1
|
Kubagawa H, Mahmoudi Aliabadi P, Al-Qaisi K, Jani PK, Honjo K, Izui S, Radbruch A, Melchers F. Functions of IgM fc receptor (FcµR) related to autoimmunity. Autoimmunity 2024; 57:2323563. [PMID: 38465789 DOI: 10.1080/08916934.2024.2323563] [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: 10/29/2023] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
Unlike Fc receptors for switched immunoglobulin (Ig) isotypes, Fc receptor for IgM (FcµR) is selectively expressed by lymphocytes. The ablation of the FcµR gene in mice impairs B cell tolerance as evidenced by concomitant production of autoantibodies of IgM and IgG isotypes. In this essay, we reiterate the autoimmune phenotypes observed in mutant mice, ie IgM homeostasis, dysregulated humoral immune responses including autoantibodies, and Mott cell formation. We also propose the potential phenotypes in individuals with FCMR deficiency and the model for FcµR-mediated regulation of self-reactive B cells.
Collapse
Affiliation(s)
| | | | | | - Peter K Jani
- Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Kazuhito Honjo
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shozo Izui
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| |
Collapse
|
2
|
Xiang Y, Xu J, McGovern BL, Ranzenigo A, Huang W, Sang Z, Shen J, Diaz-Tapia R, Pham ND, Teunissen AJP, Rodriguez ML, Benjamin J, Taylor DJ, van Leent MMT, White KM, García-Sastre A, Zhang P, Shi Y. Adaptive multi-epitope targeting and avidity-enhanced nanobody platform for ultrapotent, durable antiviral therapy. Cell 2024; 187:6966-6980.e23. [PMID: 39447570 DOI: 10.1016/j.cell.2024.09.043] [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: 02/26/2024] [Revised: 07/30/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Pathogens constantly evolve and can develop mutations that evade host immunity and treatment. Addressing these escape mechanisms requires targeting evolutionarily conserved vulnerabilities, as mutations in these regions often impose fitness costs. We introduce adaptive multi-epitope targeting with enhanced avidity (AMETA), a modular and multivalent nanobody platform that conjugates potent bispecific nanobodies to a human immunoglobulin M (IgM) scaffold. AMETA can display 20+ nanobodies, enabling superior avidity binding to multiple conserved and neutralizing epitopes. By leveraging multi-epitope SARS-CoV-2 nanobodies and structure-guided design, AMETA constructs exponentially enhance antiviral potency, surpassing monomeric nanobodies by over a million-fold. These constructs demonstrate ultrapotent, broad, and durable efficacy against pathogenic sarbecoviruses, including Omicron sublineages, with robust preclinical results. Structural analysis through cryoelectron microscopy and modeling has uncovered multiple antiviral mechanisms within a single construct. At picomolar to nanomolar concentrations, AMETA efficiently induces inter-spike and inter-virus cross-linking, promoting spike post-fusion and striking viral disarmament. AMETA's modularity enables rapid, cost-effective production and adaptation to evolving pathogens.
Collapse
Affiliation(s)
- Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialu Xu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Briana L McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Ranzenigo
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan Shen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ngoc Dung Pham
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Luis Rodriguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jared Benjamin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek J Taylor
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Mandy M T van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Peijun Zhang
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK; Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK.
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
3
|
Peng C, Talreja J, Steinbauer B, Shinki K, Koth LL, Samavati L. Discovery of Two Novel Immunoepitopes and Development of a Peptide-based Sarcoidosis Immunoassay. Am J Respir Crit Care Med 2024; 210:908-918. [PMID: 38385694 PMCID: PMC11506913 DOI: 10.1164/rccm.202306-1054oc] [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: 06/19/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
Rationale: Sarcoidosis is a systemic granulomatous disorder associated with hypergammaglobulinemia and the presence of autoantibodies. The specific antigens initiating granulomatous inflammation in sarcoidosis are unknown, and there is no specific test available to diagnose sarcoidosis. To discover novel sarcoidosis antigens, we developed a high-throughput T7 phage display library derived from the sarcoidosis cDNA and identified numerous clones differentiating sarcoidosis from other respiratory diseases. After clone sequencing and a homology search, we identified two epitopes (cofilin μ and chain A) that specifically bind to serum IgGs of patients with sarcoidosis. Objectives: To develop and validate an epitope-specific IgG-based immunoassay specific for sarcoidosis. Methods: We chemically synthesized both immunoepitopes (cofilin μ and chain A) and generated rabbit polyclonal antibodies against both neoantigens. After extensive standardization, we developed a direct peptide ELISA and measured epitope-specific IgG in the sera of 386 subjects, including healthy control subjects (n = 100), three sarcoidosis cohorts (n = 186), pulmonary tuberculosis (n = 70), and lung cancer (n = 30). Measurements and Main Results: To develop a model to classify sarcoidosis distinctly from other groups, data were analyzed using fivefold cross-validation when adjusting for confounders. The cofilin μ IgG model yielded a mean sensitivity, specificity, and positive and negative predictive value of 0.97, 0.9, 0.9, and 0.96, respectively. Those same measures for chain A IgG antibody were 0.9, 0.83, 0.84, and 0.9, respectively. Combining both biomarkers improved the area under the curve, sensitivity, specificity, and positive and negative predictive value. Conclusions: These results provide a novel immunoassay for sarcoidosis. The discovery of two neoantigens facilitates the development of biospecific drug discovery and the sarcoidosis-specific model.
Collapse
Affiliation(s)
- Changya Peng
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan
| | - Jaya Talreja
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan
| | - Brennen Steinbauer
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan
| | - Kazuhiko Shinki
- Department of Mathematics, Wayne State University, Detroit, Michigan
| | - Laura L. Koth
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California; and
| | - Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
4
|
Murata K, Murao A, Tan C, Wang P, Aziz M. B-1a cells scavenge NETs to attenuate sepsis. J Leukoc Biol 2024; 116:632-643. [PMID: 38484156 PMCID: PMC11367732 DOI: 10.1093/jleuko/qiae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 09/03/2024] Open
Abstract
B-1a cells, a regulatory subset of B lymphocytes, produce natural IgM and interleukin-10. Neutrophil extracellular traps (NETs) play a crucial role in pathogen defense, but their excessive formation during sepsis can cause further inflammation and tissue damage. In sepsis, extracellular cold-inducible RNA-binding protein (eCIRP), a damage-associated molecular pattern, is released to induce NET formation. We hypothesize that B-1a cells clear NETs to prevent sepsis-induced injury. Sepsis in mice was induced by injecting 1 × 107 and 5 × 107 colony-forming units of Escherichia coli intraperitoneally. After 4 and 20 h, we assessed the number of B-1a cells in the peritoneal cavity using flow cytometry. Our results showed that the number of peritoneal B-1a cells was significantly decreased in E. coli sepsis mice. Importantly, replenishing B-1a cells via intraperitoneal injection in sepsis mice significantly decreased NETs in peritoneal neutrophils. We also observed a decrease in serum inflammation and injury markers and a significant increase in the overall survival rate in B-1a cell-treated septic mice. To understand the mechanism, we cocultured bone marrow-derived neutrophils with peritoneal B-1a cells in a contact or noncontact condition using an insert and stimulated them with eCIRP. After 4 h, we found that eCIRP significantly increased NET formation in bone marrow-derived neutrophils. Interestingly, we observed that B-1a cells inhibited NETs by 67% in a contact-dependent manner. Surprisingly, when B-1a cells were cultured in inserts, there was no significant decrease in NET formation, suggesting that direct cell-to-cell contact is crucial for this inhibitory effect. We further determined that B-1a cells promoted NET phagocytosis, and this was mediated through natural IgM, as blocking the IgM receptor attenuated the engulfment of NETs by B-1a cells. Finally, we identified that following their engulfment, NETs were localized into the lysosomal compartment for lysis. Thus, our study suggests that B-1a cells decrease NET content in eCIRP-treated neutrophils and E. coli sepsis mice.
Collapse
Affiliation(s)
- Kensuke Murata
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Chuyi Tan
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| |
Collapse
|
5
|
Stosik M, Tokarz-Deptuła B, Deptuła W. Polymeric immunoglobulin receptor (pIgR) in ray-finned fish (Actinopterygii). FISH & SHELLFISH IMMUNOLOGY 2023; 138:108814. [PMID: 37211331 DOI: 10.1016/j.fsi.2023.108814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Affiliation(s)
- Michał Stosik
- Institute of Biological Sciences, Faculty of Biological Sciences University of Zielona Góra, Poland
| | | | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
6
|
Bartsch YC, Cizmeci D, Kang J, Gao H, Shi W, Chandrashekar A, Collier ARY, Chen B, Barouch DH, Alter G. Selective SARS-CoV2 BA.2 escape of antibody Fc/Fc-receptor interactions. iScience 2023; 26:106582. [PMID: 37082529 PMCID: PMC10079316 DOI: 10.1016/j.isci.2023.106582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/02/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
The number of mutations in the omicron (B.1.1.529) BA.1 variant of concern led to an unprecedented evasion of vaccine induced immunity. However, despite rise in global infections, severe disease did not increase proportionally and is likely linked to persistent recognition of BA.1 by T cells and non-neutralizing opsonophagocytic antibodies. Yet, the emergence of new sublineage BA.2, which is more transmissible than BA.1 despite relatively preserved neutralizing antibody responses, has raised the possibility that BA.2 may evade other vaccine-induced responses. Here, we comprehensively profiled the BNT162b2 vaccine-induced response to several VOCs, including omicron BA.1 and BA.2. While vaccine-induced immune responses were compromised against both omicron sublineages, vaccine-induced antibody isotype titers, and non-neutralizing Fc effector functions were attenuated to the omicron BA.2 spike compared to BA.1. Conversely, FcγR2a and FcγR2b binding was elevated to BA.2, albeit lower than BA.1 responses, potentially contributing to persistent protection against severity of disease.
Collapse
Affiliation(s)
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Hailong Gao
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Shi
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Bing Chen
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dan H. Barouch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
7
|
Kubagawa H, Clark C, Skopnik CM, Mahmoudi Aliabadi P, Al-Qaisi K, Teuber R, Jani PK, Radbruch A, Melchers F, Engels N, Wienands J. Physiological and Pathophysiological Roles of IgM Fc Receptor (FcµR) Isoforms. Int J Mol Sci 2023; 24:ijms24065728. [PMID: 36982860 PMCID: PMC10058298 DOI: 10.3390/ijms24065728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
IgM is the first antibody to emerge during phylogeny, ontogeny, and immune responses and serves as a first line of defense. Effector proteins interacting with the Fc portion of IgM, such as complement and its receptors, have been extensively studied for their functions. IgM Fc receptor (FcµR), identified in 2009, is the newest member of the FcR family and is intriguingly expressed by lymphocytes only, suggesting the existence of distinct functions as compared to the FcRs for switched Ig isotypes, which are expressed by various immune and non-hematopoietic cells as central mediators of antibody-triggered responses by coupling the adaptive and innate immune responses. Results from FcµR-deficient mice suggest a regulatory function of FcµR in B cell tolerance, as evidenced by their propensity to produce autoantibodies of both IgM and IgG isotypes. In this article, we discuss conflicting views about the cellular distribution and potential functions of FcµR. The signaling function of the Ig-tail tyrosine-like motif in the FcµR cytoplasmic domain is now formally shown by substitutional experiments with the IgG2 B cell receptor. The potential adaptor protein associating with FcµR and the potential cleavage of its C-terminal cytoplasmic tail after IgM binding are still enigmatic. Critical amino acid residues in the Ig-like domain of FcµR for interacting with the IgM Cµ4 domain and the mode of interaction are now defined by crystallographic and cryo-electron microscopic analyses. Some discrepancies on these interactions are discussed. Finally, elevated levels of a soluble FcµR isoform in serum samples are described as the consequence of persistent B cell receptor stimulation, as seen in chronic lymphocytic leukemia and probably in antibody-mediated autoimmune disorders.
Collapse
Affiliation(s)
| | - Caren Clark
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | | | | | | | - Ruth Teuber
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Peter K Jani
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | | | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| |
Collapse
|
8
|
TLR7 and IgM: Dangerous Partners in Autoimmunity. Antibodies (Basel) 2023; 12:antib12010004. [PMID: 36648888 PMCID: PMC9844493 DOI: 10.3390/antib12010004] [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: 10/09/2022] [Revised: 10/27/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
The B cell antigen receptor (BCR)-repertoire is capable of recognizing a nearly unlimited number of antigens. Inevitably, the random nature of antibody gene segment rearrangement, needed in order to provide mature B cells, will generate autoreactive specificities. Once tolerance mechanisms fail to block the activation and differentiation of autoreactive B cells, harmful autoantibodies may get secreted establishing autoimmune diseases. Besides the hallmark of autoimmunity, namely IgG autoantibodies, IgM autoantibodies are also found in many autoimmune diseases. In addition to pathogenic functions of secreted IgM the IgM-BCR expressing B cell might be the initial check-point where, in conjunction with innate receptor signals, B cell mediated autoimmunity starts it fateful course. Recently, pentameric IgM autoantibodies have been shown to contribute significantly to the pathogenesis of various autoimmune diseases, such as rheumatoid arthritis (RA), autoimmune hemolytic anemia (AIHA), pemphigus or autoimmune neuropathy. Further, recent studies suggest differences in the recognition of autoantigen by IgG and IgM autoantibodies, or propose a central role of anti-ACE2-IgM autoantibodies in severe COVID-19. However, exact mechanisms still remain to be uncovered in detail. This article focuses on summarizing recent findings regarding the importance of autoreactive IgM in establishing autoimmune diseases.
Collapse
|
9
|
Zhang L, Shi Y, Han X. Immunogenomic correlates of immune-related adverse events for anti-programmed cell death 1 therapy. Front Immunol 2022; 13:1032221. [PMID: 36505471 PMCID: PMC9733471 DOI: 10.3389/fimmu.2022.1032221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
Despite impressive antitumor efficacy of programmed cell death 1 (PD-1) inhibitors, this inhibition can induce mild to severe autoimmune toxicities, termed immune-related adverse events (irAEs). Yet, predictive pretreatment biomarkers for irAEs development across cancer types remain elusive. We first assessed cellular and molecular factors. To determine factors predicting the risk of irAEs for anti-PD-1 immunotherapy across multiple cancer types, an integrative analysis of cellular and molecular factors from 9104 patients across 21 cancer types and 4865522 postmarketing adverse event reports retrieved from adverse event reporting system was then performed. Accuracy of predictions was quantified as Pearson correlation coefficient determined using leave-one-out cross-validation. Independent validation sets included small cell lung cancer and melanoma cohorts. Out of 4865522 eligible adverse events reports, 10412 cases received anti-PD-1 monotherapy, of which, 2997 (28.78%) exhibited at least one irAE. Among established immunogenomic factors, dendritic cells (DC) abundance showed the strongest correlation with irAEs risk, followed by tumor mutational burden (TMB). Further predictive accuracy was achieved by DC and TMB in combination with CD4+ naive T-cells abundance, and then validated in the small cell lung cancer cohort. Additionally, global screening of multiomics data identified 11 novel predictors of irAEs. Of these, IRF4 showed the highest correlation. Best predictive performance was observed in the IRF4 - TCL1A - SHC-pY317 trivariate model. Associations of IRF4 and TCL1A expression with irAEs development were verified in the melanoma cohort receiving immune checkpoint inhibitors. Collectively, pretreatment cellular and molecular irAEs-associated features as well as their combinations are identified regardless of cancer types. These findings may deepen our knowledge of irAEs pathogenesis and, ultimately, aid in early detection of high-risk patients and management of irAEs.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China,Medical Research Center, Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China,*Correspondence: Yuankai Shi, ; Xiaohong Han,
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China,*Correspondence: Yuankai Shi, ; Xiaohong Han,
| |
Collapse
|
10
|
Naranjo L, Stojanovich L, Djokovic A, Andreoli L, Tincani A, Maślińska M, Sciascia S, Infantino M, Garcinuño S, Kostyra-Grabczak K, Manfredi M, Regola F, Stanisavljevic N, Milanovic M, Saponjski J, Roccatello D, Cecchi I, Radin M, Benucci M, Pleguezuelo D, Serrano M, Shoenfeld Y, Serrano A. Circulating immune-complexes of IgG/IgM bound to B2-glycoprotein-I associated with complement consumption and thrombocytopenia in antiphospholipid syndrome. Front Immunol 2022; 13:957201. [PMID: 36172349 PMCID: PMC9511106 DOI: 10.3389/fimmu.2022.957201] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022] Open
Abstract
Background Antiphospholipid syndrome (APS) is a multisystemic autoimmune disorder characterized by thrombotic events and/or gestational morbidity in patients with antiphospholipid antibodies (aPL). In a previous single center study, APS-related clinical manifestations that were not included in the classification criteria (livedo reticularis, thrombocytopenia, leukopenia) were associated with the presence of circulating immune-complexes (CIC) formed by beta-2-glycoprotein-I (B2GP1) and anti-B2GP1 antibodies (B2-CIC). We have performed a multicenter study on APS features associated with the presence of B2-CIC. Methods A multicenter, cross-sectional and observational study was conducted on 303 patients recruited from six European hospitals who fulfilled APS classification criteria: 165 patients had primary APS and 138 APS associated with other systemic autoimmune diseases (mainly systemic lupus erythematosus, N=112). Prevalence of B2-CIC (IgG/IgM isotypes) and its association with clinical manifestations and biomarkers related to the disease activity were evaluated. Results B2-CIC prevalence in APS patients was 39.3%. B2-CIC-positive patients with thrombotic APS presented a higher incidence of thrombocytopenia (OR: 2.32, p=0.007), heart valve thickening and dysfunction (OR: 9.06, p=0.015) and triple aPL positivity (OR: 1.83, p=0.027), as well as lower levels of C3, C4 and platelets (p-values: <0.001, <0.001 and 0.001) compared to B2-CIC-negative patients. B2-CIC of IgM isotype were significantly more prevalent in gestational than thrombotic APS. Conclusions Patients with thrombotic events and positive for B2-CIC had lower platelet count and complement levels than those who were negative, suggesting a greater degree of platelet activation.
Collapse
Affiliation(s)
- Laura Naranjo
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Aleksandra Djokovic
- Cardiology Department, University Hospital Center Bezanijska Kosa, Belgrade, Serbia
- School of Medicine , University of Belgrade, Belgrade, Serbia
| | - Laura Andreoli
- Unit of Rheumatology and Clinical Immunology, ASST Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Angela Tincani
- Unit of Rheumatology and Clinical Immunology, ASST Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Maria Maślińska
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Savino Sciascia
- Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital, Torino, Italy
| | - Maria Infantino
- Immunology and Allergy Laboratory, San Giovanni di Dio Hospital, Florence, Italy
| | - Sara Garcinuño
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Kinga Kostyra-Grabczak
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Mariangela Manfredi
- Immunology and Allergy Laboratory, San Giovanni di Dio Hospital, Florence, Italy
| | - Francesca Regola
- Unit of Rheumatology and Clinical Immunology, ASST Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Natasa Stanisavljevic
- Internal Medicine, University Hospital Center Bezanijska Kosa, Belgrade, Serbia
- School of Medicine , University of Belgrade, Belgrade, Serbia
| | - Milomir Milanovic
- Internal Medicine Department, Clinic for Infectious and Tropical Diseases, Military Medical Academy, Belgrade, Serbia
| | - Jovica Saponjski
- Cardiology Department, University Clinical Center of Serbia, Belgrade, Serbia
| | - Dario Roccatello
- Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital, Torino, Italy
| | - Irene Cecchi
- Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital, Torino, Italy
| | - Massimo Radin
- Nephrology and Dialysis Unit (ERK-net Member), Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital, Torino, Italy
| | - Maurizio Benucci
- Rheumatology Unit, San Giovanni di Dio Hospital, Florence, Italy
| | - Daniel Pleguezuelo
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Manuel Serrano
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- *Correspondence: Manuel Serrano,
| | - Yehuda Shoenfeld
- Ariel University, Ariel, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Antonio Serrano
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
11
|
Kubli SP, Ramachandran P, Duncan G, Brokx R, Mak TW. Reply to: Questioning whether the IgM Fc receptor (FcμR) is expressed by innate immune cells. Nat Commun 2022; 13:3950. [PMID: 35817786 PMCID: PMC9273603 DOI: 10.1038/s41467-022-31226-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shawn P Kubli
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | | | - Gordon Duncan
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | - Rich Brokx
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2M9, Canada.
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada.
- Department of Immunology, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada.
- Department of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
12
|
Altvater-Hughes TE, Hodgins DC, Wagter-Lesperance L, Beard SC, Cartwright SL, Mallard BA. Concentration and heritability of immunoglobulin G and natural antibody immunoglobulin M in dairy and beef colostrum along with serum total protein in their calves. J Anim Sci 2022; 100:skac006. [PMID: 35022742 PMCID: PMC8867588 DOI: 10.1093/jas/skac006] [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: 07/16/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Immunoglobulin (Ig) G and natural antibody (NAb) IgM are passively transferred to the neonatal calf through bovine colostrum. Maternal IgG provides pathogen- or vaccine-specific protection and comprises about 85% of colostral Ig. NAb-IgM is less abundant but provides broad and nonspecific reactivity, potentially contributing to protection against the dissemination of pathogens in the blood (septicemia) in a calf's first days of life. In the dairy and beef industries, failure of passive transfer (FPT) of colostral Ig (serum total protein [STP] <5.2 g/dL) is still a common concern. The objectives of this study were to: (1) compare colostral IgG concentrations and NAb-IgM titers between dairy and beef cows; (2) assess the effect of beef breed on colostral IgG; (3) compare passive transfer of colostral Ig in dairy and beef calves; and (4) estimate the heritability of colostral IgG and NAb-IgM. Colostrum was collected from Holstein dairy (n = 282) and crossbred beef (n = 168) cows at the University of Guelph dairy and beef research centers. Colostral IgG was quantified by radial immunodiffusion and NAb-IgM was quantified by an enzyme-linked immunosorbent assay. In dairy (n = 308) and beef (n = 169) calves, STP was estimated by digital refractometry. Beef cows had significantly greater colostral IgG (146.5 ± 9.5 standard error of the mean [SEM] g/L) than dairy cows (92.4 ± 5.2 g/L, P <0.01). Beef cows with a higher proportion of Angus ancestry had significantly lower colostral IgG (125.5 ± 5.8 g/L) than cows grouped as "Other" (142.5 ± 4.9 g/L, P = 0.02). Using the FPT cutoff, 13% of dairy and 16% of beef calves had FPT; still, beef calves had a significantly larger proportion with excellent passive transfer (STP ≥6.2 g/dL, P <0.01). The heritability of colostral IgG was 0.04 (±0.14) in dairy and 0.14 (±0.32) in beef. Colostral NAb-IgM titers in dairy (12.12 ± 0.22, log2 [reciprocal of titer]) and beef cows (12.03 ± 0.19) did not differ significantly (P = 0.71). The range of NAb-IgM titers was 9.18-14.60, equivalent to a 42-fold range in antibody concentration. The heritability of colostral NAb was 0.24 (±0.16) in dairy and 0.11 (±0.19) in beef cows. This study is the first to compare colostral NAb-IgM between dairy and beef cows. Based on the range in NAb-IgM titers and the heritability, selective breeding may improve colostrum quality and protection for neonatal calves in the early days of life.
Collapse
Affiliation(s)
- Tess E Altvater-Hughes
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Douglas C Hodgins
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauraine Wagter-Lesperance
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shannon C Beard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shannon L Cartwright
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Bonnie A Mallard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
13
|
Amendt T, Jumaa H. Adaptive tolerance: Protection through self-recognition. Bioessays 2022; 44:e2100236. [PMID: 34984705 DOI: 10.1002/bies.202100236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 01/10/2023]
Abstract
The random nature of immunoglobulin gene segment rearrangement inevitably leads to the generation of self-reactive B cells. Avoidance of destructive autoimmune reactions is necessary in order to maintain physiological homeostasis. However, current central and peripheral tolerance concepts fail to explain the massive number of autoantibody-borne autoimmune diseases. Moreover, recent studies have shown that in physiological mouse models autoreactive B cells were neither clonally deleted nor kept in an anergic state, but were instead able to mount autoantibody responses. We propose that activation of autoreactive B cells is induced by polyvalent autoantigen complexes that can occur under physiological conditions. Repeated encounter of autoantigen complexes leads to the production of affinity-matured autoreactive IgM that protects its respective self-targets from degradation. We refer to this novel mechanism as adaptive tolerance. This article discusses the discovery of adaptive tolerance and the unexpected role of high affinity IgM autoantibodies.
Collapse
Affiliation(s)
- Timm Amendt
- Institute of Immunology, University Hospital Ulm, Ulm, Germany
| | - Hassan Jumaa
- Institute of Immunology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
14
|
Ozcelik F, Tanoglu A, Guven BB, Keskin U, Kaplan M. Assessment of severity and mortality of COVID-19 with anti-A1 and anti-B IgM isohaemagglutinins, a reflection of the innate immune status. Int J Clin Pract 2021; 75:e14624. [PMID: 34251730 PMCID: PMC8420313 DOI: 10.1111/ijcp.14624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS The relationship between the innate immune system that creates the polysaccharide antibody response and COVID-19 is not fully understood. In this study, it was aimed to determine the predictive values of isohaemagglutinins in COVID-19 severity/mortality. METHODS Approximately 15 440 patients diagnosed with COVID-19 were examined, and a total of 286 patients with anti-B and anti-A1 IgM isohaemagglutinins test results were randomly enrolled in the study. These patients were stratified into two groups according to anti-A1 (n: 138 blood type B or O) and anti-B (n: 148 blood type A) IgM isohaemagglutinins. Anti-A1 or/and anti-B IgM, biochemical parameters, symptoms, chronic diseases, hospitalisation status, intubation status, admission to intensive care unit (ICU) and exitus status were recorded and evaluated for all patients. RESULTS Anti-A1 IgM and anti-B IgM were significantly lower in ICU patients (7.5 ± 9.9 vs 18.0 ± 20.4 and 5.5 ± 6.3 vs 19.3 ± 33.6 titres, respectively; P < .01) and in exitus patients (3.8 ± 3.6 vs 16.7 ± 18.7 and 3.5 ± 4.7 vs 16.9 ± 29.6 titres respectively; P < .01). In the ROC analysis performed to differentiate between exitus and discharge within groups, the sensitivity of anti-B IgM and anti-A1 IgM at cut-off ≤4 was 88.9% and 79.6%, specificity 66.0% and 73.4%, and AUC 0.831 and 0.861, respectively (P < .01). Anti-A1 IgM decreased the mortality risk 0.811 times per unit while anti-B IgM decreased 0.717 times (P < .01). CONCLUSION Anti-B and anti-A1 isohaemagglutinins, which are an expression of the innate immune system, can be used to predict the severity and mortality of COVID-19 disease.
Collapse
Affiliation(s)
- Fatih Ozcelik
- Department of Medical BiochemistryUniversity of Health Sciences TurkeySultan 2. Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Alpaslan Tanoglu
- Department of GastroenterologyUniversity of Health Sciences TurkeySancaktepe Şehit Prof Dr Ilhan Varank Training and Research HospitalIstanbulTurkey
| | - Bulent Barıs Guven
- Department of Anesthesia and ReanimationUniversity of Health Sciences TurkeySultan 2. Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Umran Keskin
- Department of Internal MedicineUniversity of Health Sciences TurkeyHaydarpasa Numune Training and Research HospitalIstanbulTurkey
| | - Mustafa Kaplan
- Department of Internal MedicineUniversity of Health Sciences TurkeySultan 2. Abdulhamid Han Training and Research HospitalIstanbulTurkey
| |
Collapse
|
15
|
Flowers EM, Neely HR, Guo J, Almeida T, Ohta Y, Castro CD, Flajnik MF. Identification of the Fc-alpha/mu receptor in Xenopus provides insight into the emergence of the poly-Ig receptor (pIgR) and mucosal Ig transport. Eur J Immunol 2021; 51:2590-2606. [PMID: 34411303 DOI: 10.1002/eji.202149383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Indexed: 01/17/2023]
Abstract
The polyimmunoglobulin receptor (pIgR) transcytoses J chain-containing antibodies through mucosal epithelia. In mammals, two cis-duplicates of PIGR, FCMR, and FCAMR, flank the PIGR gene. A PIGR duplication is first found in amphibians, previously annotated as PIGR2 (herein xlFCAMR), and is expressed by APCs. We demonstrate that xlFcamR is the equivalent of mammalian FcamR. It has been assumed that pIgR is the oldest member of this family, yet our data could not distinguish whether PIGR or FCAMR emerged first; however, FCMR was the last family member to emerge. Interestingly, bony fish "pIgR" is not an orthologue of tetrapod pIgR, and possibly acquired its function via convergent evolution. PIGR/FCAMR/FCMR are members of a larger superfamily, including TREM, CD300, and NKp44, which we name the "double-disulfide Ig superfamily" (ddIgSF). Domains related to each ddIgSF family were identified in cartilaginous fish (sharks, chimeras) and encoded in a single gene cluster syntenic to the human pIgR locus. Thus, the ddIgSF families date back to the earliest antibody-based adaptive immunity, but apparently not before. Finally, our data strongly suggest that the J chain arose in evolution only for Ig multimerization. This study provides a framework for further studies of pIgR and the ddIgSF in vertebrates.
Collapse
Affiliation(s)
- Emily M Flowers
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Harold R Neely
- Department of Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Jacqueline Guo
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tereza Almeida
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caitlin D Castro
- Committee on Immunology and Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Amendt T, Ayoubi OE, Linder AT, Allies G, Young M, Setz CS, Jumaa H. Primary Immune Responses and Affinity Maturation Are Controlled by IgD. Front Immunol 2021; 12:709240. [PMID: 34434193 PMCID: PMC8381280 DOI: 10.3389/fimmu.2021.709240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/15/2021] [Indexed: 01/17/2023] Open
Abstract
Mature B cells co-express IgM and IgD B cell antigen receptors (BCR) on their surface. While IgM BCR expression is already essential at early stages of development, the role of the IgD-class BCR remains unclear as most B cell functions appeared unchanged in IgD-deficient mice. Here, we show that IgD-deficient mice have an accelerated rate of B cell responsiveness as they activate antibody production within 24h after immunization, whereas wildtype (WT) animals required 3 days to activate primary antibody responses. Strikingly, soluble monovalent antigen suppresses IgG antibody production induced by multivalent antigen in WT mice. In contrast, IgD-deficient mice were not able to modulate IgG responses suggesting that IgD controls the activation rate of B cells and subsequent antibody production by sensing and distinguishing antigen-valences. Using an insulin-derived peptide we tested the role of IgD in autoimmunity. We show that primary autoreactive antibody responses are generated in WT and in IgD-deficient mice. However, insulin-specific autoantibodies were detected earlier and caused more severe symptoms of autoimmune diabetes in IgD-deficient mice as compared to WT mice. The rapid control of autoimmune diabetes in WT animals was associated with the generation of high-affinity IgM that protects insulin from autoimmune degradation. In IgD-deficient mice, however, the generation of high-affinity protective IgM is delayed resulting in prolonged autoimmune diabetes. Our data suggest that IgD is required for the transition from primary, highly autoreactive, to secondary antigen-specific antibody responses generated by affinity maturation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hassan Jumaa
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
17
|
De M, Bhushan A, Chinnaswamy S. Monocytes differentiated into macrophages and dendritic cells in the presence of human IFN-λ3 or IFN-λ4 show distinct phenotypes. J Leukoc Biol 2021; 110:357-374. [PMID: 33205487 PMCID: PMC7611425 DOI: 10.1002/jlb.3a0120-001rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022] Open
Abstract
Human IFN-λ4 is expressed by only a subset of individuals who possess the ΔG variant allele at the dinucleotide polymorphism rs368234815. Recent genetic studies have shown an association between rs368234815 and different infectious and inflammatory disorders. It is not known if IFN-λ4 has immunomodulatory activity. The expression of another type III IFN, IFN-λ3, is also controlled by genetic polymorphisms that are strongly linked to rs368234815. Therefore, it is of interest to compare these two IFNs for their effects on immune cells. Herein, using THP-1 cells, it was confirmed that IFN-λ4 could affect the differentiation status of macrophage-like cells and dendritic cells (DCs). The global gene expression changes induced by IFN-λ4 were also characterized in in vitro generated primary macrophages. Next, human PBMC-derived CD14+ monocytes were used to obtain M1 and M2 macrophages and DCs in the presence of IFN-λ3 or IFN-λ4. These DCs were cocultured with CD4+ Th cells derived from allogenic donors and their in vitro cytokine responses were measured. The specific activity of recombinant IFN-λ4 was much lower than that of IFN-λ3, as shown by induction of IFN-stimulated genes. M1 macrophages differentiated in the presence of IFN-λ4 showed higher IL-10 secretion than those differentiated in IFN-λ3. Coculture experiments suggested that IFN-λ4 could confer a Th2-biased phenotype to allogenic Th cells, wherein IFN-λ3, under similar circumstances, did not induce a significant bias toward either a Th1 or Th2 phenotype. This study shows for the first time that IFN-λ4 may influence immune responses by immunomodulation.
Collapse
Affiliation(s)
- Manjarika De
- National Institute of Biomedical GenomicsKalyaniWest BengalIndia
| | - Anand Bhushan
- National Institute of Biomedical GenomicsKalyaniWest BengalIndia
| | | |
Collapse
|
18
|
Kubagawa H, Skopnik CM, Al-Qaisi K, Calvert RA, Honjo K, Kubagawa Y, Teuber R, Aliabadi PM, Enghard P, Radbruch A, Sutton BJ. Differences between Human and Mouse IgM Fc Receptor (FcµR). Int J Mol Sci 2021; 22:ijms22137024. [PMID: 34209905 PMCID: PMC8267714 DOI: 10.3390/ijms22137024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023] Open
Abstract
Both non-immune "natural" and antigen-induced "immune" IgM are important for protection against pathogens and for regulation of immune responses to self-antigens. Since the bona fide IgM Fc receptor (FcµR) was identified in humans by a functional cloning strategy in 2009, the roles of FcµR in these IgM effector functions have begun to be explored. In this short essay, we describe the differences between human and mouse FcµRs in terms of their identification processes, cellular distributions and ligand binding activities with emphasis on our recent findings from the mutational analysis of human FcµR. We have identified at least three sites of human FcµR, i.e., Asn66 in the CDR2, Lys79 to Arg83 in the DE loop and Asn109 in the CDR3, responsible for its constitutive IgM-ligand binding. Results of computational structural modeling analysis are consistent with these mutational data and a model of the ligand binding, Ig-like domain of human FcµR is proposed. Serendipitously, substitution of Glu41 and Met42 in the CDR1 of human FcµR with mouse equivalents Gln and Leu, either single or more prominently in combination, enhances both the receptor expression and IgM binding. These findings would help in the future development of preventive and therapeutic interventions targeting FcµR.
Collapse
Affiliation(s)
- Hiromi Kubagawa
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
- Correspondence: ; Tel.: +49-030-2846-0782
| | - Christopher M. Skopnik
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Khlowd Al-Qaisi
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Rosaleen A. Calvert
- Randall Centre for Cell and Molecular Biophysics, King’s College, London SE1 1UL, UK; (R.A.C.); (B.J.S.)
| | - Kazuhito Honjo
- Department of Pathology of University of Alabama at Birmingham, Birmingham, AL 35294, USA.; (K.H.); (Y.K.)
| | - Yoshiki Kubagawa
- Department of Pathology of University of Alabama at Birmingham, Birmingham, AL 35294, USA.; (K.H.); (Y.K.)
| | - Ruth Teuber
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Pedram Mahmoudi Aliabadi
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité-Universitätmedizin, 10117 Berlin, Germany;
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Brian J. Sutton
- Randall Centre for Cell and Molecular Biophysics, King’s College, London SE1 1UL, UK; (R.A.C.); (B.J.S.)
| |
Collapse
|
19
|
Arend P. Why blood group A individuals are at risk whereas blood group O individuals are protected from SARS-CoV-2 (COVID-19) infection: A hypothesis regarding how the virus invades the human body via ABO(H) blood group-determining carbohydrates. Immunobiology 2021; 226:152027. [PMID: 33706067 PMCID: PMC7609233 DOI: 10.1016/j.imbio.2020.152027] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/16/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022]
Abstract
While the angiotensin converting enzyme 2 (ACE2) protein is defined as the primary severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor, the viral serine molecule might be mobilized by the host's transmembrane protease serine subtype 2 (TMPRSS2) enzyme from the viral spike (S) protein and hijack the host's N-acetyl-D-galactosamine (GalNAc) metabolism. The resulting hybrid, serologically A-like/Tn (T nouvelle) structure potentially acts as a host-pathogen functional molecular bridge. In humans, this intermediate structure will hypothetically be replaced by ABO(H) blood group-specific, mucin-type structures, in the case of infection hybrid epitopes, implicating the phenotypically glycosidic accommodation of plasma proteins. The virus may, by mimicking the synthetic pathways of the ABO(H) blood groups, bind to the cell surfaces of the blood group O(H) by formation of a hybrid H-type antigen as the potential precursor of hybrid non-O blood groups, which does not affect the highly anti-glycan aggressive anti-A and anti-B isoagglutinin activities, exerted by the germline-encoded nonimmune immunoglobulin M (IgM). In the non-O blood groups, which have developed from the H-type antigen, these IgM activities are downregulated by phenotypic glycosylation, while adaptive immunoglobulins might arise in response to the hybrid A and B blood group structures, bonds between autologous carbohydrates and foreign peptides, suggesting the exertion of autoreactivity. The non-O blood groups thus become a preferred target for the virus, whereas blood group O(H) individuals, lacking the A/B phenotype-determining enzymes and binding the virus alone by hybrid H-type antigen formation, have the least molecular contact with the virus and maintain the critical anti-A and anti-B isoagglutinin activities, exerted by the ancestral IgM, which is considered the humoral spearhead of innate immunity.
Collapse
Affiliation(s)
- Peter Arend
- Philipps University Marburg, Department of Medicine, D-355 Marburg, Lahn, Germany(2); Gastroenterology Research Laboratory, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA(2); Research Laboratories, Chemie Grünenthal GmbH, D-52062 Aachen, Germany(2).
| |
Collapse
|
20
|
Skopnik CM, Al-Qaisi K, Calvert RA, Enghard P, Radbruch A, Sutton BJ, Kubagawa H. Identification of Amino Acid Residues in Human IgM Fc Receptor (FcµR) Critical for IgM Binding. Front Immunol 2021; 11:618327. [PMID: 33584711 PMCID: PMC7873564 DOI: 10.3389/fimmu.2020.618327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
Both non-immune “natural” and antigen-induced “immune” IgM are important for protection against infections and for regulation of immune responses to self-antigens. The roles of its Fc receptor (FcµR) in these IgM effector functions have begun to be explored. In the present study, by taking advantage of the difference in IgM-ligand binding of FcµRs of human (constitutive binding) and mouse (transient binding), we replaced non-conserved amino acid residues of human FcµR with mouse equivalents before establishment of cell lines stably expressing mutant or wild-type (WT) receptors. The resultant eight-different mutant FcµR-bearing cells were compared with WT receptor-bearing cells for cell-surface expression and IgM-binding by flow cytometric assessments using receptor-specific mAbs and IgM paraproteins as ligands. Three sites Asn66, Lys79-Arg83, and Asn109, which are likely in the CDR2, DE loop and CDR3 of the human FcµR Ig-like domain, respectively, were responsible for constitutive IgM binding. Intriguingly, substitution of Glu41 and Met42 in the presumed CDR1 with the corresponding mouse residues Gln and Leu, either single or more prominently in combination, enhanced both the receptor expression and IgM binding. A four-aa stretch of Lys24-Gly27 in the predicted A ß-strand of human FcµR appeared to be essential for maintenance of its proper receptor conformation on plasma membranes because of reduction of both receptor expression and IgM-binding potential when these were mutated. Results from a computational structural modeling analysis were consistent with these mutational data and identified a possible mode of binding of FcµR with IgM involving the loops including Asn66, Arg83 and Asn109 of FcµR interacting principally with the Cµ4 domain including Gln510 and to a lesser extent Cµ3 domain including Glu398, of human IgM. To our knowledge, this is the first experimental report describing the identification of amino acid residues of human FcµR critical for binding to IgM Fc.
Collapse
Affiliation(s)
| | - Khlowd Al-Qaisi
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Rosaleen A Calvert
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité-Universitätmedizin, Berlin, Germany
| | - Andreas Radbruch
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Brian J Sutton
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Hiromi Kubagawa
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
21
|
Matsuda Y, Hiramitsu T, Li XK, Watanabe T. Characteristics of Immunoglobulin M Type Antibodies of Different Origins from the Immunologic and Clinical Viewpoints and Their Application in Controlling Antibody-Mediated Allograft Rejection. Pathogens 2020; 10:pathogens10010004. [PMID: 33374617 PMCID: PMC7822424 DOI: 10.3390/pathogens10010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/25/2022] Open
Abstract
Antibody-mediated allograft rejection (AMR) hinders patient prognosis after organ transplantation. Current studies concerning AMR have mainly focused on the diagnostic value of immunoglobulin G (IgG)-type donor-specific antihuman leukocyte antigen antibodies (DSAs), primarily because of their antigen specificity, whereas the clinical significance of immunoglobulin M (IgM)-type DSAs has not been thoroughly investigated in the context of organ transplantation because of their nonspecificity against antigens. Although consensus regarding the clinical significance and role of IgM antibodies is not clear, as discussed in this review, recent findings strongly suggest that they also have a huge potential in novel diagnostic as well as therapeutic application for the prevention of AMR. Most serum IgM antibodies are known to comprise natural antibodies with low affinity toward antigens, and this is derived from B-1 cells (innate B cells). However, some of the serum IgM-type antibodies reportedly also produced by B-2 cells (conventional B cells). The latter are known to have a high affinity for donor-specific antigens. In this review, we initially discuss how IgM-type antibodies of different origins participate in the pathology of various diseases, directly or through cell surface receptors, complement activation, or cytokine production. Then, we discuss the clinical applicability of B-1 and B-2 cell-derived IgM-type antibodies for controlling AMR with reference to the involvement of IgM antibodies in various pathological conditions.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplant Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan;
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Correspondence:
| | - Takahisa Hiramitsu
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross-Hospital, Aichi 466-8650, Japan;
| | - Xiao-kang Li
- Division of Transplant Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan;
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan;
| |
Collapse
|
22
|
In silico immune infiltration profiling combined with functional enrichment analysis reveals a potential role for naïve B cells as a trigger for severe immune responses in the lungs of COVID-19 patients. PLoS One 2020; 15:e0242900. [PMID: 33264345 PMCID: PMC7710067 DOI: 10.1371/journal.pone.0242900] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, has rapidly spread to more than 160 countries worldwide since 2020. Despite tremendous efforts and resources spent worldwide trying to explore antiviral drugs, there is still no effective clinical treatment for COVID-19 to date. Approximately 15% of COVID-19 cases progress to pneumonia, and patients with severe pneumonia may die from acute respiratory distress syndrome (ARDS). It is believed that pulmonary fibrosis from SARS-CoV-2 infection further leads to ARDS, often resulting in irreversible impairment of lung function. If the mechanisms by which SARS-CoV-2 infection primarily causes an immune response or immune cell infiltration can be identified, it may be possible to mitigate excessive immune responses by modulating the infiltration and activation of specific targets, thereby reducing or preventing severe lung damage. However, the extent to which immune cell subsets are significantly altered in the lung tissues of COVID-19 patients remains to be elucidated. This study applied the CIBERSORT-X method to comprehensively evaluate the transcriptional estimated immune infiltration landscape in the lung tissues of COVID-19 patients and further compare it with the lung tissues of patients with idiopathic pulmonary fibrosis (IPF). We found a variety of immune cell subtypes in the COVID-19 group, especially naïve B cells were highly infiltrated. Comparison of functional transcriptomic analyses revealed that non-differentiated naïve B cells may be the main cause of the over-active humoral immune response. Using several publicly available single-cell RNA sequencing data to validate the genetic differences in B-cell populations, it was found that the B-cells collected from COVID-19 patients were inclined towards naïve B-cells, whereas those collected from IPF patients were inclined towards memory B-cells. Further differentiation of B cells between COVID-19 mild and severe patients showed that B cells from severe patients tended to be antibody-secreting cells, and gene expression showed that B cells from severe patients were similar to DN2 B cells that trigger extrafollicular response. Moreover, a higher percentage of B-cell infiltration seems associated with poorer clinical outcome. Finally, a comparison of several specific COVID-19 cases treated with targeted B-cell therapy suggests that appropriate suppression of naïve B cells might potentially be a novel strategy to alleviate the severe symptoms of COVID-19.
Collapse
|
23
|
Reese B, Silwal A, Daugherity E, Daugherity M, Arabi M, Daly P, Paterson Y, Woolford L, Christie A, Elias R, Brugarolas J, Wang T, Karbowniczek M, Markiewski MM. Complement as Prognostic Biomarker and Potential Therapeutic Target in Renal Cell Carcinoma. THE JOURNAL OF IMMUNOLOGY 2020; 205:3218-3229. [PMID: 33158953 DOI: 10.4049/jimmunol.2000511] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022]
Abstract
Preclinical studies demonstrated that complement promotes tumor growth. Therefore, we sought to determine the best target for complement-based therapy among common human malignancies. High expression of 11 complement genes was linked to unfavorable prognosis in renal cell carcinoma. Complement protein expression or deposition was observed mainly in stroma, leukocytes, and tumor vasculature, corresponding to a role of complement in regulating the tumor microenvironment. Complement abundance in tumors correlated with a high nuclear grade. Complement genes clustered within an aggressive inflammatory subtype of renal cancer characterized by poor prognosis, markers of T cell dysfunction, and alternatively activated macrophages. Plasma levels of complement proteins correlated with response to immune checkpoint inhibitors. Corroborating human data, complement deficiencies and blockade reduced tumor growth by enhancing antitumor immunity and seemingly reducing angiogenesis in a mouse model of kidney cancer resistant to PD-1 blockade. Overall, this study implicates complement in the immune landscape of renal cell carcinoma, and notwithstanding cohort size and preclinical model limitations, the data suggest that tumors resistant to immune checkpoint inhibitors might be suitable targets for complement-based therapy.
Collapse
Affiliation(s)
- Britney Reese
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Ashok Silwal
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Elizabeth Daugherity
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Michael Daugherity
- Department of Engineering and Physics, Abilene Christian University, Abilene, TX 79601
| | - Mahshid Arabi
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Pierce Daly
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Yvonne Paterson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Layton Woolford
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Alana Christie
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Roy Elias
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - James Brugarolas
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Tao Wang
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and.,The Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601;
| |
Collapse
|
24
|
Singh VK, Kumar S, Dhaked RK, Ansari AS, Lohiya NK, Tapryal S. Generation of oligomers of subunit vaccine candidate glycoprotein D of Herpes Simplex Virus-2 expressed in fusion with IgM Fc domain(s) in Escherichia coli: A strategy to enhance the immunogenicity of the antigen. 3 Biotech 2020; 10:463. [PMID: 33047090 PMCID: PMC7541101 DOI: 10.1007/s13205-020-02452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022] Open
Abstract
Glycoprotein D (gD) of Herpes Simplex Virus-2 is used as an antigen in various anti-herpes subunit vaccines owing to its involvement in binding the host cell receptors for host infectivity. However, most of these monomeric protein based candidates have shown low immunogenicity in animal models. To enhance the immunogenicity of gD, a fresh approach of fusing its ectodomain with the Fc domain(s) of IgM has been adopted to oligomerize the viral antigen and to exploite the immune-modulating potential of IgM Fc. Six vaccine constructs, generated by fusing three gD-ectodomain-length-variants with the Ig µ-chain domain 4 (µCH4) and µCH3-CH4 fragment, were cloned in Escherichia coli using pET28b( +) vector. The vaccine proteins were expressed in the form of inclusion bodies (IBs) and were in vitro refolded into protein oligomers of high stoichiometries of ~ 15–24, with 70–80% refolding yields. The conformations of gD and Fc components of the refolded oligomers were analyzed by ELISA and CD spectroscopy and were found to be native-like. The sizes and profiles of the size-distribution of oligomers were determined by dynamic light scattering (DLS). The candidate C2 (gD-μCH3-CH4), showing the most compact oligomer size and uniform distribution of its particles was chosen as the suitable candidate for mice immunization studies to assess the immunogenicity of the antigen gD. The C2 oligomer stimulated a strong anti-gD humoral response with an antibody titer of 102,400 and a strong, biased Th1 immune response in C57BL/6 mice, indicating its potential as a strong immunogen which may serve as an effective vaccine candidate.
Collapse
Affiliation(s)
- Vikas Kumar Singh
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| | - Sandeep Kumar
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| | - Rajeev Kumar Dhaked
- Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Abdul S. Ansari
- Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Nirmal K. Lohiya
- Indian Society for the Study of Reproduction and Fertility, Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Suman Tapryal
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| |
Collapse
|
25
|
Kumar S, Singh VK, Vasam M, Patil PS, Dhaked RK, Ansari AS, Lohiya NK, Parashar D, Tapryal S. An in vitro refolding method to produce oligomers of anti-CHIKV, E2-IgM Fc fusion subunit vaccine candidates expressed in E. coli. J Immunol Methods 2020; 487:112869. [PMID: 32971119 DOI: 10.1016/j.jim.2020.112869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022]
Abstract
Recombinant envelope protein-1 (E1) and E2 of Chikungunya virus (CHIKV) has been shown to elicit neutralizing antibodies and a balanced Th1/Th2 response in mice however with limited protection. Recently reported CHIK virus-like particles showed augmented immunity and protection in adult mice in comparison to E1 and E2, however exacerbated the disease in aged subjects. In order to improve the overall efficacy of protein based vaccines, novel strategies need to be adopted. The discovery of IgM Fc receptor (FcμR) and its role in humoral immune response led us to hypothesise that fusion of an antigen with Fc of IgM may enhance its immunogenicity by polymerizing it and FcμR mediated activation of B and other immune cells. We report in the current study, expression of E2 subunit of CHIKV in fusion with various IgM Fc domains/peptides in E. coli, their in-vitro refolding, characterization and immune response in C57BL/6 mice. Candidates fused with CH3-CH4 Fc fragment produced stable oligomers, whereas the one fused with peptides remained monomeric. The latter elicited a strong humoral and a balanced Th1/Th2 response in mice, whereas the polymeric candidate despite eliciting a strong humoral response, stimulated a biased Th1 response and exhibited higher virus neutralization in Vero cells.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Vikas Kumar Singh
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Manohar Vasam
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Poonam Shewale Patil
- Dengue & Chikungunya Group, ICMR-National Institute of Virology, 20-A Dr. Ambedkar Road, Pune 411001, India
| | - Rajeev K Dhaked
- Department of Zoology, Centre for Advanced Studies, University of Rajasthan, JLN Marg, Jaipur, Rajasthan 302004, India
| | - Abdul S Ansari
- Department of Zoology, Centre for Advanced Studies, University of Rajasthan, JLN Marg, Jaipur, Rajasthan 302004, India
| | - Nirmal K Lohiya
- Department of Zoology, Centre for Advanced Studies, Indian Society for the Study of Reproduction & Fertility, University of Rajasthan, Jaipur, Rajasthan 302004, India
| | - Deepti Parashar
- Dengue & Chikungunya Group, ICMR-National Institute of Virology, 20-A Dr. Ambedkar Road, Pune 411001, India
| | - Suman Tapryal
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
26
|
Jani PK, Kubagawa H, Melchers F. A rheostat sets B-cell receptor repertoire selection to distinguish self from non-self. Curr Opin Immunol 2020; 67:42-49. [PMID: 32916645 DOI: 10.1016/j.coi.2020.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
In bone marrow VDJ-recombination continuously generates original repertoires of immature B cells expressing IgM-B cell receptor (BcR), in which each cell recognizes the wide variety of self and non-self antigens with an individually different spectrum of avidities. High avidity self-reactive B cells try to edit their BcRs by secondary or multiple VL-rearrangements to JL-rearrangements. If they do not manage to change their self reactivity, they are deleted by apoptosis. Low avidity self-reactive B cells are anergized, while B cells with no avidity to self are ignored. A rheostat crosslinking antigen-binding BcRs, self antigen complexed with pentameric IgM and Fcμ-receptor monitors high, low or no binding. PI3K and PTEN are the effectors of this self antigen-sensing device. In mature B cells this rheostat continues to function in the activation of resting B cells by foreign antigens which crosslink BcR, antigen and pentameric IgM with Fcμ-receptors.
Collapse
Affiliation(s)
- Peter K Jani
- Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, D-10117 Berlin, Germany.
| | - Hiromi Kubagawa
- Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, D-10117 Berlin, Germany
| | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, D-10117 Berlin, Germany
| |
Collapse
|
27
|
Both maternal and newborn IgMs inhibit influenza virus-induced hemagglutination in vitro. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
28
|
Chambers ET, Gbadegesin RA. Aberrant IgM on T cells: biomarker or pathogenic factor in childhood nephrotic syndrome? Kidney Int 2020; 96:818-820. [PMID: 31543150 DOI: 10.1016/j.kint.2019.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/14/2019] [Indexed: 11/13/2022]
Abstract
Although the pathogenesis of steroid-sensitive nephrotic syndrome (SSNS) remains elusive, multiple epidemiologic, clinical, and experimental studies converge on the common theme of immune dysregulation. Initially, T-cell adaptive immunity was solely emphasized; however, the role of humoral immunity in nephrotic syndrome has gained recognition. The study by Colucci and colleagues provides preliminary evidence that production of deglycosylated IgM that is unable to regulate T-cell function in the presence or absence of corticosteroid may be responsible for a steroid-dependence course in SSNS. This study provides invaluable insights into the mechanistic roles of both T-cell and B-cell responses in the pathogenesis and clinical course of SSNS.
Collapse
Affiliation(s)
- Eileen Tsai Chambers
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Rasheed A Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
29
|
Antiphospholipid Antibodies and Autoimmune Haemolytic Anaemia: A Systematic Review and Meta-Analysis. Int J Mol Sci 2020; 21:ijms21114120. [PMID: 32527000 PMCID: PMC7313475 DOI: 10.3390/ijms21114120] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 02/02/2023] Open
Abstract
The relationship between antiphospholipid antibodies (aPL) and autoimmune haemolytic anaemia (AIHA) has never been systematically addressed. The aim of this study is to assess the link between aPL and AIHA in adult systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS). This study performed an EMBASE/PubMed search from inception to June 2019 and meta-analysis using Peto’s odds ratios. The pooled prevalence (PP) of IgG/IgM anticardiolipin (aCL) and lupus anticoagulant (LA) was greater in AIHA +ve than AIHA −ve patients (34.7% vs. 27.6%, p = 0.03; 33.3% vs. 21.8%, p < 0.0001; 20.9% vs. 8.3%, p = 0.01). The PP of AIHA was greater in: (1) IgG and IgM aCL +ve than −ve patients (21.8% vs. 11.1%, p = 0.001 and 18.7% vs. 6.3%, p < 0.0001), (2) in SLE related APS than in primary APS patients (22.8% vs. 3.9% p < 0.0001), (3) in APS +ve than APS −ve SLE patients (23.2% vs. 8.4%, p = 0.01), and (4) in thrombotic APS than non-thrombotic APS/SLE patients (26.8% vs. 10%, p = 0.03). The PP of IgG/IgM aCL and LA was greater in DAT +ve than DAT −ve patients (42.4% vs. 12.8%, p < 0.0001; 26.2% vs. 12.8%, p = 0.03 and 29.2% vs. 15.7%, p = 0.004 respectively). It was found that AIHA prevalence is maximal in SLE with aPL/APS, low-moderate in SLE without aPL and minimal in PAPS. Moreover, AIHA is rightly included among the classification criteria for SLE but not for APS/aPL. The significance of an isolated DAT positivity remains unclear in this setting
Collapse
|
30
|
Zepeda-Cervantes J, Ramírez-Jarquín JO, Vaca L. Interaction Between Virus-Like Particles (VLPs) and Pattern Recognition Receptors (PRRs) From Dendritic Cells (DCs): Toward Better Engineering of VLPs. Front Immunol 2020; 11:1100. [PMID: 32582186 PMCID: PMC7297083 DOI: 10.3389/fimmu.2020.01100] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Virus-like particles (VLPs) have been shown to be strong activators of dendritic cells (DCs). DCs are the most potent antigen presenting cells (APCs) and their activation prompts the priming of immunity mediators based on B and T cells. The first step for the activation of DCs is the binding of VLPs to pattern recognition receptors (PRRs) on the surface of DCs, followed by VLP internalization. Like wild-type viruses, VLPs use specific PRRs from the DC; however, these recognition interactions between VLPs and PRRs from DCs have not been thoroughly reviewed. In this review, we focused on the interaction between proteins that form VLPs and PRRs from DCs. Several proteins that form VLP contain glycosylations that allow the direct interaction with PRRs sensing carbohydrates, prompting DC maturation and leading to the development of strong adaptive immune responses. We also discussed how the knowledge of the molecular interaction between VLPs and PRRs from DCs can lead to the smart design of VLPs, whether based on the fusion of foreign epitopes or their chemical conjugation, as well as other modifications that have been shown to induce a stronger adaptive immune response and protection against infectious pathogens of importance in human and veterinary medicine. Finally, we address the use of VLPs as tools against cancer and allergic diseases.
Collapse
Affiliation(s)
- Jesús Zepeda-Cervantes
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Josué Orlando Ramírez-Jarquín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
31
|
Samsudin F, Yeo JY, Gan SKE, Bond PJ. Not all therapeutic antibody isotypes are equal: the case of IgM versus IgG in Pertuzumab and Trastuzumab. Chem Sci 2020; 11:2843-2854. [PMID: 32206268 PMCID: PMC7069520 DOI: 10.1039/c9sc04722k] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/12/2020] [Indexed: 01/06/2023] Open
Abstract
The therapeutic potential of immunoglobulin M (IgM) is of considerable interest in immunotherapy due to its complement-activating and cell-agglutinating abilities. Pertuzumab and Trastuzumab are monoclonal antibodies used to treat human epidermal growth factor receptor 2 (HER2)-positive breast cancer but exhibit significantly different binding affinities as IgM when compared to its IgG isotype. Using integrative multiscale modelling and simulations of complete antibody assemblies, we show that Pertuzumab IgM is able to utilize all of its V-regions to bind multiple HER2 receptors simultaneously, while similar binding in Trastuzumab IgM is prohibited by steric clashes caused by the large globular domain of HER2. This is subsequently validated by confirming that Pertuzumab IgM inhibits proliferation in HER2 over-expressing live cells more effectively than its IgG counterpart and Trastuzumab IgM. Our study highlights the importance of understanding the molecular details of antibody-antigen interactions for the design and isotype selection of therapeutic antibodies.
Collapse
Affiliation(s)
- Firdaus Samsudin
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
| | - Joshua Yi Yeo
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
| | - Samuel Ken-En Gan
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
- p53 Laboratory (ASTAR) , 8A Biomedical Grove, #06-04/05 Neuros/Immunos , Singapore 138648
- Experimental Drug Development Center (ASTAR) , 10 Biopolis Road Chromos #05-01 , Singapore 138670
| | - Peter J Bond
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
- Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , Singapore 117543 , Singapore
| |
Collapse
|
32
|
Ghouse SM, Vadrevu SK, Manne S, Reese B, Patel J, Patel B, Silwal A, Lodhi N, Paterson Y, Srivastava SK, Karbowniczek M, Markiewski MM. Therapeutic Targeting of Vasculature in the Premetastatic and Metastatic Niches Reduces Lung Metastasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:990-1000. [PMID: 31900334 PMCID: PMC7012400 DOI: 10.4049/jimmunol.1901208] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
In the metastasis-targeted organs, angiogenesis is essential for the progression of dormant micrometastases to rapidly growing and clinically overt lesions. However, we observed changes suggesting angiogenic switching in the mouse lungs prior to arrival of tumor cells (i.e., in the premetastatic niche) in the models of breast carcinoma. This angiogenic switching appears to be caused by myeloid-derived suppressor cells recruited to the premetastatic lungs through complement C5a receptor 1 signaling. These myeloid cells are known to secrete several proangiogenic factors in tumors, including IL-1β and matrix metalloproteinase-9, and we found upregulation of these genes in the premetastatic lungs. Blockade of C5a receptor 1 synergized with antiangiogenic Listeria monocytogenes-based vaccines to decrease the lung metastatic burden by reducing vascular density and improving antitumor immunity in the lungs. This was mediated even when growth of primary breast tumors was not affected by these treatments. This work provides initial evidence that angiogenesis contributes to the premetastatic niche in rapidly progressing cancers and that inhibiting this process through immunotherapy is beneficial for reducing or even preventing metastasis.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Cancer Vaccines/administration & dosage
- Cell Line, Tumor
- Combined Modality Therapy/methods
- Complement C5a/immunology
- Complement C5a/metabolism
- Female
- Humans
- Immunotherapy/methods
- Listeria monocytogenes/immunology
- Lung/blood supply
- Lung/immunology
- Lung/pathology
- Lung Neoplasms/blood supply
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Knockout
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- Neoplasm Metastasis/immunology
- Neoplasm Metastasis/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- Receptor, Anaphylatoxin C5a/antagonists & inhibitors
- Receptor, Anaphylatoxin C5a/genetics
- Receptor, Anaphylatoxin C5a/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Shanawaz M Ghouse
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Surya K Vadrevu
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Britney Reese
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Jalpa Patel
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Bhaumik Patel
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Ashok Silwal
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Niraj Lodhi
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Yvonne Paterson
- Department of Microbiology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601;
| |
Collapse
|
33
|
Regulation of Humoral Immune Responses and B Cell Tolerance by the IgM Fc Receptor (FcμR). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1254:75-86. [DOI: 10.1007/978-981-15-3532-1_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Goulet DR, Atkins WM. Considerations for the Design of Antibody-Based Therapeutics. J Pharm Sci 2020; 109:74-103. [PMID: 31173761 PMCID: PMC6891151 DOI: 10.1016/j.xphs.2019.05.031] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/02/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Antibody-based proteins have become an important class of biologic therapeutics, due in large part to the stability, specificity, and adaptability of the antibody framework. Indeed, antibodies not only have the inherent ability to bind both antigens and endogenous immune receptors but also have proven extremely amenable to protein engineering. Thus, several derivatives of the monoclonal antibody format, including bispecific antibodies, antibody-drug conjugates, and antibody fragments, have demonstrated efficacy for treating human disease, particularly in the fields of immunology and oncology. Reviewed here are considerations for the design of antibody-based therapeutics, including immunological context, therapeutic mechanisms, and engineering strategies. First, characteristics of antibodies are introduced, with emphasis on structural domains, functionally important receptors, isotypic and allotypic differences, and modifications such as glycosylation. Then, aspects of therapeutic antibody design are discussed, including identification of antigen-specific variable regions, choice of expression system, use of multispecific formats, and design of antibody derivatives based on fragmentation, oligomerization, or conjugation to other functional moieties. Finally, strategies to enhance antibody function through protein engineering are reviewed while highlighting the impact of fundamental biophysical properties on protein developability.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195.
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
35
|
Reyes-Caballero H, Park B, Loube J, Sanchez I, Vinayachandran V, Choi Y, Woo J, Edwards J, Brinkman MC, Sussan T, Mitzner W, Biswal S. Immune modulation by chronic exposure to waterpipe smoke and immediate-early gene regulation in murine lungs. Tob Control 2019; 29:s80-s89. [PMID: 31852817 DOI: 10.1136/tobaccocontrol-2019-054965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVE We investigated the effects of chronic waterpipe (WP) smoke on pulmonary function and immune response in a murine model using a research-grade WP and the effects of acute exposure on the regulation of immediate-early genes (IEGs). METHODS WP smoke was generated using three WP smoke puffing regimens based on the Beirut regimen. WP smoke samples generated under these puffing regimens were quantified for nicotine concentration. Mice were chronically exposed for 6 months followed by assessment of pulmonary function and airway inflammation. Transcriptomic analysis using RNAseq was conducted after acute exposure to characterise the IEG response. These biomarkers were then compared with those generated after exposure to dry smoke (without water added to the WP bowl). RESULTS We determined that nicotine composition in WP smoke ranged from 0.4 to 2.5 mg per puffing session. The lung immune response was sensitive to the incremental severity of chronic exposure, with modest decreases in airway inflammatory cells and chemokine levels compared with air-exposed controls. Pulmonary function was unmodified by chronic WP exposure. Acute WP exposure was found to activate the immune response and identified known and novel IEG as potential biomarkers of WP exposure. CONCLUSION Chronic exposure to WP smoke leads to immune suppression without significant changes to pulmonary function. Transcriptomic analysis of the lung after acute exposure to WP smoke showed activation of the immune response and revealed IEGs that are common to WP and dry smoke, as well as pools of IEGs unique to each exposure, identifying potential biomarkers specific to WP exposure.
Collapse
Affiliation(s)
- Hermes Reyes-Caballero
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bongsoo Park
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeffrey Loube
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ian Sanchez
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vinesh Vinayachandran
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Youngshim Choi
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Juhyung Woo
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin Edwards
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Thomas Sussan
- Toxicology Directorate, US Army Public Health Command, Aberdeen Proving Ground, Maryland, USA
| | - Wayne Mitzner
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shyam Biswal
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Blandino R, Baumgarth N. Secreted IgM: New tricks for an old molecule. J Leukoc Biol 2019; 106:1021-1034. [PMID: 31302940 PMCID: PMC6803036 DOI: 10.1002/jlb.3ri0519-161r] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/27/2022] Open
Abstract
Secreted IgM (sIgM) is a multifunctional evolutionary conserved antibody that is critical for the maintenance of tissue homeostasis as well as the development of fully protective humoral responses to pathogens. Constitutive secretion of self- and polyreactive natural IgM, produced mainly by B-1 cells, provides a circulating antibody that engages with autoantigens as well as invading pathogens, removing apoptotic and other cell debris and initiating strong immune responses. Pathogen-induced IgM production by B-1 and conventional B-2 cells strengthens this early, passive layer of IgM-mediated immune defense and regulates subsequent IgG production. The varied effects of secreted IgM on immune homeostasis and immune defense are facilitated through its binding to numerous different cell types via different receptors. Recent studies identified a novel function for pentameric IgM, namely as a transporter for the effector protein ″apoptosis-inhibitor of macrophages″ (AIM/CD5L). This review aims to provide a summary of the known functions and effects of sIgM on immune homeostasis and immune defense, and its interaction with its various receptors, and to highlight the many critical immune regulatory functions of this ancient and fascinating immunoglobulin.
Collapse
Affiliation(s)
- Rebecca Blandino
- Biochemistry, Molecular, Cellular and Developmental Biology Graduate Group, University of California, Davis
- Center for Comparative Medicine and University of California, Davis
| | - Nicole Baumgarth
- Biochemistry, Molecular, Cellular and Developmental Biology Graduate Group, University of California, Davis
- Center for Comparative Medicine and University of California, Davis
- Dept. Pathology, Microbiology & Immunology, University of California, Davis
| |
Collapse
|
37
|
Peppas I, Sollie S, Josephs DH, Hammar N, Walldius G, Karagiannis SN, Van Hemelrijck M. Serum immunoglobulin levels and the risk of bladder cancer in the AMORIS Cohort. Cancer Epidemiol 2019; 62:101584. [DOI: 10.1016/j.canep.2019.101584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/04/2019] [Accepted: 08/07/2019] [Indexed: 11/29/2022]
|
38
|
Xu L, Li Y, Yang C, Loughran P, Liao H, Hoffman R, Billiar TR, Deng M. TLR9 signaling in fibroblastic reticular cells regulates peritoneal immunity. J Clin Invest 2019; 129:3657-3669. [PMID: 31380807 DOI: 10.1172/jci127542] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Fibroblastic reticular cells (FRCs), a subpopulation of stromal cells in lymphoid organs and fat-associated lymphoid clusters (FALCs) in adipose tissue, play immune-regulatory roles in the host response to infection and may be useful as a form of cell therapy in sepsis. Here, we found an unexpected major role of TLR9 in controlling peritoneal immune cell recruitment and FALC formation at baseline and after sepsis induced by cecal ligation and puncture (CLP). TLR9 regulated peritoneal immunity via suppression of chemokine production by FRCs. Adoptive transfer of TLR9-deficient FRCs more effectively decreased mortality, bacterial load, and systemic inflammation after CLP than WT FRCs. Importantly, we found that activation of TLR9 signaling suppressed chemokine production by human adipose tissue-derived FRCs. Together, our results indicate that TLR9 plays critical roles in regulating peritoneal immunity via suppression of chemokine production by FRCs. These data form a knowledge basis upon which to design new therapeutic strategies to improve the therapeutic efficacy of FRC-based treatments for sepsis and immune dysregulation diseases.
Collapse
Affiliation(s)
- Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Yiming Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chenxuan Yang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Tsinghua University School of Medicine, Beijing, China
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hong Liao
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rosemary Hoffman
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
39
|
Law ECY, Leung DTM, Tam FCH, Cheung KKT, Cheng NHY, Lim PL. IgM Antibodies Can Access Cryptic Antigens Denied to IgG: Hypothesis on Novel Binding Mechanism. Front Immunol 2019; 10:1820. [PMID: 31428101 PMCID: PMC6688401 DOI: 10.3389/fimmu.2019.01820] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/18/2019] [Indexed: 11/13/2022] Open
Abstract
Antibodies are well-known protein mediators of immunity. IgM is the primordial member and the neglected sibling of the later-evolved and more proficient IgG in regard to their therapeutic and diagnostic use. Serendipitously, however, we found a paradox: While murine IgM antibodies specific for guanosine triphosphate (GTP) were able to recognize native guanylyl antigens found in primate or rat muscle tissues by immunofluorescence assays (which mimicked the auto-antibodies from autoimmune patients to skeletal or smooth muscle), the murine and human IgG counterparts failed. The results were replicated in cell-free direct binding assays using small latex microspheres decorated densely with GTP. The IgG antibodies could bind, however, if GTP was presented more spaciously on larger particles or as a univalent hapten. Accordingly, oligomerization of GTP (30-mer) destroyed the binding of the IgG antibodies but enhanced that of the IgMs in inhibition ELISA. We reason that, contrary to current belief, IgM does not bind in a lock-and-key manner like IgG. We hypothesize that whereas the intact and rigid antigen-binding site of IgG hinders the antibody from docking with antigens that are obstructed, in IgM, the two component polypeptides of the antigen-binding site can dissociate from each other and navigate individually through obstacles like the ancestral single-polypeptide antibodies found in sharks and camelids, both components eventually re-grouping around the antigen. We further speculate that polyreactive IgMs, which enigmatically bind to more than one type of antigen, use the same modus operandi. These findings call for a re-look at the clinical potential of IgM antibodies particularly in specific areas of cancer therapy, tissue pathology and vaccine design, where IgG antibodies have failed due to target inaccessibility.
Collapse
Affiliation(s)
- Eric Chun Yiu Law
- Clinical Immunology Unit, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Frankie Chi Hang Tam
- Clinical Immunology Unit, The Chinese University of Hong Kong, Hong Kong, China.,IgGENE, FoTan, Hong Kong, China
| | | | - Naomi Hua Yin Cheng
- Clinical Immunology Unit, The Chinese University of Hong Kong, Hong Kong, China
| | - Pak Leong Lim
- Clinical Immunology Unit, The Chinese University of Hong Kong, Hong Kong, China.,IgGENE, FoTan, Hong Kong, China
| |
Collapse
|
40
|
Fcmr regulates mononuclear phagocyte control of anti-tumor immunity. Nat Commun 2019; 10:2678. [PMID: 31213601 PMCID: PMC6581943 DOI: 10.1038/s41467-019-10619-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 05/09/2019] [Indexed: 12/13/2022] Open
Abstract
Myeloid cells contribute to tumor progression, but how the constellation of receptors they express regulates their functions within the tumor microenvironment (TME) is unclear. We demonstrate that Fcmr (Toso), the putative receptor for soluble IgM, modulates myeloid cell responses to cancer. In a syngeneic melanoma model, Fcmr ablation in myeloid cells suppressed tumor growth and extended mouse survival. Fcmr deficiency increased myeloid cell population density in this malignancy and enhanced anti-tumor immunity. Single-cell RNA sequencing of Fcmr-deficient tumor-associated mononuclear phagocytes revealed a unique subset with enhanced antigen processing/presenting properties. Conversely, Fcmr activity negatively regulated the activation and migratory capacity of myeloid cells in vivo, and T cell activation by bone marrow-derived dendritic cells in vitro. Therapeutic targeting of Fcmr during oncogenesis decreased tumor growth when used as a single agent or in combination with anti-PD-1. Thus, Fcmr regulates myeloid cell activation within the TME and may be a potential therapeutic target. Myeloid cells modulate the immune response within the tumour microenvironment, but the underlying mechanisms remain largely unknown. Here, the authors show that Fcmr – the putative receptor for soluble IgM – is a potent negative regulator of anti-tumour immunity.
Collapse
|
41
|
Kubagawa H, Honjo K, Ohkura N, Sakaguchi S, Radbruch A, Melchers F, Jani PK. Functional Roles of the IgM Fc Receptor in the Immune System. Front Immunol 2019; 10:945. [PMID: 31130948 PMCID: PMC6509151 DOI: 10.3389/fimmu.2019.00945] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/12/2019] [Indexed: 12/14/2022] Open
Abstract
It is now evident from studies of mice unable to secrete IgM that both non-immune “natural” and antigen-induced “immune” IgM are important for protection against pathogens and for regulation of immune responses to self-antigens. Since identification of its Fc receptor (FcμR) by a functional cloning strategy in 2009, the roles of FcμR in these IgM effector functions have begun to be explored. Unlike Fc receptors for switched Ig isotypes (e.g., FcγRs, FcεRs, FcαR, Fcα/μR, pIgR, FcRn), FcμR is selectively expressed by lymphocytes: B, T, and NK cells in humans and only B cells in mice. FcμR may have dual signaling ability: one through a potential as yet unidentified adaptor protein non-covalently associating with the FcμR ligand-binding chain via a His in transmembrane segment and the other through its own Tyr and Ser residues in the cytoplasmic tail. FcμR binds pentameric and hexameric IgM with a high avidity of ~10 nM in solution, but more efficiently binds IgM when it is attached to a membrane component via its Fab region on the same cell surface (cis engagement). Four different laboratories have generated Fcmr-ablated mice and eight different groups of investigators have examined the resultant phenotypes. There have been some clear discrepancies reported that appear to be due to factors including differences in the exons of Fcmr that were targeted to generate the knockouts. One common feature among these different mutant mice, however, is their propensity to produce autoantibodies of both IgM and IgG isotypes. In this review, we briefly describe recent findings concerning the functions of FcμR in both mice and humans and propose a model for how FcμR plays a regulatory role in B cell tolerance.
Collapse
Affiliation(s)
| | - Kazuhito Honjo
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Naganari Ohkura
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | | | | | - Peter K Jani
- Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
42
|
Transcription factors IRF8 and PU.1 are required for follicular B cell development and BCL6-driven germinal center responses. Proc Natl Acad Sci U S A 2019; 116:9511-9520. [PMID: 31000603 DOI: 10.1073/pnas.1901258116] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The IRF and Ets families of transcription factors regulate the expression of a range of genes involved in immune cell development and function. However, the understanding of the molecular mechanisms of each family member has been limited due to their redundancy and broad effects on multiple lineages of cells. Here, we report that double deletion of floxed Irf8 and Spi1 (encoding PU.1) by Mb1-Cre (designated DKO mice) in the B cell lineage resulted in severe defects in the development of follicular and germinal center (GC) B cells. Class-switch recombination and antibody affinity maturation were also compromised in DKO mice. RNA-seq (sequencing) and ChIP-seq analyses revealed distinct IRF8 and PU.1 target genes in follicular and activated B cells. DKO B cells had diminished expression of target genes vital for maintaining follicular B cell identity and GC development. Moreover, our findings reveal that expression of B-cell lymphoma protein 6 (BCL6), which is critical for development of germinal center B cells, is dependent on IRF8 and PU.1 in vivo, providing a mechanism for the critical role for IRF8 and PU.1 in the development of GC B cells.
Collapse
|
43
|
Berghof TVL, Matthijs MGR, Arts JAJ, Bovenhuis H, Dwars RM, van der Poel JJ, Visker MHPW, Parmentier HK. Selective breeding for high natural antibody level increases resistance to avian pathogenic Escherichia coli (APEC) in chickens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 93:45-57. [PMID: 30579935 DOI: 10.1016/j.dci.2018.12.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/15/2018] [Indexed: 06/09/2023]
Abstract
Keyhole limpet hemocyanin (KLH)-binding natural antibody (NAb) titers in chickens are heritable, and higher levels have previously been associated with a higher survival. This suggests that selective breeding for higher NAb levels might increase survival by means of improved general disease resistance. Chickens were divergently selected and bred for total NAb levels binding KLH at 16 weeks of age for six generations, resulting in a High NAb selection line and a Low NAb selection line. To for test differences in disease resistance, chickens were challenged with avian pathogenic Escherichia coli (APEC) in two separate experiments. Chickens at 8 days of age received one of four intratracheal inoculations of 0.2 mL phosphate buffered saline (PBS): 1) mock inoculate, 2) with 0.2 mL PBS containing 108.20 colony-forming units (CFU)/mL APEC, 3) with 0.2 mL PBS containing 106.64 CFU/mL APEC, and 4) with 0.2 mL PBS containing 107.55 CFU/mL APEC. Mortality was recorded during 7 days post inoculation. Overall, 50-60% reduced mortality was observed in the High line compared to the Low line for all APEC doses. In addition, morbidity was determined of the surviving chickens at 15 days of age. The High line had lower morbidity scores compared to the Low line. We conclude that selective breeding for high KLH-binding NAb levels at 16 weeks of age increase APEC resistance in early life. This study and previous studies support the hypothesis that KLH-binding NAb might be used as an indicator trait for to selective breed for general disease resistance in an antigen non-specific fashion.
Collapse
Affiliation(s)
- T V L Berghof
- Wageningen University & Research Adaptation Physiology, Wageningen, The Netherlands; Wageningen University & Research Animal Breeding and Genomics, Wageningen, The Netherlands.
| | - M G R Matthijs
- Faculty of Veterinary Medicine, Department of Farm Animal Health, Utrecht University, The Netherlands
| | - J A J Arts
- Wageningen University & Research Adaptation Physiology, Wageningen, The Netherlands
| | - H Bovenhuis
- Wageningen University & Research Animal Breeding and Genomics, Wageningen, The Netherlands
| | - R M Dwars
- Faculty of Veterinary Medicine, Department of Farm Animal Health, Utrecht University, The Netherlands
| | - J J van der Poel
- Wageningen University & Research Animal Breeding and Genomics, Wageningen, The Netherlands
| | - M H P W Visker
- Wageningen University & Research Animal Breeding and Genomics, Wageningen, The Netherlands
| | - H K Parmentier
- Wageningen University & Research Adaptation Physiology, Wageningen, The Netherlands
| |
Collapse
|
44
|
Liu J, Wang Y, Xiong E, Hong R, Lu Q, Ohno H, Wang JY. Role of the IgM Fc Receptor in Immunity and Tolerance. Front Immunol 2019; 10:529. [PMID: 30967868 PMCID: PMC6438924 DOI: 10.3389/fimmu.2019.00529] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Immunoglobulin (Ig) M is the first antibody isotype to appear during evolution, ontogeny and immune responses. IgM not only serves as the first line of host defense against infections but also plays an important role in immune regulation and immunological tolerance. For many years, IgM is thought to function by binding to antigen and activating complement system. With the discovery of the IgM Fc receptor (FcμR), it is now clear that IgM can also elicit its function through FcμR. In this review, we will describe the molecular characteristics of FcμR, its role in B cell development, maturation and activation, humoral immune responses, host defense, and immunological tolerance. We will also discuss the functional relationship between IgM-complement and IgM-FcμR pathways in regulating immunity and tolerance. Finally, we will discuss the potential involvement of FcμR in human diseases.
Collapse
Affiliation(s)
- Jun Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ermeng Xiong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rongjian Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Spencer DA, Quiniou SMA, Crider J, Musungu B, Bengten E, Wilson M. Insights into the dynamics of memory, effector and apoptotic cytotoxic T lymphocytes in channel catfish, Ictalurus punctatus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:116-128. [PMID: 30447233 DOI: 10.1016/j.dci.2018.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 06/09/2023]
Abstract
In this study, we used the channel catfish model clonal TS32.15 alloantigen-specific cytotoxic T cell (CTL) line to examine the dynamics of memory CTL expansion and senescence in teleosts. Although TS32.15 has been routinely cultured to study catfish CTL responses and killing mechanisms, little is known about the dynamics of the CTLs in these cultures. Here we show that this cell line consists of small non-cytotoxic T cells and larger granular effector T cells and that their ratios vary with time after stimulation. Small CTLs, when exposed to their irradiated targets, replicate and differentiate to morphologically distinct cytotoxic effectors, which do not replicate. After lysing target cells, or with prolonged absence of stimulation, the effector cells transition to a non-cytolytic senescent stage or become apoptotic. In addition, we demonstrate that natural IgM in catfish serum binds lipids, including PIP2, on early apoptotic CTLs, and that these IgM+ CTL can be cleared by catfish head kidney-derived macrophages.
Collapse
Affiliation(s)
- David A Spencer
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Sylvie M A Quiniou
- Warmwater Aquaculture Research Unit, U.S. Department of Agriculture-Agricultural Research Service, Stoneville, MS, 38776, USA
| | - Jonathan Crider
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Bryan Musungu
- Warmwater Aquaculture Research Unit, U.S. Department of Agriculture-Agricultural Research Service, Stoneville, MS, 38776, USA
| | - Eva Bengten
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Melanie Wilson
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| |
Collapse
|
46
|
Tay MZ, Wiehe K, Pollara J. Antibody-Dependent Cellular Phagocytosis in Antiviral Immune Responses. Front Immunol 2019; 10:332. [PMID: 30873178 PMCID: PMC6404786 DOI: 10.3389/fimmu.2019.00332] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
Antiviral activities of antibodies may either be dependent only on interactions between the antibody and cognate antigen, as in binding and neutralization of an infectious virion, or instead may require interactions between antibody-antigen immune complexes and immunoproteins or Fc receptor expressing immune effector cells. These Fc receptor-dependent antibody functions provide a direct link between the innate and adaptive immune systems by combining the potent antiviral activity of innate effector cells with the diversity and specificity of the adaptive humoral response. The Fc receptor-dependent function of antibody-dependent cellular phagocytosis (ADCP) provides mechanisms for clearance of virus and virus-infected cells, as well as for stimulation of downstream adaptive immune responses by facilitating antigen presentation, or by stimulating the secretion of inflammatory mediators. In this review, we discuss the properties of Fc receptors, antibodies, and effector cells that influence ADCP. We also provide and interpret evidence from studies that support a potential role for ADCP in either inhibiting or enhancing viral infection. Finally, we describe current approaches used to measure antiviral ADCP and discuss considerations for the translation of studies performed in animal models. We propose that additional investigation into the role of ADCP in protective viral responses, the specific virus epitopes targeted by ADCP antibodies, and the types of phagocytes and Fc receptors involved in ADCP at sites of virus infection will provide insight into strategies to successfully leverage this important immune response for improved antiviral immunity through rational vaccine design.
Collapse
Affiliation(s)
- Matthew Zirui Tay
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin Wiehe
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Justin Pollara
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
47
|
Lam JH, Baumgarth N. The Multifaceted B Cell Response to Influenza Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:351-359. [PMID: 30617116 PMCID: PMC6327962 DOI: 10.4049/jimmunol.1801208] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/17/2018] [Indexed: 01/08/2023]
Abstract
Protection from yearly recurring, highly acute infections with a pathogen that rapidly and continuously evades previously induced protective neutralizing Abs, as seen during seasonal influenza virus infections, can be expected to require a B cell response that is too highly variable, able to adapt rapidly, and able to reduce morbidity and death when sterile immunity cannot be garnered quickly enough. As we outline in this Brief Review, the influenza-specific B cell response is exactly that: it is multifaceted, involves both innate-like and conventional B cells, provides early and later immune protection, employs B cells with distinct BCR repertoires and distinct modes of activation, and continuously adapts to the ever-changing virus while enhancing overall protection. A formidable response to a formidable pathogen.
Collapse
Affiliation(s)
- Jonathan H Lam
- Center for Comparative Medicine, University of California, Davis, Davis, CA 95616
- Graduate Group in Immunology, University of California, Davis, Davis, CA 95616; and
| | - Nicole Baumgarth
- Center for Comparative Medicine, University of California, Davis, Davis, CA 95616;
- Graduate Group in Immunology, University of California, Davis, Davis, CA 95616; and
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616
| |
Collapse
|
48
|
Abstract
In contrast to adaptive antibodies, natural antibodies are present in a non-immunised organism from birth, and they do not include anti-Gal antibodies and/or anti-Gal natural antibodies, which are developed as a result of the effect of the α-Gal epitope and physiological flora. Natural antibodies are the first line of the organism’s defence before the formation of the immunity created via the stimulation of elements that determine specific and non-specific immunity. This is especially important in the case of infants. Despite the fact that natural antibodies differ in their function from adaptive antibodies, they are polyreactive and they detect autoantigens and new antigenic determinants. Natural antibodies are formed from the subpopulation of B lymphocytes, mainly B1 lymphocytes and B lymphocytes of the marginal zone. This phenomenon is supported by the fact that when the quantity of these cells in the organism decreases, which happens with age, the level of natural antibodies also decreases and the risk of illnesses of old age becomes higher. During ontogenesis, these antibodies participate in many physiological processes, including the “support” of the immune system and homeostasis, the prevention of inflammation, infections and other pathological states, such as autoimmune and cardiovascular diseases, or the process of carcinogenesis. The best known natural antibody is IgM, but the role of IgGs and IgAs is also considered important. Nowadays, many researchers also mention intravenous immunoglobulins, which are used in the treatment of numerous illnesses, and there are discussions on the possibility of increasing their potential if they were based on natural antibodies.
Collapse
|
49
|
Pérez D, Stojanovich L, Naranjo L, Stanisavljevic N, Bogdanovic G, Serrano M, Serrano A. Presence of Immune Complexes of IgG/IgM Bound to B2-glycoprotein I Is Associated With Non-criteria Clinical Manifestations in Patients With Antiphospholipid Syndrome. Front Immunol 2018; 9:2644. [PMID: 30524428 PMCID: PMC6256181 DOI: 10.3389/fimmu.2018.02644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/26/2018] [Indexed: 11/26/2022] Open
Abstract
Background: Antiphospholipid syndrome (APS) is an acquired autoimmune disorder defined by the presence of both clinical (thromboembolic events or pregnancy morbidity) and laboratory (antiphospholipid antibodies, aPL) manifestations. Despite their importance, several clinical manifestations strongly associated with APS such as livedo reticularis (LR), thrombocytopenia, sicca-ophthalmic(sicca), heart, or neurological manifestations are not included in the APS clinical classification criteria. Circulating immune complexes (CIC) formed by Beta-2-glycoprotein I (B2GPI) and aPL (B2-CIC) have been described and their presence has been related with thrombotic events. Methods: Cross-sectional and observational cohort study in APS patients with thrombotic symptomatology. Setting and Participants: Fifty-seven patients from the University Hospital Center Bezanijska Kosa (Belgrade, Serbia) who met the APS classification criteria (35 with primary APS and 22 with APS associated to systemic lupus erythematosus). This study aimed to determine the prevalence of B2-CIC in APS patients and to evaluate their association with clinical manifestations of APS not included in the classification criteria. Results: B2-CIC prevalence in APS patients was 19.3%. The presence of thrombocytopenia (OR:5.7), livedo reticularis (OR:5.6), sicca (OR:12.6), and leukopenia (OR:5.6) was significantly higher in patients with B2-CIC than in the rest of APS patients. C3 and C4 complement factor levels were significantly lower in B2-CIC positive patients, which suggests a greater consumption of complement. Patients with quadruple aPL positivity (triple aPL-positivity plus the presence of B2-CIC) showed a higher prevalence of thrombocytopenia, leucopenia and LR than those with single/double aPL-positivity. No significant differences were found in the frequencies observed in patients with triple-only vs. single/double aPL-positivity. There were no significant differences between patients with primary APS and lupus-associated APS regarding the prevalence of B2-CIC and outcomes. Conclusions: Presence of B2-CIC is strongly associated with several non-criteria clinical manifestations related to APS and to higher complement consumption. More studies are required to better understand the clinical significance of B2-CIC.
Collapse
Affiliation(s)
- Dolores Pérez
- Immunology Department, Hospital 12 de Octubre, Madrid, Spain
| | - Ljudmila Stojanovich
- Internal Medicine, "Bezanijska Kosa", University Medical Center, Belgrade, Serbia
| | - Laura Naranjo
- Immunology Department, Hospital 12 de Octubre, Madrid, Spain
| | | | - Gordana Bogdanovic
- Internal Medicine, "Bezanijska Kosa", University Medical Center, Belgrade, Serbia
| | - Manuel Serrano
- Immunology Department, Hospital 12 de Octubre, Madrid, Spain
| | - Antonio Serrano
- Immunology Department, Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
50
|
Human IgM antibody rHIgM22 promotes phagocytic clearance of myelin debris by microglia. Sci Rep 2018; 8:9392. [PMID: 29925848 PMCID: PMC6010437 DOI: 10.1038/s41598-018-27559-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/01/2018] [Indexed: 12/12/2022] Open
Abstract
In multiple sclerosis (MS), demyelinated CNS lesions fail to sufficiently remyelinate, despite the presence of oligodendrocyte precursor cells (OPCs) capable of differentiating into mature oligodendrocytes. MS lesions contain damaged myelin debris that can inhibit OPC maturation and hinder repair. rHIgM22 is an experimental human recombinant IgM antibody that promotes remyelination in animal models and is being examined in patients with MS. rHIgM22 binds to CNS myelin and partially rescues OPC process outgrowth on myelin. Since rHIgM22 does not affect OPC process outgrowth in vitro on permissive substrate, we examined the possibility that it acts by enhancing phagocytic clearance of myelin debris by microglia. In this study, we tested if rHIgM22 binding could tag myelin for microglial phagocytosis. A mouse microglial cell line and primary rat microglia were treated with myelin and rHIgM22 and assayed for myelin phagocytosis. We found that: 1) rHIgM22 stimulates myelin phagocytosis in a dose-dependent manner; 2) rHIgM22-mediated myelin phagocytosis requires actin polymerization; and 3) rHIgM22-stimulation of myelin phagocytosis requires activity of rHIgM22 Fc domain and activation of Complement Receptor 3. Since myelin inhibits OPC differentiation, stimulation of phagocytic clearance of damaged myelin may be an important means by which rHIgM22 promotes remyelination.
Collapse
|