentially mark a significant breakthrough in treating diverse cancers. By delving into signal transduction and engineering novel technologies that utilize distinct "eat me" and "Don't eat me" signals, a valuable avenue may emerge for advancing cancer treatment methodologies. Macrophages, functioning as vital components of the immune system, regulate metabolic equilibrium, manage inflammatory disorders, oversee fibrosis, and aid in the repair of injuries. However, in the context of tumor cells, the overexpression of "Don't eat me" signals like CD47, PD-L1, and beta-2 microglobulin (B2M), an anti-phagocytic subunit of the primary histocompatibility complex class I, enables these cells to evade macrophages and proliferate uncontrollably. Conversely, the presentation of an "eat me" signal, such as Phosphatidylserine (PS), along with alterations in charge and glycosylation patterns on the cellular surface, modifications in intercellular adhesion molecule-1 (ICAM-1) epitopes, and the exposure of Calreticulin and PS on the outer layer of the plasma membrane represent universally observed changes on the surface of apoptotic cells, preventing phagocytosis from causing harm to adjacent non-tumoral cells. The current review provides insight into how signaling pathways and immune cells either stimulate or obstruct these signals, aiming to address challenges that may arise in future immunotherapy research. A potential solution lies in combination therapies targeting the "eat me" and "Don't eat me" signals in conjunction with other targeted therapeutic approaches. This innovative strategy holds promise as a novel avenue for the future treatment of cancer.
Collapse
Affiliation(s)
- Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatereh Baharlouei Yancheshmeh
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Farham
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arya Khorram
- Department of Laboratory Sciences, School of Allied Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Shiva Sheshbolouki
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Veterinary Medicine, Beyza Branch, Islamic Azad University, Beyza, Iran
| | - Fatemeh Vatankhah
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Soleymani-Goloujeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
14
|
Van Wagoner CM, Rivera-Escalera F, Delgadillo NJ, Chu CC, Zent CS, Elliott MR. Antibody-mediated phagocytosis in cancer immunotherapy. Immunol Rev 2023; 319:128-141. [PMID: 37602915 PMCID: PMC10615698 DOI: 10.1111/imr.13265] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023]
Abstract
Unconjugated monoclonal antibodies (mAb) have revolutionized the treatment of many types of cancer. Some of these mAbs promote the clearance of malignant cells via direct cytotoxic effects. More recently, antibody-dependent cellular phagocytosis (ADCP) has been appreciated as a major mechanism of action for a number of widely-used mAbs, including anti-CD20 (rituximab, obinutuzumab), anti-HER2 (trazituzumab), and anti-CD38 (daratumumab). However, as a monotherapy these ADCP-inducing mAbs produce insufficient levels of cytotoxicity in vivo and are not curative. As a result, these mAbs are most effectively used in combination therapies. The efficacy of these mAbs is further hampered by the apparent development of drug resistance by many patients. Here we will explore the role of ADCP in cancer immunotherapy and discuss the key factors that could limit the efficacy of ADCP-inducing mAbs in vivo. Finally, we will discuss current insights and approaches being applied to overcome these limitations.
Collapse
Affiliation(s)
- Carly M. Van Wagoner
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Fátima Rivera-Escalera
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | | | - Charles C. Chu
- Division of Hematology/Oncology, University of Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, NY, USA
| | - Clive S. Zent
- Division of Hematology/Oncology, University of Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, NY, USA
| | - Michael R. Elliott
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
15
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley KB, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. GeroScience 2023; 45:3019-3043. [PMID: 37393197 PMCID: PMC10643718 DOI: 10.1007/s11357-023-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023] Open
Abstract
Major histocompatibility complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses, but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here, we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating ribosome affinity purification-qPCR analysis of 3-6- and 18-22-month-old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m, H2-D1, H2-K1, H2-M3, H2-Q6, and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I-binding leukocyte immunoglobulin-like (Lilrs) and paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell -autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A, suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M Kellogg
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Adeline H Machalinski
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Hunter L Porter
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Harris E Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
16
|
Xie MM, Dai B, Hackney JA, Sun T, Zhang J, Jackman JK, Jeet S, Irizarry-Caro RA, Fu Y, Liang Y, Bender H, Shamir ER, Keir ME, Bevers J, Nakamura G, Townsend MJ, Fox DA, Scherl A, Lee WP, Martin F, Godowski PJ, Pappu R, Yi T. An agonistic anti-signal regulatory protein α antibody for chronic inflammatory diseases. Cell Rep Med 2023; 4:101130. [PMID: 37490914 PMCID: PMC10439247 DOI: 10.1016/j.xcrm.2023.101130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
Signal regulatory protein (SIRPα) is an immune inhibitory receptor expressed by myeloid cells to inhibit immune cell phagocytosis, migration, and activation. Despite the progress of SIRPα and CD47 antagonist antibodies to promote anti-cancer immunity, it is not yet known whether SIRPα receptor agonism could restrain excessive autoimmune tissue inflammation. Here, we report that neutrophil- and monocyte-associated genes including SIRPA are increased in inflamed tissue biopsies from patients with rheumatoid arthritis and inflammatory bowel diseases, and elevated SIRPA is associated with treatment-refractory ulcerative colitis. We next identify an agonistic anti-SIRPα antibody that exhibits potent anti-inflammatory effects in reducing neutrophil and monocyte chemotaxis and tissue infiltration. In preclinical models of arthritis and colitis, anti-SIRPα agonistic antibody ameliorates autoimmune joint inflammation and inflammatory colitis by reducing neutrophils and monocytes in tissues. Our work provides a proof of concept for SIRPα receptor agonism for suppressing excessive innate immune activation and chronic inflammatory disease treatment.
Collapse
Affiliation(s)
- Markus M Xie
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Bingbing Dai
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Jason A Hackney
- Department of OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Tianhe Sun
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Juan Zhang
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Janet K Jackman
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Ricardo A Irizarry-Caro
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA; Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Yongyao Fu
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Microchemistry, Proteomics, and Lipidomics and Next Generation Sequencing, Genentech, Inc., South San Francisco, CA, USA
| | - Hannah Bender
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Eliah R Shamir
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Mary E Keir
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Jack Bevers
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Michael J Townsend
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alexis Scherl
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Flavius Martin
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Paul J Godowski
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Rajita Pappu
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Tangsheng Yi
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
17
|
Benadda S, Nugue M, Koumantou D, Bens M, De Luca M, Pellé O, Monteiro RC, Evnouchidou I, Saveanu L. Activating FcγR function depends on endosomal-signaling platforms. iScience 2023; 26:107055. [PMID: 37360697 PMCID: PMC10285637 DOI: 10.1016/j.isci.2023.107055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/02/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Cell surface receptor internalization can either terminate signaling or activate alternative endosomal signaling pathways. We investigated here whether endosomal signaling is involved in the function of the human receptors for Fc immunoglobulin fragments (FcRs): FcαRI, FcγRIIA, and FcγRI. All these receptors were internalized after their cross-linking with receptor-specific antibodies, but their intracellular trafficking was different. FcαRI was targeted directly to lysosomes, while FcγRIIA and FcγRI were internalized in particular endosomal compartments described by the insulin esponsive minoeptidase (IRAP), where they recruited signaling molecules, such as the active form of the kinase Syk, PLCγ and the adaptor LAT. Destabilization of FcγR endosomal signaling in the absence of IRAP compromised cytokine secretion downstream FcγR activation and macrophage ability to kill tumor cells by antibody-dependent cell-mediated cytotoxicity (ADCC). Our results indicate that FcγR endosomal signaling is required for the FcγR-driven inflammatory reaction and possibly for the therapeutic action of monoclonal antibodies.
Collapse
Affiliation(s)
- Samira Benadda
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Mathilde Nugue
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Despoina Koumantou
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Marcelle Bens
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Mariacristina De Luca
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Olivier Pellé
- INSERM UMR 1163, Cell Sorting Facility, Paris, France
- INSERM UMR 1163, Laboratoire of Immunogenetics of Pediatric Autoimmunity, Paris, France
| | - Renato C. Monteiro
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Irini Evnouchidou
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
- Inovarion, Paris, France
| | - Loredana Saveanu
- INSERM U1149, CRI, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université de Paris, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| |
Collapse
|
18
|
Sawant J, Patil A, Kurle S. A Review: Understanding Molecular Mechanisms of Antibody-Dependent Enhancement in Viral Infections. Vaccines (Basel) 2023; 11:1240. [PMID: 37515055 PMCID: PMC10384352 DOI: 10.3390/vaccines11071240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Antibody Dependent Enhancement (ADE) of an infection has been of interest in the investigation of many viruses. It is associated with the severity of the infection. ADE is mediated by non-neutralizing antibodies, antibodies at sub-neutralizing concentrations, or cross-reactive non-neutralizing antibodies. Treatments like plasma therapy, B cell immunizations, and antibody therapies may trigger ADE. It is seen as an impediment to vaccine development as well. In viruses including the Dengue virus (DENV), severe acute respiratory syndrome (SARS) virus, Middle East respiratory syndrome (MERS) virus, human immunodeficiency virus (HIV), Ebola virus, Zika virus, and influenza virus, the likely mechanisms of ADE are postulated and described. ADE improves the likelihood of productively infecting cells that are expressing the complement receptor or the Fc receptor (FcR) rather than the viral receptors. ADE occurs when the FcR, particularly the Fc gamma receptor, and/or complement system, particularly Complement 1q (C1q), allow the entry of the virus-antibody complex into the cell. Moreover, ADE alters the innate immune pathways to escape from lysis, promoting viral replication inside the cell that produces viral particles. This review discusses the involvement of FcR and the downstream immunomodulatory pathways in ADE, the complement system, and innate antiviral signaling pathways modification in ADE and its impact on facilitating viral replication. Additionally, we have outlined the modes of ADE in the cases of different viruses reported until now.
Collapse
Affiliation(s)
- Jyoti Sawant
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| | - Ajit Patil
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| | - Swarali Kurle
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| |
Collapse
|
19
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley K, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531435. [PMID: 36945372 PMCID: PMC10028873 DOI: 10.1101/2023.03.07.531435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Major Histocompatibility Complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating Ribosome Affinity Purification-qPCR analysis of 3-6 and 18-22 month old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m , H2-D1 , H2-K1 , H2-M3 , H2-Q6 , and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I binding Leukocyte Immunoglobulin-like (Lilrs) and Paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell-autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A , suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M. Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Kevin Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Adeline H. Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Hunter L. Porter
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Harris E. Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla Tooley
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Heather C. Rice
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J. Beckstead
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Ana J. Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Sarah R. Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Willard M. Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| |
Collapse
|
20
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [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: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
21
|
Adnan A, Acharya S, Alenazy LA, de las Vecillas L, Giavina Bianchi P, Picard M, Calbache-Gil L, Romero-Pinedo S, Abadí´a-Molina AC, Kerr W, Pedicone C, Nagai J, Hollers E, Dwyer D, Castells M. Multistep IgE Mast Cell Desensitization Is a Dose- and Time-Dependent Process Partially Regulated by SHIP-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:709-720. [PMID: 36881903 PMCID: PMC9986054 DOI: 10.4049/jimmunol.2100485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/22/2022] [Indexed: 03/09/2023]
Abstract
Multistep mast cell desensitization blocks the release of mediators following IgE crosslinking with increasing doses of Ag. Although its in vivo application has led to the safe reintroduction of drugs and foods in IgE-sensitized patients at risk for anaphylaxis, the mechanisms of the inhibitory process have remained elusive. We sought to investigate the kinetics, membrane, and cytoskeletal changes and to identify molecular targets. IgE-sensitized wild-type murine (WT) and FcεRIα humanized (h) bone marrow mast cells were activated and desensitized with DNP, nitrophenyl, dust mites, and peanut Ags. The movements of membrane receptors, FcεRI/IgE/Ag, actin, and tubulin and the phosphorylation of Syk, Lyn, P38-MAPK, and SHIP-1 were assessed. Silencing SHIP-1 protein was used to dissect the SHIP-1 role. Multistep IgE desensitization of WT and transgenic human bone marrow mast cells blocked the release of β-hexosaminidase in an Ag-specific fashion and prevented actin and tubulin movements. Desensitization was regulated by the initial Ag dose, number of doses, and time between doses. FcεRI, IgE, Ags, and surface receptors were not internalized during desensitization. Phosphorylation of Syk, Lyn, p38 MAPK, and SHIP-1 increased in a dose-response manner during activation; in contrast, only SHIP-1 phosphorylation increased in early desensitization. SHIP-1 phosphatase function had no impact on desensitization, but silencing SHIP-1 increased β-hexoxaminidase release, preventing desensitization. Multistep IgE mast cell desensitization is a dose- and time-regulated process that blocks β-hexosaminidase, impacting membrane and cytoskeletal movements. Signal transduction is uncoupled, favoring early phosphorylation of SHIP-1. Silencing SHIP-1 impairs desensitization without implicating its phosphatase function.
Collapse
Affiliation(s)
- Ather Adnan
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Texas A&M Health Science Center, College of Medicine, Houston, TX
| | - Shree Acharya
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Leila A. Alenazy
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Allergy and Clinical Immunology, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Leticia de las Vecillas
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Allergy, Marqués de Valdecilla University Hospital – Instituto de Investigación Marques de Valdecilla, Santander, Spain
| | - Pedro Giavina Bianchi
- Clinical Immunology and Allergy Division, School of Medicine, Universidade de São Paulo, R. Prof. Artur Ramos Sao Paulo, Brazil
| | - Matthieu Picard
- Division of Allergy and Clinical Immunology, Department of Medicine, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, QC, Canada
| | - Lucia Calbache-Gil
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Salvador Romero-Pinedo
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Ana Clara Abadí´a-Molina
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - William Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Chiara Pedicone
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Jun Nagai
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Eleanor Hollers
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Daniel Dwyer
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Mariana Castells
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Rojas-Ortega DA, Rojas-Hernández S, Sánchez-Mendoza ME, Gómez-López M, Sánchez-Camacho JV, Rosales-Cruz E, Yépez MMC. Role of FcγRIII in the nasal cavity of BALB/c mice in the primary amebic meningoencephalitis protection model. Parasitol Res 2023; 122:1087-1105. [PMID: 36913025 PMCID: PMC10009362 DOI: 10.1007/s00436-023-07810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/01/2023] [Indexed: 03/14/2023]
Abstract
Different mechanisms of the host immune response against the primary amebic meningoencephalitis (PAM) in the mouse protection model have been described. It has been proposed that antibodies opsonize Naegleria fowleri trophozoites; subsequently, the polymorphonuclear cells (PMNs) surround the trophozoites to avoid the infection. FcγRs activate signaling pathways of adapter proteins such as Syk and Hck on PMNs to promote different effector cell functions which are induced by the Fc portion of the antibody-antigen complexes. In this work, we analyzed the activation of PMNs, epithelial cells, and nasal passage cells via the expression of Syk and Hck genes. Our results showed an increment of the FcγRIII and IgG subclasses in the nasal cavity from immunized mice as well as Syk and Hck expression was increased, whereas in the in vitro assay, we observed that when the trophozoites of N. fowleri were opsonized with IgG anti-N. fowleri and interacted with PMN, the expression of Syk and Hck was also increased. We suggest that PMNs are activated via their FcγRIII, which leads to the elimination of the trophozoites in vitro, while in the nasal cavity, the adhesion and consequently infection are avoided.
Collapse
Affiliation(s)
- Diego Alexander Rojas-Ortega
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Saúl Rojas-Hernández
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - María Elena Sánchez-Mendoza
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Modesto Gómez-López
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Jennifer Viridiana Sánchez-Camacho
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Erika Rosales-Cruz
- Laboratorio de Investigación en Hematopatología, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Ciudad de Mexico, México
| | | |
Collapse
|
23
|
Willis EF, Gillespie ER, Guse K, Zuercher AW, Käsermann F, Ruitenberg MJ, Vukovic J. Intravenous immunoglobulin (IVIG) promotes brain repair and improves cognitive outcomes after traumatic brain injury in a FcγRIIB receptor-dependent manner. Brain Behav Immun 2023; 109:37-50. [PMID: 36581304 DOI: 10.1016/j.bbi.2022.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is a promising immune-modulatory therapy for limiting harmful inflammation and associated secondary tissue loss in neurotrauma. Here, we show that IVIG therapy attenuates spatial learning and memory deficits following a controlled cortical impact mouse model of traumatic brain injury (TBI). These improvements in cognitive outcomes were associated with increased neuronal survival, an overall reduction in brain tissue loss, and a greater preservation of neural connectivity. Furthermore, we demonstrate that the presence of the main inhibitory FcγRIIB receptor is required for the beneficial effects of IVIG treatment in TBI, with our results simultaneously highlighting the role of this receptor in reducing secondary damage arising from brain injury.
Collapse
Affiliation(s)
- Emily F Willis
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Kirsten Guse
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Adrian W Zuercher
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Fabian Käsermann
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Marc J Ruitenberg
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia; Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
24
|
Cottignies-Calamarte A, Tudor D, Bomsel M. Antibody Fc-chimerism and effector functions: When IgG takes advantage of IgA. Front Immunol 2023; 14:1037033. [PMID: 36817447 PMCID: PMC9933243 DOI: 10.3389/fimmu.2023.1037033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Recent advances in the development of therapeutic antibodies (Abs) have greatly improved the treatment of otherwise drug-resistant cancers and autoimmune diseases. Antibody activities are mediated by both their Fab and the Fc. However, therapeutic Abs base their protective mechanisms on Fc-mediated effector functions resulting in the activation of innate immune cells by FcRs. Therefore, Fc-bioengineering has been widely used to maximise the efficacy and convenience of therapeutic antibodies. Today, IgG remains the only commercially available therapeutic Abs, at the expense of other isotypes. Indeed, production, sampling, analysis and related in vivo studies are easier to perform with IgG than with IgA due to well-developed tools. However, interest in IgA is growing, despite a shorter serum half-life and a more difficult sampling and purification methods than IgG. Indeed, the paradigm that the effector functions of IgG surpass those of IgA has been experimentally challenged. Firstly, IgA has been shown to bind to its Fc receptor (FcR) on effector cells of innate immunity with greater efficiency than IgG, resulting in more robust IgA-mediated effector functions in vitro and better survival of treated animals. In addition, the two isotypes have been shown to act synergistically. From these results, new therapeutic formats of Abs are currently emerging, in particular chimeric Abs containing two tandemly expressed Fc, one from IgG (Fcγ) and one from IgA (Fcα). By binding both FcγR and FcαR on effector cells, these new chimeras showed improved effector functions in vitro that were translated in vivo. Furthermore, these chimeras retain an IgG-like half-life in the blood, which could improve Ab-based therapies, including in AIDS. This review provides the rationale, based on the biology of IgA and IgG, for the development of Fcγ and Fcα chimeras as therapeutic Abs, offering promising opportunities for HIV-1 infected patients. We will first describe the main features of the IgA- and IgG-specific Fc-mediated signalling pathways and their respective functional differences. We will then summarise the very promising results on Fcγ and Fcα containing chimeras in cancer treatment. Finally, we will discuss the impact of Fcα-Fcγ chimerism in prevention/treatment strategies against infectious diseases such as HIV-1.
Collapse
Affiliation(s)
- Andréa Cottignies-Calamarte
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Daniela Tudor
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
25
|
Xiao J, Zhang T, Gao F, Zhou Z, Shu G, Zou Y, Yin G. Natural Killer Cells: A Promising Kit in the Adoptive Cell Therapy Toolbox. Cancers (Basel) 2022; 14:cancers14225657. [PMID: 36428748 PMCID: PMC9688567 DOI: 10.3390/cancers14225657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
As an important component of the innate immune system, natural killer (NK) cells have gained increasing attention in adoptive cell therapy for their safety and efficacious tumor-killing effect. Unlike T cells which rely on the interaction between TCRs and specific peptide-MHC complexes, NK cells are more prone to be served as "off-the-shelf" cell therapy products due to their rapid recognition and killing of tumor cells without MHC restriction. In recent years, constantly emerging sources of therapeutic NK cells have provided flexible options for cancer immunotherapy. Advanced genetic engineering techniques, especially chimeric antigen receptor (CAR) modification, have yielded exciting effectiveness in enhancing NK cell specificity and cytotoxicity, improving in vivo persistence, and overcoming immunosuppressive factors derived from tumors. In this review, we highlight current advances in NK-based adoptive cell therapy, including alternative sources of NK cells for adoptive infusion, various CAR modifications that confer different targeting specificity to NK cells, multiple genetic engineering strategies to enhance NK cell function, as well as the latest clinical research on adoptive NK cell therapy.
Collapse
Affiliation(s)
- Jiani Xiao
- Department of Pathology, School of Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Fei Gao
- Department of Pathology, School of Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zhengwei Zhou
- Department of Pathology, School of Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Guang Shu
- Department of Pathology, School of Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine, Central South University, Changsha 410000, China
- Correspondence: (Y.Z.); (G.Y.)
| | - Gang Yin
- Department of Pathology, School of Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha 410000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410000, China
- Correspondence: (Y.Z.); (G.Y.)
| |
Collapse
|
26
|
Oosterhoff JJ, Larsen MD, van der Schoot CE, Vidarsson G. Afucosylated IgG responses in humans - structural clues to the regulation of humoral immunity. Trends Immunol 2022; 43:800-814. [PMID: 36008258 PMCID: PMC9395167 DOI: 10.1016/j.it.2022.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
Healthy immune responses require efficient protection without excessive inflammation. Recent discoveries on the degree of fucosylation of a human N-linked glycan at a conserved site in the immunoglobulin IgG-Fc domain might add an additional regulatory layer to adaptive humoral immunity. Specifically, afucosylation of IgG-Fc enhances the interaction of IgG with FcγRIII and thereby its activity. Although plasma IgG is generally fucosylated, afucosylated IgG is raised in responses to enveloped viruses and Plasmodium falciparum proteins expressed on infected erythrocytes, as well as during alloimmune responses. Moreover, while afucosylation can exacerbate some infectious diseases (e.g., COVID-19), it also correlates with traits of protective immunity against malaria and HIV-1. Herein we discuss the implications of IgG afucosylation for health and disease, as well as for vaccination.
Collapse
Affiliation(s)
- Janita J Oosterhoff
- Immunoglobulin Research Laboratory, Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Mads Delbo Larsen
- Immunoglobulin Research Laboratory, Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - C Ellen van der Schoot
- Immunoglobulin Research Laboratory, Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands; Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Immunoglobulin Research Laboratory, Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
27
|
Li Y, Lv C, Yu Y, Wu B, Zhang Y, Lang Q, Liang Z, Zhong C, Shi Y, Han S, Xu F, Tian Y. KIR3DL3-HHLA2 and TMIGD2-HHLA2 pathways: The dual role of HHLA2 in immune responses and its potential therapeutic approach for cancer immunotherapy. J Adv Res 2022; 47:137-150. [PMID: 35933091 PMCID: PMC10173190 DOI: 10.1016/j.jare.2022.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/15/2022] [Accepted: 07/29/2022] [Indexed: 10/16/2022] Open
Abstract
BACKGROUND T cells and natural killer (NK) cells are essential components of the immune system and are regulated by coinhibitory and costimulatory molecules in which the B7 family and CD28 family play significant roles. Previous immune checkpoint studies on B7/CD28 family members, such as PD-1, have led to remarkable success in cancer immunotherapy. However, there is still a need to find new immune checkpoint molecules. Recent studies have demonstrated that HHLA2 exerts inhibitory and stimulatory functions on the immune system by binding to different receptors on different sites. However, the pathways between HHLA2 and its two receptors on T cells and NK cells remain controversial. AIM OF REVIEW Here, we reviewed recent studies about HHLA2 ligand interactions with KIR3DL3 and TMIGD2. We focused on elucidating the pathways between KIR3DL3/TMIGD2 and HHLA2 as well as their function in tumour progression. We also addressed the relationship between HHLA2 expression and the clinical prognosis of cancer patients. KEY SCIENTIFIC CONCEPTS OF REVIEW KIR3DL3/TMIGD2-HHLA2 may represent novel pathways within the tumour microenvironment and serve as crucial immune checkpoints for developing novel therapeutic drugs against human cancer.
Collapse
Affiliation(s)
- Yang Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Yang Yu
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Yizhou Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Qi Lang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Zhiyun Liang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Yu Shi
- The First Clinical College of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Shukun Han
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China.
| |
Collapse
|
28
|
Gonzalez JC, Chakraborty S, Thulin NK, Wang TT. Heterogeneity in IgG-CD16 signaling in infectious disease outcomes. Immunol Rev 2022; 309:64-74. [PMID: 35781671 PMCID: PMC9539944 DOI: 10.1111/imr.13109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Joseph C. Gonzalez
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Saborni Chakraborty
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA
| | - Natalie K. Thulin
- Department of ImmunologyUniversity of WashingtonSeattleWashingtonUSA
| | - Taia T. Wang
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA,Department of Microbiology and ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
29
|
Simpson AP, Roghanian A, Oldham RJ, Chan HTC, Penfold CA, Kim HJ, Inzhelevskaya T, Mockridge CI, Cox KL, Bogdanov YD, James S, Tutt AL, Rycroft D, Morley P, Dahal LN, Teige I, Frendeus B, Beers SA, Cragg MS. FcγRIIB controls antibody-mediated target cell depletion by ITIM-independent mechanisms. Cell Rep 2022; 40:111099. [PMID: 35858562 PMCID: PMC9638011 DOI: 10.1016/j.celrep.2022.111099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/10/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
Many therapeutic antibodies deplete target cells and elicit immunotherapy by engaging activating Fc gamma receptors (FcγRs) on host effector cells. These antibodies are negatively regulated by the inhibitory FcγRIIB (CD32B). Dogma suggests inhibition is mediated through the FcγRIIB immunoreceptor tyrosine-based inhibition motif (ITIM), negatively regulating immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling from activating FcγR. To assess this, we generated experimental models expressing human (h)FcγRIIB on targets or effectors, lacking or retaining ITIM signaling capacity. We demonstrate that signaling through the hFcγRIIB ITIM is dispensable for impairing monoclonal antibody (mAb)-mediated depletion of normal and malignant murine target cells through three therapeutically relevant surface receptors (CD20, CD25, and OX40) affecting immunotherapy. We demonstrate that hFcγRIIB competition with activating FcγRs for antibody Fc, rather than ITIM signaling, is sufficient to impair activating FcγR engagement, inhibiting effector function and immunotherapy.
Collapse
Affiliation(s)
- Alexander P Simpson
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Robert J Oldham
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - H T Claude Chan
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Christine A Penfold
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Hyung J Kim
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Tatyana Inzhelevskaya
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - C Ian Mockridge
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Kerry L Cox
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Yury D Bogdanov
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Sonya James
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Alison L Tutt
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Daniel Rycroft
- Biopharm Discovery, GSK, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - Peter Morley
- Biopharm Discovery, GSK, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Ingrid Teige
- BioInvent International AB, Sölvegatan 41, 22370 Lund, Sweden
| | - Björn Frendeus
- BioInvent International AB, Sölvegatan 41, 22370 Lund, Sweden.
| | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
30
|
El Ansari YS, Kanagaratham C, Burton OT, Santos JV, Hollister BMA, Lewis OL, Renz H, Oettgen HC. Allergen-Specific IgA Antibodies Block IgE-Mediated Activation of Mast Cells and Basophils. Front Immunol 2022; 13:881655. [PMID: 35865546 PMCID: PMC9294179 DOI: 10.3389/fimmu.2022.881655] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mast cells and basophils have long been implicated in the pathogenesis of IgE-mediated hypersensitivity reactions. They express the high-affinity IgE receptor, FcϵRI, on their surface. Antigen-induced crosslinking of IgE antibodies bound to that receptor triggers a signaling cascade that results in activation, leading to the release of an array of preformed vasoactive mediators and rapidly synthesized lipids, as well as the de novo production of inflammatory cytokines. In addition to bearing activating receptors like FcεRI, these effector cells of allergy express inhibitory ones including FcγR2b, an IgG Fc receptor with a cytosolic inhibitory motif that activates protein tyrosine phosphatases that suppress IgE-mediated activation. We and others have shown that food allergen-specific IgG antibodies strongly induced during the course of oral immunotherapy (OIT), signal via FcγR2b to suppress IgE-mediated mast cell and basophil activation triggered by food allergen challenge. However, the potential inhibitory effects of IgA antibodies, which are also produced in response to OIT and are present at high levels at mucosal sites, including the intestine where food allergens are encountered, have not been well studied. Here we uncover an inhibitory function for IgA. We observe that IgA binds mouse bone marrow-derived mast cells (BMMCs) and peritoneal mast cells. Binding to BMMCs is dependent on calcium and sialic acid. We also found that IgA antibodies inhibit IgE-mediated mast cell degranulation in an allergen-specific fashion. Antigen-specific IgA inhibits IgE-mediated mast cell activation early in the signaling cascade, suppressing the phosphorylation of Syk, the proximal protein kinase mediating FcεRI signaling, and suppresses mast cell production of cytokines. Furthermore, using basophils from a peanut allergic donor we found that IgA binds to basophils and that activation by exposure to peanuts is effectively suppressed by IgA. We conclude that IgA serves as a regulator of mast cell and basophil degranulation, suggesting a physiologic role for IgA in the maintenance of immune homeostasis at mucosal sites.
Collapse
Affiliation(s)
- Yasmeen S. El Ansari
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Oliver T. Burton
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Jenna V. Santos
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | | | - Owen L. Lewis
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Harald Renz
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- *Correspondence: Hans C. Oettgen,
| |
Collapse
|
31
|
Mata-Molanes JJ, Rebollo-Liceaga J, Martínez-Navarro EM, Manzano RG, Brugarolas A, Juan M, Sureda M. Relevance of Fc Gamma Receptor Polymorphisms in Cancer Therapy With Monoclonal Antibodies. Front Oncol 2022; 12:926289. [PMID: 35814459 PMCID: PMC9263556 DOI: 10.3389/fonc.2022.926289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs), including immune checkpoint inhibitors (ICIs), are an important breakthrough for the treatment of cancer and have dramatically changed clinical outcomes in a wide variety of tumours. However, clinical response varies among patients receiving mAb-based treatment, so it is necessary to search for predictive biomarkers of response to identify the patients who will derive the greatest therapeutic benefit. The interaction of mAbs with Fc gamma receptors (FcγR) expressed by innate immune cells is essential for antibody-dependent cellular cytotoxicity (ADCC) and this binding is often critical for their in vivo efficacy. FcγRIIa (H131R) and FcγRIIIa (V158F) polymorphisms have been reported to correlate with response to therapeutic mAbs. These polymorphisms play a major role in the affinity of mAb receptors and, therefore, can exert a profound impact on antitumor response in these therapies. Furthermore, recent reports have revealed potential mechanisms of ICIs to modulate myeloid subset composition within the tumour microenvironment through FcγR-binding, optimizing their anti-tumour activity. The purpose of this review is to highlight the clinical contribution of FcγR polymorphisms to predict response to mAbs in cancer patients.
Collapse
Affiliation(s)
- Juan J. Mata-Molanes
- Oncology Platform, Hospital Quirónsalud Torrevieja, Alicante, Spain
- *Correspondence: Juan J. Mata-Molanes,
| | | | | | | | | | - Manel Juan
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Manuel Sureda
- Oncology Platform, Hospital Quirónsalud Torrevieja, Alicante, Spain
| |
Collapse
|
32
|
Zhao P, Xu Y, Jiang LL, Fan X, Ku Z, Li L, Liu X, Deng M, Arase H, Zhu JJ, Huang TY, Zhao Y, Zhang C, Xu H, Tong Q, Zhang N, An Z. LILRB2-mediated TREM2 signaling inhibition suppresses microglia functions. Mol Neurodegener 2022; 17:44. [PMID: 35717259 PMCID: PMC9206387 DOI: 10.1186/s13024-022-00550-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 06/08/2022] [Indexed: 12/18/2022] Open
Abstract
Background Microglia plays crucial roles in Alzheimer’s disease (AD) development. Triggering receptor expressed on myeloid cells 2 (TREM2) in association with DAP12 mediates signaling affecting microglia function. Here we study the negative regulation of TREM2 functions by leukocyte immunoglobulin-like receptor subfamily B member 2 (LILRB2), an inhibitory receptor bearing ITIM motifs. Methods To specifically interrogate LILRB2-ligand (oAβ and PS) interactions and microglia functions, we generated potent antagonistic LILRB2 antibodies with sub-nanomolar level activities. The biological effects of LILRB2 antagonist antibody (Ab29) were studied in human induced pluripotent stem cell (iPSC)–derived microglia (hMGLs) for migration, oAβ phagocytosis, and upregulation of inflammatory cytokines. Effects of the LILRB2 antagonist antibody on microglial responses to amyloid plaques were further studied in vivo using stereotaxic grafted microglia in 5XFAD mice. Results We confirmed the expression of both LILRB2 and TREM2 in human brain microglia using immunofluorescence. Upon co-ligation of the LILRB2 and TREM2 by shared ligands oAβ or PS, TREM2 signaling was significantly inhibited. We identified a monoclonal antibody (Ab29) that blocks LILRB2/ligand interactions and prevents TREM2 signaling inhibition mediated by LILRB2. Further, Ab29 enhanced microglia phagocytosis, TREM2 signaling, migration, and cytokine responses to the oAβ-lipoprotein complex in hMGL and microglia cell line HMC3. In vivo studies showed significantly enhanced clustering of microglia around plaques with a prominent increase in microglial amyloid plaque phagocytosis when 5XFAD mice were treated with Ab29. Conclusions This study revealed for the first time the molecular mechanisms of LILRB2-mediated inhibition of TREM2 signaling in microglia and demonstrated a novel approach of enhancing TREM2-mediated microglia functions by blocking LILRB2-ligand interactions. Translationally, a LILRB2 antagonist antibody completely rescued the inhibition of TREM2 signaling by LILRB2, suggesting a novel therapeutic strategy for improving microglial functions. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-022-00550-y.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yuanzhong Xu
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lu-Lin Jiang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaoye Liu
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mi Deng
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Jay-Jiguang Zhu
- Department of Neurosurgery, University of Texas Health Science Center in Houston, McGovern Medical School and Memorial Hermann, Houston, TX, USA
| | - Timothy Y Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Yingjun Zhao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chengcheng Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Huaxi Xu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qingchun Tong
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
33
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
34
|
Delidakis G, Kim JE, George K, Georgiou G. Improving Antibody Therapeutics by Manipulating the Fc Domain: Immunological and Structural Considerations. Annu Rev Biomed Eng 2022; 24:249-274. [PMID: 35363537 PMCID: PMC9648538 DOI: 10.1146/annurev-bioeng-082721-024500] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interactions between the crystallizable fragment (Fc) domain of antibodies and a plethora of cellular Fc receptors (FcRs) or soluble proteins form a critical link between humoral and innate immunity. In particular, the immunoglobulin G Fc domain is critical for the clearance of target cells by processes that include (a) cytotoxicity, phagocytosis, or complement lysis; (b) modulation of inflammation; (c) antigen presentation; (d) antibody-mediated receptor clustering; and (e) cytokine release. More than 30 Fc-engineered antibodies aimed primarily at tailoring these effects for optimal therapeutic outcomes are in clinical evaluation or have already been approved. Nonetheless, our understanding of how FcR engagement impacts various immune cell phenotypes is still largely incomplete. Recent insights into FcR biology coupled with advances in Fc:FcR structural analysis, Fc engineering, and mouse models that recapitulate human biology are helping to fill in existing knowledge gaps. These advances will provide a blueprint on how to fine-tune the Fc domain to achieve optimal therapeutic efficacy. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA;
| | - Jin Eyun Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Katia George
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA; .,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
35
|
Chen H, Maul‐Pavicic A, Holzer M, Huber M, Salzer U, Chevalier N, Voll RE, Hengel H, Kolb P. Detection and functional resolution of soluble immune complexes by an FcγR reporter cell panel. EMBO Mol Med 2022; 14:e14182. [PMID: 34842342 PMCID: PMC8749491 DOI: 10.15252/emmm.202114182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Fc-gamma receptor (FcγR) activation by soluble IgG immune complexes (sICs) represents a major mechanism of inflammation in certain autoimmune diseases such as systemic lupus erythematosus (SLE). A robust and scalable test system allowing for the detection and quantification of sIC bioactivity is missing. We developed a comprehensive reporter cell panel detecting activation of FcγRs. The reporter cell lines were integrated into an assay that enables the quantification of sIC reactivity via ELISA or a faster detection using flow cytometry. This identified FcγRIIA(H) and FcγRIIIA as the most sIC-sensitive FcγRs in our test system. Reaching a detection limit in the very low nanomolar range, the assay proved also to be sensitive to sIC stoichiometry and size reproducing for the first time a complete Heidelberger-Kendall curve in terms of immune receptor activation. Analyzing sera from SLE patients and mouse models of lupus and arthritis proved that sIC-dependent FcγR activation has predictive capabilities regarding severity of SLE disease. The assay provides a sensitive and scalable tool to evaluate the size, amount, and bioactivity of sICs in all settings.
Collapse
Affiliation(s)
- Haizhang Chen
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Andrea Maul‐Pavicic
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Center for Chronic Immunodeficiency (CCI)Medical Center‐University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Martin Holzer
- Institute for Pharmaceutical SciencesAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Magdalena Huber
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Ulrich Salzer
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Nina Chevalier
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical ImmunologyMedical Center – University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Center for Chronic Immunodeficiency (CCI)Medical Center‐University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Hartmut Hengel
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Philipp Kolb
- Institute of VirologyUniversity Medical CenterAlbert‐Ludwigs‐University FreiburgFreiburgGermany
- Faculty of MedicineAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
36
|
Jin J, Liu Y, Xu X, Wang Z, Niu J. The association between Fc gamma RIIb expression levels and chronic hepatitis B virus infection progression. BMC Infect Dis 2021; 21:1235. [PMID: 34879827 PMCID: PMC8653572 DOI: 10.1186/s12879-021-06918-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fc gamma receptor IIb (FcγRIIb) is an important inhibitory receptor that plays vital roles in regulating various immune response processes and the pathogenesis of many infectious diseases. The purpose of our research was to evaluate FcγRIIb expression in serum and liver biopsy specimens from hepatitis B virus (HBV)-infected patients and to explore the association of FcγRIIb with chronic HBV infection. METHODS Enzyme-linked immunosorbent assay (ELISA) was adopted to measure the serum FcγRIIb levels in 119 HBV-infected patients and 24 healthy controls. An immunohistochemical method was then employed to identify FcγRIIb expression in biopsy specimens from patients with chronic hepatitis B (CHB). The integrated optical density (IOD) value was measured to represent FcγRIIb expression levels. RESULTS Serum FcγRIIb levels were decreased in CHB patients compared to controls (P < 0.001). The FcγRIIb levels in the CHB patient group were remarkably lower than those in the HBV carrier group (P < 0.001). In addition, FcγRIIb levels were negatively associated with AST and ALT (r = -0.3936, P = 0.0063; r = -0.3459, P = 0.0097, respectively). The IOD values of FcγRIIb expression in the moderate and severe CHB groups were significantly lower than those in the control group (P = 0.006 and P < 0.001, respectively). The FcγRIIb level tended to be lower with pathological changes related to hepatitis. Furthermore, correlation analysis revealed that FcγRIIb had negative correlations with AST and ALT (r = -0.688, P = 0.0016; r = -0.686, P = 0.0017, respectively) but a positive association with the platelet count (r = 0.6464, P = 0.0038). CONCLUSIONS FcγRIIb levels are significantly related to chronic HBV infection and the progression of CHB. Changes in FcγRIIb may affect the progression of liver inflammation and fibrosis in CHB patients.
Collapse
Affiliation(s)
- Jinglan Jin
- Department of Hepatology, First Bethune Hospital of Jilin University, 71 Xin Min Street, Changchun, Jilin, 130021, People's Republic of China
| | - Yuwei Liu
- Department of Hepatology, First Bethune Hospital of Jilin University, 71 Xin Min Street, Changchun, Jilin, 130021, People's Republic of China
| | - Xiaotong Xu
- Department of Hepatology, First Bethune Hospital of Jilin University, 71 Xin Min Street, Changchun, Jilin, 130021, People's Republic of China
| | - Zhongfeng Wang
- Department of Hepatology, First Bethune Hospital of Jilin University, 71 Xin Min Street, Changchun, Jilin, 130021, People's Republic of China
| | - Junqi Niu
- Department of Hepatology, First Bethune Hospital of Jilin University, 71 Xin Min Street, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
37
|
DeLuca JM, Murphy MK, Wang X, Wilson TJ. FCRL1 Regulates B Cell Receptor-Induced ERK Activation through GRB2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2688-2698. [PMID: 34697226 PMCID: PMC8629370 DOI: 10.4049/jimmunol.2100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Regulation of BCR signaling has important consequences for generating effective Ab responses to pathogens and preventing production of autoreactive B cells during development. Currently defined functions of Fc receptor-like (FCRL) 1 include positive regulation of BCR-induced calcium flux, proliferation, and Ab production; however, the mechanistic basis of FCRL1 signaling and its contributions to B cell development remain undefined. Molecular characterization of FCRL1 signaling shows phosphotyrosine-dependent associations with GRB2, GRAP, SHIP-1, and SOS1, all of which can profoundly influence MAPK signaling. In contrast with previous characterizations of FCRL1 as a strictly activating receptor, we discover a role for FCRL1 in suppressing ERK activation under homeostatic and BCR-stimulated conditions in a GRB2-dependent manner. Our analysis of B cells in Fcrl1 -/- mice shows that ERK suppression by FCRL1 is associated with a restriction in the number of cells surviving splenic maturation in vivo. The capacity of FCRL1 to modulate ERK activation presents a potential for FCRL1 to be a regulator of peripheral B cell tolerance, homeostasis, and activation.
Collapse
Affiliation(s)
- Jenna M DeLuca
- Department of Microbiology, Miami University, Oxford, OH
| | | | - Xin Wang
- Department of Microbiology, Miami University, Oxford, OH
| | | |
Collapse
|
38
|
Hu W, Bagramyan K, Bhatticharya S, Hong T, Tapia A, Wong P, Kalkum M, Shively JE. Phosphorylation of human CEACAM1-LF by PKA and GSK3β promotes its interaction with β-catenin. J Biol Chem 2021; 297:101305. [PMID: 34656562 PMCID: PMC8564729 DOI: 10.1016/j.jbc.2021.101305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
CEACAM1-LF, a homotypic cell adhesion adhesion molecule, transduces intracellular signals via a 72 amino acid cytoplasmic domain that contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and a binding site for β-catenin. Phosphorylation of Ser503 by PKC in rodent CEACAM1 was shown to affect bile acid transport or hepatosteatosis via the level of ITIM phosphorylation, but the phosphorylation of the equivalent residue in human CEACAM1 (Ser508) was unclear. Here we studied this analogous phosphorylation by NMR analysis of the 15N labeled cytoplasmic domain peptide. Incubation with a variety of Ser/Thr kinases revealed phosphorylation of Ser508 by GSK3bβ but not by PKC. The lack of phosphorylation by PKC is likely due to evolutionary sequence changes between the rodent and human genes. Phosphorylation site assignment by mass spectrometry and NMR revealed phosphorylation of Ser472, Ser461 and Ser512 by PKA, of which Ser512 is part of a conserved consensus site for GSK3β binding. We showed here that only after phosphorylation of Ser512 by PKA was GSK3β able to phosphorylate Ser508. Phosphorylation of Ser512 by PKA promoted a tight association with the armadillo repeat domain of β-catenin at an extended region spanning the ITIMs of CEACAM1. The kinetics of phosphorylation of the ITIMs by Src, as well dephosphorylation by SHP2, were affected by the presence of Ser508/512 phosphorylation, suggesting that PKA and GSK3β may regulate the signal transduction activity of human CEACAM1-LF. The interaction of CEACAM1-LF with β-catenin promoted by PKA is suggestive of a tight association between the two ITIMs of CEACAM1-LF.
Collapse
Affiliation(s)
- Weidong Hu
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Karine Bagramyan
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Supriyo Bhatticharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Teresa Hong
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Alonso Tapia
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Patty Wong
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - John E Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA.
| |
Collapse
|
39
|
Macri C, Morgan H, Villadangos JA, Mintern JD. Regulation of dendritic cell function by Fc-γ-receptors and the neonatal Fc receptor. Mol Immunol 2021; 139:193-201. [PMID: 34560415 DOI: 10.1016/j.molimm.2021.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023]
Abstract
Dendritic cells (DCs) express receptors to sense pathogens and/or tissue damage and to communicate with other immune cells. Among those receptors, Fc receptors (FcRs) are triggered by the Fc region of antibodies produced during adaptive immunity. In this review, the role of FcγR and neonatal Fc receptor (FcRn) in DC immunity will be discussed. Their expression in DC subsets and impact on antigen uptake and presentation, DC maturation and polarisation of T cell responses will be described. Lastly, we will discuss the importance of FcR-mediated DC function in the context of immunity during viral infection, inflammatory disease, cancer and immunotherapy.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Huw Morgan
- ACRF Translational Research Laboratory, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, 3050, Australia; Department of Medicine, University of Melbourne, Parkville, Melbourne, Victoria, 3010, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
40
|
Zillikens H, Kasprick A, Osterloh C, Gross N, Radziewitz M, Hass C, Hartmann V, Behnen-Härer M, Ernst N, Boch K, Vidarsson G, Visser R, Laskay T, Yu X, Petersen F, Ludwig RJ, Bieber K. Topical Application of the PI3Kβ-Selective Small Molecule Inhibitor TGX-221 Is an Effective Treatment Option for Experimental Epidermolysis Bullosa Acquisita. Front Med (Lausanne) 2021; 8:713312. [PMID: 34557502 PMCID: PMC8452940 DOI: 10.3389/fmed.2021.713312] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/17/2021] [Indexed: 11/23/2022] Open
Abstract
Class I phosphoinositide 3-kinases (PI3K) have been implemented in pathogenesis of experimental epidermolysis bullosa acquisita (EBA), an autoimmune skin disease caused by type VII collagen (COL7) autoantibodies. Mechanistically, inhibition of specific PI3K isoforms, namely PI3Kβ or PI3Kδ, impaired immune complex (IC)-induced neutrophil activation, a key prerequisite for EBA pathogenesis. Data unrelated to EBA showed that neutrophil activation is also modulated by PI3Kα and γ, but their impact on the EBA has, so far, remained elusive. To address this and to identify potential therapeutic targets, we evaluated the impact of a panel of PI3K isoform-selective inhibitors (PI3Ki) on neutrophil function in vitro, and in pre-clinical EBA mouse models. We document that distinctive, and EBA pathogenesis-related activation-induced neutrophil in vitro functions depend on distinctive PI3K isoforms. When mice were treated with the different PI3Ki, selective blockade of PI3Kα (alpelisib), PI3Kγ (AS-604850), or PI3Kβ (TGX-221) impaired clinical disease manifestation. When applied topically, only TGX-221 impaired induction of experimental EBA. Ultimately, multiplex kinase activity profiling in the presence of disease-modifying PI3Ki identified unique signatures of different PI3K isoform-selective inhibitors on the kinome of IC-activated human neutrophils. Collectively, we here identify topical PI3Kβ inhibition as a potential therapeutic target for the treatment of EBA.
Collapse
Affiliation(s)
- Hannah Zillikens
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Anika Kasprick
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Natalie Gross
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Michael Radziewitz
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Cindy Hass
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Veronika Hartmann
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Martina Behnen-Härer
- Department for Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Nancy Ernst
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Katharina Boch
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Remco Visser
- Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Tamás Laskay
- Department for Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Xinhua Yu
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Frank Petersen
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| |
Collapse
|
41
|
Shin C, Kim SS, Jo YH. Extending traditional antibody therapies: Novel discoveries in immunotherapy and clinical applications. Mol Ther Oncolytics 2021; 22:166-179. [PMID: 34514097 PMCID: PMC8416972 DOI: 10.1016/j.omto.2021.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Immunotherapy has been well regarded as one of the safer and antigen-specific anti-cancer treatments compared to first-generation chemotherapy. Since Coley's discovery, researchers focused on engineering novel antibody-based therapies. Including artificial and modified antibodies, such as antibody fragments, antibody-drug conjugates, and synthetic mimetics, the variety of immunotherapy has been rapidly expanding in the last few decades. Genetic and chemical modifications to monoclonal antibody have been brought into academia, in vivo trials, and clinical applications. Here, we have looked around antibodies overall. First, we elucidate the antibody structure and its cytotoxicity mechanisms. Second, types of therapeutic antibodies are presented. Additionally, there is a summarized list of US Food and Drug Administration (FDA)-approved therapeutic antibodies and recent clinical trials. This review provides a comprehensive overview of both the general function of therapeutic antibodies and a few main variations in development, including recent advent with the proposed mechanism of actions, and we introduce types of therapeutic antibodies, clinical trials, and approved commercial immunotherapeutic drugs.
Collapse
Affiliation(s)
- Charles Shin
- Chadwick International, Incheon 22002, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Hwa Jo
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
42
|
Phitthayaphong P, Kumfu S, Chattipakorn N, Chattipakorn SC. Blockage of Fc Gamma Receptors Alleviates Neuronal and Microglial Toxicity Induced by Palmitic Acid. J Alzheimers Dis 2021; 82:1315-1332. [PMID: 34151811 DOI: 10.3233/jad-210417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Palmitic acid (PA) promotes brain pathologies including Alzheimer's disease (AD)-related proteins, neuroinflammation, and microglial activation. The activation of neurons and microglia via their Fc gamma receptors (FcγRs) results in producing inflammatory cytokines. OBJECTIVE To investigate the expression of FcγRs, FcγR signaling proteins, AD-related proteins, proinflammatory cytokines, and cell viability of neurons and microglia in association with PA exposure as well as the effects of FcγR blockade on these parameters in response to PA. METHODS 200 and 400μM PA-conjugated BSA were applied to SH-SY5Y and HMC3 cells for 24 h. For FcγR blockage experiment, both cells were exposed to FcγR blocker before receiving of 200 and 400μM of PA-conjugated BSA for 24 h. RESULTS PA significantly increased AD-related proteins, including Aβ and BACE1, as well as increasing TNFα, IL-1β, and IL-6 in SH-SY5Y and HMC3 cells. However, the p-Tau/Tau ratio was only increased in SH-SY5Y cells. These results were associated with an increase in FcγRs activation and a decrease in cell viability in both cell types. FcγRs blockage diminished the activation of FcγR in SH-SY5Y and HMC3 cells. Interestingly, blocking FcγRs before PA exposure reduced the increment of AD-related proteins, proinflammatory cytokines caused by PA. FcγRs blocking also inhibits cell death for 23%of SH-SY5Y cells and 64%of HMC3 cells, respectively. CONCLUSION These findings suggest that PA is a risk factor for AD via the increased AD-related pathologies, inflammation, FcγRs activation, and brain cell death, while FcγR blockage can alleviate these effects.
Collapse
Affiliation(s)
- Phansa Phitthayaphong
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinart Kumfu
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
43
|
Kelley SM, Ravichandran KS. Putting the brakes on phagocytosis: "don't-eat-me" signaling in physiology and disease. EMBO Rep 2021; 22:e52564. [PMID: 34041845 DOI: 10.15252/embr.202152564] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Timely removal of dying or pathogenic cells by phagocytes is essential to maintaining host homeostasis. Phagocytes execute the clearance process with high fidelity while sparing healthy neighboring cells, and this process is at least partially regulated by the balance of "eat-me" and "don't-eat-me" signals expressed on the surface of host cells. Upon contact, eat-me signals activate "pro-phagocytic" receptors expressed on the phagocyte membrane and signal to promote phagocytosis. Conversely, don't-eat-me signals engage "anti-phagocytic" receptors to suppress phagocytosis. We review the current knowledge of don't-eat-me signaling in normal physiology and disease contexts where aberrant don't-eat-me signaling contributes to pathology.
Collapse
Affiliation(s)
- Shannon M Kelley
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,VIB-UGent Center for Inflammation Research, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
44
|
Geyer CE, Mes L, Newling M, den Dunnen J, Hoepel W. Physiological and Pathological Inflammation Induced by Antibodies and Pentraxins. Cells 2021; 10:1175. [PMID: 34065953 PMCID: PMC8150799 DOI: 10.3390/cells10051175] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages play a key role in induction of inflammatory responses. These inflammatory responses are mostly considered to be instigated by activation of pattern recognition receptors (PRRs) or cytokine receptors. However, recently it has become clear that also antibodies and pentraxins, which can both activate Fc receptors (FcRs), induce very powerful inflammatory responses by macrophages that can even be an order of magnitude greater than PRRs. While the physiological function of this antibody-dependent inflammation (ADI) is to counteract infections, undesired activation or over-activation of this mechanism will lead to pathology, as observed in a variety of disorders, including viral infections such as COVID-19, chronic inflammatory disorders such as Crohn's disease, and autoimmune diseases such as rheumatoid arthritis. In this review we discuss how physiological ADI provides host defense by inducing pathogen-specific immunity, and how erroneous activation of this mechanism leads to pathology. Moreover, we will provide an overview of the currently known signaling and metabolic pathways that underlie ADI, and how these can be targeted to counteract pathological inflammation.
Collapse
Affiliation(s)
- Chiara Elisabeth Geyer
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Lynn Mes
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Melissa Newling
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jeroen den Dunnen
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Willianne Hoepel
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
45
|
Liang X, Cao Y, Li C, Yu H, Yang C, Liu H. MALT1 as a promising target to treat lymphoma and other diseases related to MALT1 anomalies. Med Res Rev 2021; 41:2388-2422. [PMID: 33763890 DOI: 10.1002/med.21799] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a key adaptor protein that regulates the NF-κB pathway, in which MALT1 functions as a scaffold protein and protease to trigger downstream signals. The abnormal expression of MALT1 is closely associated with lymphomagenesis and other diseases, including solid tumors and autoimmune diseases. MALT1 is the only protease in the underlying pathogenesis of these diseases, and its proteolytic activity can be pharmacologically regulated. Therefore, MALT1 is a potential and promising target for anti-lymphoma and other MALT1-related disease treatments. Currently, the development of MALT1 inhibitors is still in its early stages. This review presents an overview of MALT1, particularly its X-ray structures and biological functions, and elaborates on the pathogenesis of diseases associated with its dysregulation. We then summarize previously reported MALT1 inhibitors, focusing on their molecular structure, biological activity, structure-activity relationship, and limitations. Finally, we propose future research directions to accelerate the discovery of novel MALT1 inhibitors with clinical applications. Overall, this review provides a comprehensive and systematic overview of MALT1-related research advances and serves as a theoretical basis for drug discovery and research.
Collapse
Affiliation(s)
- Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - YiChun Cao
- School of Pharmacy, Fudan University, Shanghai, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
46
|
Dixon KJ, Wu J, Walcheck B. Engineering Anti-Tumor Monoclonal Antibodies and Fc Receptors to Enhance ADCC by Human NK Cells. Cancers (Basel) 2021; 13:312. [PMID: 33467027 PMCID: PMC7829765 DOI: 10.3390/cancers13020312] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor-targeting monoclonal antibodies (mAbs) are the most widely used and characterized immunotherapy for hematologic and solid tumors. The significance of this therapy is their direct and indirect effects on tumor cells, facilitated by the antibody's antigen-binding fragment (Fab) and fragment crystallizable region (Fc region), respectively. The Fab can modulate the function of cell surface markers on tumor cells in an agonistic or antagonistic manner, whereas the Fc region can be recognized by an Fc receptor (FcR) on leukocytes through which various effector functions, including antibody-dependent cell-mediated cytotoxicity (ADCC), can be elicited. This process is a key cytolytic mechanism of natural killer (NK) cells. These innate lymphocytes in the human body recognize tumor-bound antibodies exclusively by the IgG Fc receptor CD16A (FcγRIIIA). Two allelic versions of CD16A bind IgG with either lower or higher affinity. Cancer patients homozygous for the higher affinity allele of CD16A have been reported to respond significantly better to mAb therapies for various malignancies. These studies revealed that mAb therapy efficacy positively correlates with higher affinity binding to CD16A. Approaches to enhance tumor antigen targeting by NK cells by modifying the Fc portion of antibodies or the FcR on NK cells are the focus of this review.
Collapse
Affiliation(s)
| | | | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108, USA; (K.J.D.); (J.W.)
| |
Collapse
|
47
|
van Gool MMJ, van Egmond M. IgA and FcαRI: Versatile Players in Homeostasis, Infection, and Autoimmunity. Immunotargets Ther 2021; 9:351-372. [PMID: 33447585 PMCID: PMC7801909 DOI: 10.2147/itt.s266242] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Mucosal surfaces constitute the frontiers of the body and are the biggest barriers of our body for the outside world. Immunoglobulin A (IgA) is the most abundant antibody class present at these sites. It passively contributes to mucosal homeostasis via immune exclusion maintaining a tight balance between tolerating commensals and providing protection against pathogens. Once pathogens have succeeded in invading the epithelial barriers, IgA has an active role in host-pathogen defense by activating myeloid cells through divers receptors, including its Fc receptor, FcαRI (CD89). To evade elimination, several pathogens secrete proteins that interfere with either IgA neutralization or FcαRI-mediated immune responses, emphasizing the importance of IgA-FcαRI interactions in preventing infection. Depending on the IgA form, either anti- or pro-inflammatory responses can be induced. Moreover, the presence of excessive IgA immune complexes can result in continuous FcαRI-mediated activation of myeloid cells, potentially leading to severe tissue damage. On the one hand, enhancing pathogen-specific mucosal and systemic IgA by vaccination may increase protective immunity against infectious diseases. On the other hand, interfering with the IgA-FcαRI axis by monovalent targeting or blocking FcαRI may resolve IgA-induced inflammation and tissue damage. This review describes the multifaceted role of FcαRI as immune regulator between anti- and pro-inflammatory responses of IgA, and addresses potential novel therapeutic strategies that target FcαRI in disease. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/xlijXy5W0xA
Collapse
Affiliation(s)
- Melissa Maria Johanna van Gool
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
48
|
Zarrin AA, Monteiro RC. Editorial: The Role of Inhibitory Receptors in Inflammation and Cancer. Front Immunol 2020; 11:633686. [PMID: 33414792 PMCID: PMC7783443 DOI: 10.3389/fimmu.2020.633686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 01/31/2023] Open
Affiliation(s)
| | - Renato C Monteiro
- Center for Research on Inflammation, Université de Paris, Paris, France
| |
Collapse
|
49
|
Crute BW, Sheraden R, Ott VL, Harley ITW, Getahun A, Cambier JC. Inhibitory Receptor Trap: A Platform for Discovery of Inhibitory Receptors That Utilize Inositol Lipid and Phosphotyrosine Phosphatase Effectors. Front Immunol 2020; 11:592329. [PMID: 33193438 PMCID: PMC7641642 DOI: 10.3389/fimmu.2020.592329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/29/2020] [Indexed: 01/06/2023] Open
Abstract
Among the areas of most impactful recent progress in immunology is the discovery of inhibitory receptors and the subsequent translation of this knowledge to the clinic. Although the original and canonical member of this family is FcγRIIB, more recent studies defined PD1 as an inhibitory receptor that constrains T cell immunity to tumors. These studies led to development of “checkpoint blockade” immunotherapies (CBT) for cancers in which PD1 interactions with its ligand are blocked. Unfortunately, although very effective in some patients, only a small proportion respond to this therapy. This suggests that additional as yet undescribed inhibitory receptors exist, which could be exploited. Here, we describe a new platform, termed inhibitory receptor trap (IRT), for discovery of members of this family. The approach takes advantage of the fact that many of the known inhibitory receptors mediate signaling by phospho-immunoreceptor tyrosine-based inhibition motif (ITIM) mediated recruitment of Src Homology 2 (SH2) domain-containing phosphatases including the SH2 domain-containing inositol phosphatase SHIP1 encoded by the INPP5D gene and the SH2 domain-containing phosphotyrosine phosphatases SHP1 and SHP2 encoded by the PTPN6 and PTPN11 genes respectively. Here, we describe the IRT discovery platform in which the SH2 domains of inhibitory phosphatases are used for affinity-based isolation and subsequent identification of candidate effectors via immunoblotting and high sensitivity liquid chromatography–mass spectrometry. These receptors may represent alternative targets that can be exploited for improved CBT. Salient observations from these studies include the following: SH2 domains derived from the respective phosphatases bind distinct sets of candidates from different cell types. Thus, cells of different identity and different activation states express partially distinct repertoires of up and downstream phosphatase effectors. Phosphorylated PD1 binds not only SHP2 but also SHIP1, thus the latter may be important in its inhibitory function. B cell antigen receptor signaling leads predominantly to CD79 mono-phosphorylation as indicated by much greater binding to LynSH2 than Syk(SH2)2. This balance of ITAM mono- versus bi-phosphorylation likely tunes signaling by varying activation of inhibitory (Lyn) and stimulatory (Syk) pathways.
Collapse
Affiliation(s)
- Bergren W Crute
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Rachel Sheraden
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Vanessa L Ott
- Department of Biomedical Sciences, National Jewish Health, Denver, CO, United States
| | - Isaac T W Harley
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Biomedical Sciences, National Jewish Health, Denver, CO, United States
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Biomedical Sciences, National Jewish Health, Denver, CO, United States
| |
Collapse
|
50
|
Zhang L, Xia Y, Li W, Sun Y, Kong L, Xu P, Xia P, Yue J. Activation of Fc gamma receptor IIb up-regulates the production of interferon-alpha and interferon-gamma in porcine alveolar macrophages during PRRSV infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 109:103696. [PMID: 32278861 DOI: 10.1016/j.dci.2020.103696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 06/11/2023]
Abstract
Porcine Fc gamma receptor IIb (FcγRIIb) has been cloned and characterized for many years. However, the role of FcγRIIb in innate antiviral response to porcine reproductive and respiratory syndrome virus (PRRSV) infection has not yet been well investigated. In current study, our results showed that specific activation of FcγRIIb in porcine alveolar macrophages (PAMs) significantly enhanced the production of interferon-alpha (IFN-α) and interferon-gamma (IFN-γ), and significantly repressed the production of transforming growth factor beta 1 (TGF-β1). In addition, our results showed that specific activation of FcγRIIb in PAMs cells in PRRSV infection not only significantly increased the production of IFN-α and IFN-γ, but also significantly decreased the production of TGF-β1, and significantly inhibited PRRSV replication level. In summary, our studies indicated that FcγRIIb signaling up-regulated the production of IFN-α and IFN-γ in PAMs cells in vitro, in response to PRRSV infection.
Collapse
Affiliation(s)
- Liujun Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yuhao Xia
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wen Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yangyang Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Linghao Kong
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pengli Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pingan Xia
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|