1
|
Chen SS, Zhang H. Abrogation and homeostatic restoration of IgE responses by a universal IgE allergy CTL vaccine-The three signal self/non-self/self (S/NS/S) theory. Immunology 2024; 172:91-108. [PMID: 38303079 PMCID: PMC10987285 DOI: 10.1111/imm.13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024] Open
Abstract
Natural IgE cytotoxic peptides (nECPs), which are derived from the constant domain of the heavy chain of human IgE producing B cells via endoplasmic reticulum (ER) stress, are decorated onto MHC class 1a molecules (MHCIa) as unique biomarkers for CTL (cytotoxic T lymphocyte)-mediated immune surveillance. Human IgE exhibits only one isotype and lacks polymorphisms; IgE is pivotal in mediating diverse, allergen-specific allergies. Therefore, by disrupting self-IgE tolerance via costimulation, the CTLs induced by nECPs can serve as universal allergy vaccines (UAVs) in humans to dampen IgE production mediated by diverse allergen-specific IgE-secreting B cells and plasma cells expressing surface nECP-MHCIa as targets. The study herein has enabled the identification of nECPs, A32 and SP-1/SP-2 nonameric natural peptides produced through the correspondence principle. Vaccination using nECP induced nECP-specific CTL that profoundly suppressed human IgE production in vitro as well as chimeric human IgE production in human IgE/HLA-A2.01/HLA-B7.02 triple transgenic rodents. Furthermore, nECP-tetramer-specific CTLs were found to be converted into CD4 Tregs that restored IgE competence via the homeostatic principle, mediatepred by SREBP-1c suppressed DCs. Thus, nECPs showed causal efficacy and safety as UAVs for treating categorically type I hypersensitivity IgE-mediated allergies. The applied vaccination concept presented provides the foundation to unify, integrate through a singular class of tetramer-specific TCR clonotypes for regulaing human IgE production. The three signal theory pertains to mechanisms of three cells underlying central tolerance (S), breaking self tolerance (NS) and regaining peripheral tolerance (S) via homeostasis concerning nECP as an efficacious and safe UAV to treat type I IgE-mediated hypersensitivity. The three signal theory impirically extended, may be heuritic for immuno-regulation of adaptive immune repertoire in general.
Collapse
Affiliation(s)
- Swey-Shen Chen
- Department of Immunology and Inflammation, AAIIT LLC, San Diego, California, USA
- Division of Vaccinology and Immunotherapy, IGE Therapeutics and Pharmaceuticals, Inc, San Diego, California, USA
- Department of Protein Display and Molecular Evolution, The Institute of Genetics at San Diego, San Diego, California, USA
| | - Hailan Zhang
- Department of Immunology and Inflammation, AAIIT LLC, San Diego, California, USA
- Division of Vaccinology and Immunotherapy, IGE Therapeutics and Pharmaceuticals, Inc, San Diego, California, USA
| |
Collapse
|
2
|
Pomés A, Smith SA, Chruszcz M, Mueller GA, Brackett NF, Chapman MD. Precision engineering for localization, validation, and modification of allergenic epitopes. J Allergy Clin Immunol 2024; 153:560-571. [PMID: 38181840 PMCID: PMC10939758 DOI: 10.1016/j.jaci.2023.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
The allergen-IgE interaction is essential for the genesis of allergic responses, yet investigation of the molecular basis of these interactions is in its infancy. Precision engineering has unveiled the molecular features of allergen-antibody interactions at the atomic level. High-resolution technologies, including x-ray crystallography, nuclear magnetic resonance spectroscopy, and cryo-electron microscopy, determine allergen-antibody structures. X-ray crystallography of an allergen-antibody complex localizes in detail amino acid residues and interactions that define the epitope-paratope interface. Multiple structures involving murine IgG mAbs have recently been resolved. The number of amino acids forming the epitope broadly correlates with the epitope area. The production of human IgE mAbs from B cells of allergic subjects is an exciting recent development that has for the first time enabled an actual IgE epitope to be defined. The biologic activity of defined IgE epitopes can be validated in vivo in animal models or by measuring mediator release from engineered basophilic cell lines. Finally, gene-editing approaches using the Clustered Regularly Interspaced Short Palindromic Repeats technology to either remove allergen genes or make targeted epitope engineering at the source are on the horizon. This review presents an overview of the identification and validation of allergenic epitopes by precision engineering.
Collapse
Affiliation(s)
| | - Scott A Smith
- Vanderbilt University Medical Center, Nashville, Tenn
| | | | | | | | | |
Collapse
|
3
|
Chen SS, Zhang H. Abrogation and Homeostatic Restoration of IgE Responses by a Universal IgE Allergy CTL Vaccine-The Three Signal Self/Non-Self/Self (S/NS/S) Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.561777. [PMID: 37904962 PMCID: PMC10614744 DOI: 10.1101/2023.10.12.561777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Natural IgE cytotoxic peptides (nECPs), which are derived from the constant domain of the heavy chain of human IgE producing B cells via endoplasmic reticulum (ER) stress, are decorated onto MHC class 1a molecules (MHCIa) as unique biomarkers for CTL (cytotoxic T lymphocyte)-mediated immune surveillance. Human IgE exhibits only one isotype and lacks polymorphisms; IgE is pivotal in mediating diverse, allergen-specific allergies. Therefore, by disrupting self-IgE tolerance via costimulation, the cytotoxic T lymphocytes (CTLs) induced by nECPs can serve as universal allergy vaccines (UAVs) in humans to dampen IgE production mediated by diverse allergen-specific IgE- secreting B cells and plasma cells expressing surface nECP-MHCIa as targets. The study herein has enabled the identification of nECPs produced through the correspondence principle 1, 2 . Furthermore, nECP-tetramer-specific CTLs were found to be converted into CD4 Tregs that restored IgE competence via the homeostatic principle, mediated by SREBP-1c suppressed DCs. Thus, nECPs showed causal efficacy and safety as UAVs for treating type I hypersensitivity IgE-mediated allergies. The applied vaccination concept presented provides the foundation to unify, integrate through a singular class of tetramer-specific TCR clonotypes. The three signal model is proposed on the mechanisms underlying central tolerance, breaking tolerance and regaining peripheral tolerance via homeostasis concerning nECP as an efficacious and safe UAV to treat type I IgE-mediated hypersensitivity. One Sentence Summary Human IgE self-peptides are identified as universal allergy vaccines that inhibit IgE synthesis while allowing homeostatic IgE recovery.Graphic abstract textThree cell S/NS/S model of Universal Allergy Vaccines (UAV): Natural IgE peptides (nECPs) presented by enabler DCs break central IgE tolerance (Self), leading to CTLs that inhibit IgE production (Non-self). Generative DCs converted by the metabolic milieu transform the pre-existing nECP-specific CTLs into nECP-specific Tregs leading to homeostatic recovery of IgE competence (S).
Collapse
|
4
|
Guldenpfennig C, Teixeiro E, Daniels M. NF-kB's contribution to B cell fate decisions. Front Immunol 2023; 14:1214095. [PMID: 37533858 PMCID: PMC10391175 DOI: 10.3389/fimmu.2023.1214095] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
NF-κB signaling is essential to an effective innate and adaptive immune response. Many immune-specific functional and developmental outcomes depend in large on NF-κB. The formidable task of sorting out the mechanisms behind the regulation and outcome of NF-κB signaling remains an important area of immunology research. Here we briefly discuss the role of NF-κB in regulating cell fate decisions at various times in the path of B cell development, activation, and the generation of long-term humoral immunity.
Collapse
Affiliation(s)
- Caitlyn Guldenpfennig
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Emma Teixeiro
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Mark Daniels
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| |
Collapse
|
5
|
Baris S, Benamar M, Chen Q, Catak MC, Martínez-Blanco M, Wang M, Fong J, Massaad MJ, Sefer AP, Kara A, Babayeva R, Eltan SB, Yucelten AD, Bozkurtlar E, Cinel L, Karakoc-Aydiner E, Zheng Y, Wu H, Ozen A, Schmitz-Abe K, Chatila TA. Severe allergic dysregulation due to a gain of function mutation in the transcription factor STAT6. J Allergy Clin Immunol 2023; 152:182-194.e7. [PMID: 36758835 PMCID: PMC10330134 DOI: 10.1016/j.jaci.2023.01.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Inborn errors of immunity have been implicated in causing immune dysregulation, including allergic diseases. STAT6 is a key regulator of allergic responses. OBJECTIVES This study sought to characterize a novel gain-of-function STAT6 mutation identified in a child with severe allergic manifestations. METHODS Whole-exome and targeted gene sequencing, lymphocyte characterization, and molecular and functional analyses of mutated STAT6 were performed. RESULTS This study reports a child with a missense mutation in the DNA binding domain of STAT6 (c.1114G>A, p.E372K) who presented with severe atopic dermatitis, eosinophilia, and elevated IgE. Naive lymphocytes from the affected patient displayed increased TH2- and suppressed TH1- and TH17-cell responses. The mutation augmented both basal and cytokine-induced STAT6 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reversed STAT6 hyperresponsiveness to IL-4, normalized TH1 and TH17 cells, suppressed the eosinophilia, and improved the patient's atopic dermatitis. CONCLUSIONS This study identified a novel inborn error of immunity due to a STAT6 gain-of-function mutation that gave rise to severe allergic dysregulation. Janus kinase inhibitor therapy could represent an effective targeted treatment for this disorder.
Collapse
Affiliation(s)
- Safa Baris
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Mehmet Cihangir Catak
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mónica Martínez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Muyun Wang
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Jason Fong
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Asena Pinar Sefer
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Altan Kara
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Gebze, Turkey
| | - Royala Babayeva
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Sevgi Bilgic Eltan
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ayse Deniz Yucelten
- Department of Dermatology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Emine Bozkurtlar
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Leyla Cinel
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Yumei Zheng
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Klaus Schmitz-Abe
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
6
|
Zhou A, Shi C, Fan Y, Zheng Y, Wang J, Liu Z, Xie H, Liu J, Jiao Q. Involvement of CD40-CD40L and ICOS-ICOSL in the development of chronic rhinosinusitis by targeting eosinophils. Front Immunol 2023; 14:1171308. [PMID: 37325657 PMCID: PMC10267736 DOI: 10.3389/fimmu.2023.1171308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 06/17/2023] Open
Abstract
Background Chronic rhinosinusitis (CRS), whose prevalence and pathogenesis are age-related, is characterized by nasal tissue eosinophil infiltration. CD40-CD40 ligand (CD40L) pathway involves in the eosinophil-mediated inflammation, and inducible co-stimulator (ICOS)-ICOS ligand (ICOSL) signal can strengthen CD40-CD40L interaction. Whether CD40-CD40L and ICOS-ICOSL have a role in the development of CRS remains unknown. Objectives The aim of this study is to investigate the association of CD40-CD40L and ICOS-ICOSL expression with CRS and underlying mechanisms. Methods Immunohistology detected the expression of CD40, CD40L, ICOS, and ICOSL. Immunofluorescence was performed to evaluate the co-localizations of CD40 or ICOSL with eosinophils. Correlations between CD40-CD40L and ICOS-ICOSL as well as clinical parameters were analyzed. Flow cytometry was used to explore the activation of eosinophils by CD69 expression and the CD40 and ICOSL expression on eosinophils. Results Compared with the non-eCRS subset, ECRS (eosinophilic CRS) subset showed significantly increased CD40, ICOS, and ICOSL expression. The CD40, CD40L, ICOS, and ICOSL expressions were all positively correlated with eosinophil infiltration in nasal tissues. CD40 and ICOSL were mainly expressed on eosinophils. ICOS expression was significantly correlated with the expression of CD40-CD40L, whereas ICOSL expression was correlated with CD40 expression. ICOS-ICOSL expression positively correlated with blood eosinophils count and disease severity. rhCD40L and rhICOS significantly enhanced the activation of eosinophils from patients with ECRS. Tumor necrosis factor-α (TNF-α) and interleukin-5 (IL-5) obviously upregulated CD40 expression on eosinophils, which was significantly inhibited by the p38 mitogen-activated protein kinase (MAPK) inhibitor. Conclusions Increased CD40-CD40L and ICOS-ICOSL expressions in nasal tissues are linked to eosinophils infiltration and disease severity of CRS. CD40-CD40L and ICOS-ICOSL signals enhance eosinophils activation of ECRS. TNF-α and IL-5 regulate eosinophils function by increasing CD40 expression partly via p38 MAPK activation in patients with CRS.
Collapse
Affiliation(s)
- Aina Zhou
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenxi Shi
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhui Fan
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yushuang Zheng
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jue Wang
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhichen Liu
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huanxia Xie
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jisheng Liu
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingqing Jiao
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Richardson A, Terrazzini N, Gage C, Lee BJ, Bradley R, Watt P, Watkins ER. Inflammatory and psychological consequences of chronic high exposure firefighting. J Therm Biol 2023; 111:103399. [PMID: 36585074 DOI: 10.1016/j.jtherbio.2022.103399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/20/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES This study aimed to examine the impact of extreme heat exposure frequency on inflammation and well-being in UK Fire Service personnel. METHODS 136 Fire personnel and 14 controls (CON) were recruited [92 Firefighters (FF), 44 Breathing Apparatus Instructors (BAI)]. BAI were split into low (LBAI; ≤15 exposures per month) and high (HBAI; ≥20 exposures per month) categories. Measures of inflammation, mood and fatigue were collected at 0, 3 and 6 month times points. These variables were analysed for differences between groups and association with frequency of exposure. RESULTS HBAI exhibited raised IL-1β, IL-6, IL-10, IgE and lower IgM (p < 0.05). In addition, IL-1β, IL-6, IL-10 and IgM were associated with monthly exposure number, with exposures accounting for 15.4% of the variance in IL-6, 11.8% of IL-1β and 25.2% of IL-10. No differences in mood or fatigue were reported (p > 0.05). CONCLUSION High exposure firefighting consistently causes systemic inflammation without perceptual recognition of potential health risks.
Collapse
Affiliation(s)
- Alan Richardson
- Environmental Extremes Group, University of Brighton, UK; Centre for Stress, Aging and Disease, University of Brighton, UK
| | - Nadia Terrazzini
- Centre for Stress, Aging and Disease, University of Brighton, UK
| | - Catherine Gage
- Environmental Extremes Group, University of Brighton, UK
| | - Ben James Lee
- Occupational and Environmental Research Group, Coventry University, UK
| | - Rebecca Bradley
- Centre for Stress, Aging and Disease, University of Brighton, UK
| | - Peter Watt
- Environmental Extremes Group, University of Brighton, UK
| | | |
Collapse
|
8
|
Lang I, Zaitseva O, Wajant H. FcγRs and Their Relevance for the Activity of Anti-CD40 Antibodies. Int J Mol Sci 2022; 23:12869. [PMID: 36361658 PMCID: PMC9655775 DOI: 10.3390/ijms232112869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 03/14/2024] Open
Abstract
Inhibitory targeting of the CD40L-CD40 system is a promising therapeutic option in the field of organ transplantation and is also attractive in the treatment of autoimmune diseases. After early complex results with neutralizing CD40L antibodies, it turned out that lack of Fcγ receptor (FcγR)-binding is the crucial factor for the development of safe inhibitory antibodies targeting CD40L or CD40. Indeed, in recent years, blocking CD40 antibodies not interacting with FcγRs, has proven to be well tolerated in clinical studies and has shown initial clinical efficacy. Stimulation of CD40 is also of considerable therapeutic interest, especially in cancer immunotherapy. CD40 can be robustly activated by genetically engineered variants of soluble CD40L but also by anti-CD40 antibodies. However, the development of CD40L-based agonists is biotechnologically and pharmacokinetically challenging, and anti-CD40 antibodies typically display only strong agonism in complex with FcγRs or upon secondary crosslinking. The latter, however, typically results in poorly developable mixtures of molecule species of varying stoichiometry and FcγR-binding by anti-CD40 antibodies can elicit unwanted side effects such as antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP) of CD40 expressing immune cells. Here, we summarize and compare strategies to overcome the unwanted target cell-destroying activity of anti-CD40-FcγR complexes, especially the use of FcγR type-specific mutants and the FcγR-independent cell surface anchoring of bispecific anti-CD40 fusion proteins. Especially, we discuss the therapeutic potential of these strategies in view of the emerging evidence for the dose-limiting activities of systemic CD40 engagement.
Collapse
Affiliation(s)
| | | | - Harald Wajant
- Department of Internal Medicine II, Division of Molecular Internal Medicine, University Hospital Würzburg, Auvera Haus, Grombühlstrasse 12, 97080 Würzburg, Germany
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW B cells are known in food allergy pathogenesis for their production of IgE but their roles in the development of tolerance to foods are not well understood. Further understanding of B-cell biology in the context of food allergy is essential for the creation of effective prevention strategies and therapies. RECENT FINDINGS The majority of allergen-specific IgE in humans appears to arise from antigen-experienced B cells that have already undergone class switch recombination to other antibody isotypes, such as IgG1, and can also be produced by cells class switching to IgE locally in the gastrointestinal tract. Allergen-specific IgG4 can have protective effects in individuals and is associated with tolerance. Regulatory B cells, which can produce allergen-specific IgG4, are reduced in food-allergic individuals and may also be an important component of tolerance. Therapeutic approaches that block the generation and action of IgE and that enhance tolerizing immune responses are being evaluated for the treatment of food allergy. SUMMARY B cells play several roles in the development of food allergy versus tolerance. Their functions may translate into the care of food allergy as biomarkers or therapeutic targets and can be employed in other atopic diseases to better understand their pathogenesis and create new avenues for treatment.
Collapse
Affiliation(s)
- Chioma Udemgba
- Allergy and Clinical Immunology Fellowship Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Adora Lin
- Center for Cancer and Immunology Research, Children’s National Research Institute, Washington, DC
| |
Collapse
|
10
|
CD40-CD154: A perspective from type 2 immunity. Semin Immunol 2021; 53:101528. [PMID: 34810089 DOI: 10.1016/j.smim.2021.101528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
The interaction between CD40 and CD154 (CD40 ligand) is central in immunology, participating in CD4+ T cell priming by dendritic cells (DC), CD4+ T cell help to B cells and classical macrophage activation by CD4+ T cells. However, its role in the Th2 side of immunology including helminth infection remains incompletely understood. Contrary to viral and bacterial stimuli, helminth products usually do not cause CD40 up-regulation in DC, and exogenous CD40 ligation drives Th2-biased systems towards Th1. On the other hand, CD40 and CD154 are necessary for induction of most Th2 responses. We attempt to reconcile these observations, mainly by proposing that (i) CD40 up-regulation in DC in Th2 systems is mostly induced by alarmins, (ii) the Th2 to Th1 shift induced by exogenous CD40 ligation is related to the capacity of such ligation to enhance IL-12 production by myeloid cells, and (iii) signals elicited by endogenous CD154 available in Th2 contexts and by exogenous CD40 ligation are probably different. We stress that CD40-CD154 is important beyond cognate cellular interactions. In such a context, we argue that the proliferation response of B-cells to IL-4 plus CD154 reflects a Th2-specific mechanism for polyclonal B-cell amplification and IgE production at infection sites. Finally, we argue that CD154 is a general immune activation signal across immune polarization including Th2, and propose that competition for CD154 at tissue sites may provide negative feedback on response induction at each site.
Collapse
|
11
|
Zhang J, Wu Z, Yu F, Ye L, Gu W, Tan Y, Wang L, Shi Y. Role of liver-X-receptors in airway remodeling in mice with chronic allergic asthma. Exp Ther Med 2021; 22:920. [PMID: 34335881 PMCID: PMC8290420 DOI: 10.3892/etm.2021.10352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Liver X receptors (LXRs) exert anti-inflammatory effects in animal models of certain respiratory diseases. In the present study, a model of chronic airway remodeling was established in wild-type and LXR-deficient mice. Ovalbumin (OVA)-sensitized mice were chronically administered OVA via inhalation for 8 weeks. Prior to each stimulation, certain wild-type mice were treated with GW3965, which is a highly selective LXR agonist. The influence of LXRs on airway inflammation, airway hyperresponsiveness and airway remodeling was evaluated. LXRs were indicated to increase airway inflammation and airway hyperresponsiveness, as well as promote airway remodeling. These results suggest that inhibiting LXRs may be a potential method for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Jinmei Zhang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Zhengcan Wu
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Fenfang Yu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Liang Ye
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Wei Gu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yan Tan
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Li Wang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Ying Shi
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
12
|
Yang Z, Wu CAM, Targ S, Allen CDC. IL-21 is a broad negative regulator of IgE class switch recombination in mouse and human B cells. J Exp Med 2020; 217:133860. [PMID: 32130409 PMCID: PMC7201927 DOI: 10.1084/jem.20190472] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/24/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
IgE antibodies may elicit potent allergic reactions, and their production is tightly controlled. The tendency to generate IgE has been thought to reflect the balance between type 1 and type 2 cytokines, with the latter promoting IgE. Here, we reevaluated this paradigm by a direct cellular analysis, demonstrating that IgE production was not limited to type 2 immune responses yet was generally constrained in vivo. IL-21 was a critical negative regulator of IgE responses, whereas IFN-γ, IL-6, and IL-10 were dispensable. Follicular helper T cells were the primary source of IL-21 that inhibited IgE responses by directly engaging the IL-21 receptor on B cells and triggering STAT3-dependent signaling. We reconciled previous discordant results between mouse and human B cells and revealed that the inhibition of IgE class switch recombination by IL-21 was attenuated by CD40 signaling, whereas IgG1 class switch recombination was potentiated by IL-21 in the context of limited IL-4. These findings establish key features of the extrinsic regulation of IgE production by cytokines.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Chung-An M Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Sasha Targ
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA.,Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
13
|
Spencer S, Köstel Bal S, Egner W, Lango Allen H, Raza SI, Ma CA, Gürel M, Zhang Y, Sun G, Sabroe RA, Greene D, Rae W, Shahin T, Kania K, Ardy RC, Thian M, Staples E, Pecchia-Bekkum A, Worrall WPM, Stephens J, Brown M, Tuna S, York M, Shackley F, Kerrin D, Sargur R, Condliffe A, Tipu HN, Kuehn HS, Rosenzweig SD, Turro E, Tavaré S, Thrasher AJ, Jodrell DI, Smith KGC, Boztug K, Milner JD, Thaventhiran JED. Loss of the interleukin-6 receptor causes immunodeficiency, atopy, and abnormal inflammatory responses. J Exp Med 2019; 216:1986-1998. [PMID: 31235509 PMCID: PMC6719421 DOI: 10.1084/jem.20190344] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/20/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022] Open
Abstract
IL-6 excess is central to the pathogenesis of multiple inflammatory conditions and is targeted in clinical practice by immunotherapy that blocks the IL-6 receptor encoded by IL6R We describe two patients with homozygous mutations in IL6R who presented with recurrent infections, abnormal acute-phase responses, elevated IgE, eczema, and eosinophilia. This study identifies a novel primary immunodeficiency, clarifying the contribution of IL-6 to the phenotype of patients with mutations in IL6ST, STAT3, and ZNF341, genes encoding different components of the IL-6 signaling pathway, and alerts us to the potential toxicity of drugs targeting the IL-6R.
Collapse
Affiliation(s)
- Sarah Spencer
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Sevgi Köstel Bal
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - William Egner
- Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Hana Lango Allen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Syed I Raza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Chi A Ma
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Meltem Gürel
- Cancer Research UK Cambridge Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Yuan Zhang
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Guangping Sun
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ruth A Sabroe
- Department of Dermatology, Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
| | - Daniel Greene
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - William Rae
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Tala Shahin
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Katarzyna Kania
- Cancer Research UK Cambridge Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Rico Chandra Ardy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Marini Thian
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- St. Anna Kinderspital and Children's Cancer Research Institute, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Emily Staples
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Annika Pecchia-Bekkum
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - William P M Worrall
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jonathan Stephens
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew Brown
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Salih Tuna
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Melanie York
- Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Fiona Shackley
- Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Diarmuid Kerrin
- Barnsley Hospitals National Health Service Foundation Trust, Barnsley, UK
| | - Ravishankar Sargur
- Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alison Condliffe
- Sheffield Teaching Hospitals National Health Service Trust, Sheffield, UK
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Hamid Nawaz Tipu
- Immunology Department, Armed Forces Institute of Pathology, Rawalpindi, Pakistan
| | - Hye Sun Kuehn
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Sergio D Rosenzweig
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Simon Tavaré
- Cancer Research UK Cambridge Institute, Cambridge Biomedical Campus, Cambridge, UK
- Herbert and Florence Irving Institute for Cancer Dynamics, Columbia University, New York, NY
- New York Genome Center, New York, NY
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, Great Ormond Street Hospital National Health Service Trust, London, UK
| | - Duncan Ian Jodrell
- Department of Oncology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Kenneth G C Smith
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- St. Anna Kinderspital and Children's Cancer Research Institute, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Vienna Center for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - James E D Thaventhiran
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Institute, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
14
|
Tracing IgE-Producing Cells in Allergic Patients. Cells 2019; 8:cells8090994. [PMID: 31466324 PMCID: PMC6769703 DOI: 10.3390/cells8090994] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/13/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Immunoglobulin E (IgE) is the key immunoglobulin in the pathogenesis of IgE associated allergic diseases affecting 30% of the world population. Recent data suggest that allergen-specific IgE levels in serum of allergic patients are sustained by two different mechanisms: inducible IgE production through allergen exposure, and continuous IgE production occurring even in the absence of allergen stimulus that maintains IgE levels. This assumption is supported by two observations. First, allergen exposure induces transient increases of systemic IgE production. Second, reduction in IgE levels upon depletion of IgE from the blood of allergic patients using immunoapheresis is only temporary and IgE levels quickly return to pre-treatment levels even in the absence of allergen exposure. Though IgE production has been observed in the peripheral blood and locally in various human tissues (e.g., nose, lung, spleen, bone marrow), the origin and main sites of IgE production in humans remain unknown. Furthermore, IgE-producing cells in humans have yet to be fully characterized. Capturing IgE-producing cells is challenging not only because current staining technologies are inadequate, but also because the cells are rare, they are difficult to discriminate from cells bearing IgE bound to IgE-receptors, and plasma cells express little IgE on their surface. However, due to the central role in mediating both the early and late phases of allergy, free IgE, IgE-bearing effector cells and IgE-producing cells are important therapeutic targets. Here, we discuss current knowledge and unanswered questions regarding IgE production in allergic patients as well as possible therapeutic approaches targeting IgE.
Collapse
|
15
|
Piccinni MP, Lombardelli L, Logiodice F, Kullolli O, Maggi E, Barkley MS. Medroxyprogesterone Acetate Decreases Th1, Th17, and Increases Th22 Responses via AHR Signaling Which Could Affect Susceptibility to Infections and Inflammatory Disease. Front Immunol 2019; 10:642. [PMID: 31001262 PMCID: PMC6456711 DOI: 10.3389/fimmu.2019.00642] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022] Open
Abstract
A synthetic progestin, medroxyprogesterone acetate (MPA), was used in a novel study to determine progestin effects on human purified macrophages and Th1, Th2, Th17, Th22 cells. MPA concentrations were equivalent to those in the serum of women after 6 and 9 months of progestin use. MPA has no effect on the proliferation of PBMCs and CD4+ T cell clones induced by immobilized anti-CD3 antibodies or by antigen (streptokinase). However, MPA decreases production and mRNA expression of IL-5, IL-13, IFN-γ, T-bet, RORC, and IL-17A but increases production and mRNA expression of IL-22 by CD4+ Th22 cell clones and decreases IL-22 production by Th17 cells. MPA inhibits RORC, but not T-bet and AHR, by Th17 cells but increases AHR mRNA and T-bet expression of established CD4+ Th22 cell clones. This suggests that MPA, at concentrations equivalent to those found in the serum of women after treatment for contraception and hormone replacement therapy, can directly inhibit Th1 responses (against intracellular bacteria and viruses), Th17 (against extracellular bacteria and fungi), Th2 (against parasites) but MPA therapy increases IL-22 produced by Th22 cells mediated by an increased expression of AHR and T-bet controlling inflammation. MPA could be responsible for the tissue damage limited by IL-22 in absence of IL-17A.
Collapse
Affiliation(s)
- Marie-Pierre Piccinni
- Department of Experimental and Clinical Medicine and Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Florence, Italy
| | - Letizia Lombardelli
- Department of Experimental and Clinical Medicine and Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Florence, Italy
| | - Federica Logiodice
- Department of Experimental and Clinical Medicine and Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Florence, Italy
| | - Ornela Kullolli
- Department of Experimental and Clinical Medicine and Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Florence, Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Florence, Italy.,Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Marylynn S Barkley
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
| |
Collapse
|
16
|
Role of IL-35 in sublingual allergen immunotherapy. J Allergy Clin Immunol 2018; 143:1131-1142.e4. [PMID: 30053528 DOI: 10.1016/j.jaci.2018.06.041] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/06/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Grass pollen-specific immunotherapy involves immunomodulation of allergen-specific TH2 responses and induction of IL-10+ and/or TGF-β+CD4+CD25+ regulatory T cells (induced Treg cells). IL-35+CD4+CD25+ forkhead box protein 3-negative T (IL-35-inducible regulatory T [iTR35]) cells have been reported as a novel subset of induced Treg cells with modulatory characteristics. OBJECTIVE We sought to investigate mechanisms underlying the induction and maintenance of immunologic tolerance induced by IL-35 and iTR35 cells. METHODS The biological effects of IL-35 were assessed on group 2 innate lymphoid cells (ILC2s); dendritic cells primed with thymic stromal lymphopoietin, IL-25, and IL-33; and B and TH2 cells by using flow cytometry and quantitative RT-PCR. Grass pollen-driven TH2 cell proliferation and cytokine production were measured by using tritiated thymidine and Luminex MagPix, respectively. iTR35 cells were quantified in patients with grass pollen allergy (seasonal allergic rhinitis [SAR] group, n = 16), sublingual immunotherapy (SLIT)-treated patients (SLIT group, n = 16), and nonatopic control subjects (NACs; NAC group, n = 16). RESULTS The SAR group had increased proportions of ILC2s (P = .002) and IL-5+ cells (P = .042), IL-13+ cells (P = .042), and IL-5+IL-13+ ILC2s (P = .003) compared with NACs. IL-35 inhibited IL-5 and IL-13 production by ILC2s in the presence of IL-25 or IL-33 (P = .031) and allergen-driven TH2 cytokines by effector T cells. IL-35 inhibited CD40 ligand-, IL-4-, and IL-21-mediated IgE production by B cells (P = .015), allergen-driven T-cell proliferation (P = .001), and TH2 cytokine production mediated by primed dendritic cells. iTR35 cells suppressed TH2 cell proliferation and cytokine production. In addition, allergen-driven IL-35 levels and iTR35 cell counts were increased in patients receiving SLIT (all, P < .001) and NACs (all, P < .001) compared with patients with SAR. CONCLUSION IL-35 and iTR35 cells are potential novel immune regulators induced by SLIT. The clinical relevance of SLIT can be underscored by restoration of protective iTR35 cells.
Collapse
|
17
|
Fecher P, Caspell R, Naeem V, Karulin AY, Kuerten S, Lehmann PV. B Cells and B Cell Blasts Withstand Cryopreservation While Retaining Their Functionality for Producing Antibody. Cells 2018; 7:E50. [PMID: 29857548 PMCID: PMC6028916 DOI: 10.3390/cells7060050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/02/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022] Open
Abstract
In individuals who have once developed humoral immunity to an infectious/foreign antigen, the antibodies present in their body can mediate instant protection when the antigen re-enters. Such antigen-specific antibodies can be readily detected in the serum. Long term humoral immunity is, however, also critically dependent on the ability of memory B cells to engage in a secondary antibody response upon re-exposure to the antigen. Antibody molecules in the body are short lived, having a half-life of weeks, while memory B cells have a life span of decades. Therefore, the presence of serum antibodies is not always a reliable indicator of B cell memory and comprehensive monitoring of humoral immunity requires that both serum antibodies and memory B cells be assessed. The prevailing view is that resting memory B cells and B cell blasts in peripheral blood mononuclear cells (PBMC) cannot be cryopreserved without losing their antibody secreting function, and regulated high throughput immune monitoring of B cell immunity is therefore confined to-and largely limited by-the need to test freshly isolated PBMC. Using optimized protocols for freezing and thawing of PBMC, and four color ImmunoSpot® analysis for the simultaneous detection of all immunoglobulin classes/subclasses we show here that both resting memory B cells and B cell blasts retain their ability to secrete antibody after thawing, and thus demonstrate the feasibility of B cell immune monitoring using cryopreserved PBMC.
Collapse
Affiliation(s)
- Philipp Fecher
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Richard Caspell
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Villian Naeem
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Alexey Y Karulin
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Paul V Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| |
Collapse
|
18
|
Eckl-Dorna J, Fröschl R, Lupinek C, Kiss R, Gattinger P, Marth K, Campana R, Mittermann I, Blatt K, Valent P, Selb R, Mayer A, Gangl K, Steiner I, Gamper J, Perkmann T, Zieglmayer P, Gevaert P, Valenta R, Niederberger V. Intranasal administration of allergen increases specific IgE whereas intranasal omalizumab does not increase serum IgE levels-A pilot study. Allergy 2018; 73:1003-1012. [PMID: 29083477 PMCID: PMC5969304 DOI: 10.1111/all.13343] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
Background Administration of the therapeutic anti‐IgE antibody omalizumab to patients induces strong increases in IgE antibody levels. Objective To investigate the effect of intranasal administration of major birch pollen allergen Bet v 1, omalizumab or placebo on the levels of total and allergen‐specific IgE in patients with birch pollen allergy. Methods Based on the fact that intranasal allergen application induces rises of systemic allergen‐specific IgE, we performed a double‐blind placebo‐controlled pilot trial in which birch pollen allergic subjects were challenged intranasally with omalizumab, placebo or birch pollen allergen Bet v 1. Total and allergen‐specific IgE, IgG and basophil sensitivity were measured before and 8 weeks after challenge. For control purposes, total, allergen‐specific IgE levels and omalizumab‐IgE complexes as well as specific IgG levels were studied in subjects treated subcutaneously with either omalizumab or placebo. Effects of omalizumab on IgE production by IL‐4/anti‐CD40‐treated PBMCs from allergic patients were studied in vitro. Results Intranasal challenge with Bet v 1 induced increases in Bet v 1‐specific IgE levels by a median of 59.2%, and this change differed significantly from the other treatment groups (P = .016). No relevant change in allergen‐specific and total IgE levels was observed in subjects challenged with omalizumab. Addition of omalizumab did not enhance IL‐4/anti‐CD40‐induced IgE production in vitro. Significant rises in total IgE (mean IgE before: 131.83 kU/L to mean IgE after: 505.23 kU/L) and the presence of IgE‐omalizumab complexes were observed after subcutaneous administration of omalizumab. Conclusion Intranasal administration of allergen induced rises of allergen‐specific IgE levels, whereas intranasal administration of omalizumab did not enhance systemic total or allergen‐specific IgE levels.
Collapse
Affiliation(s)
- J. Eckl-Dorna
- Department of Otorhinolaryngology; Medical University of Vienna; Vienna Austria
| | - R. Fröschl
- Clinical Institute for Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - C. Lupinek
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - R. Kiss
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - P. Gattinger
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - K. Marth
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - R. Campana
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - I. Mittermann
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - K. Blatt
- Division of Hematology and Hemostaseology; Department of Internal Medicine I; Medical University of Vienna; Vienna Austria
| | - P. Valent
- Division of Hematology and Hemostaseology; Department of Internal Medicine I; Medical University of Vienna; Vienna Austria
| | - R. Selb
- Department of Otorhinolaryngology; Medical University of Vienna; Vienna Austria
| | - A. Mayer
- Department of Otorhinolaryngology; Medical University of Vienna; Vienna Austria
| | - K. Gangl
- Department of Otorhinolaryngology; Medical University of Vienna; Vienna Austria
| | - I. Steiner
- Center for Medical Statistics, Informatics, and Intelligent Systems; Section for Medical Statistics; Medical University of Vienna; Vienna Austria
| | - J. Gamper
- Center for Medical Statistics, Informatics, and Intelligent Systems; Section for Medical Statistics; Medical University of Vienna; Vienna Austria
| | - T. Perkmann
- Clinical Institute for Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | | | - P. Gevaert
- Upper Airway Research Laboratory (URL); Ghent University Hospital; Ghent Belgium
| | - R. Valenta
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - V. Niederberger
- Department of Otorhinolaryngology; Medical University of Vienna; Vienna Austria
| |
Collapse
|
19
|
Ettinger R, Karnell JL, Henault J, Panda SK, Riggs JM, Kolbeck R, Sanjuan MA. Pathogenic mechanisms of IgE-mediated inflammation in self-destructive autoimmune responses. Autoimmunity 2017; 50:25-36. [PMID: 28166684 DOI: 10.1080/08916934.2017.1280670] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autoantibodies of the IgG subclass are pathogenic in a number of autoimmune disorders such as systemic lupus erythomatosus. The presence of circulating IgE autoantibodies in autoimmune patients has also been known for almost 40 years. Despite their role in allergies, IgE autoantibodies are not associated with a higher rate of atopy in these patients. However, recently they have been recognized as active drivers of autoimmunity through mechanisms involving the secretion of Type I interferons by plasmacytoid dendritic cells (pDC), the recruitment of basophils to lymph nodes, and the activation of adaptive immune responses through B and T cells. Here, we will review the formation, prevalence, affinity, and roles of the IgE autoantibodies that have been described in autoimmunity. We also present novel evidence supporting that triggering of IgE receptors in pDC induces LC3-associated phagocytosis, a cellular process also known as LAP that is associated with interferon responses. The activation of pDC with immune complexes formed by DNA-specific IgE antibodies also induce potent B-cell differentiation and plasma cell formation, which further define IgE's role in autoimmune humoral responses.
Collapse
Affiliation(s)
- Rachel Ettinger
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Jodi L Karnell
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Jill Henault
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Santosh K Panda
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Jeffrey M Riggs
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Roland Kolbeck
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| | - Miguel A Sanjuan
- a Department of Respiratory , Inflammation & Autoimmunity, MedImmune LLC , Gaithersburg , MD , USA
| |
Collapse
|
20
|
DOCK8 and STAT3 dependent inhibition of IgE isotype switching by TLR9 ligation in human B cells. Clin Immunol 2017; 183:263-265. [PMID: 28882618 DOI: 10.1016/j.clim.2017.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 11/22/2022]
|
21
|
Gon Y, Maruoka S, Inoue T, Mizumura K, Kuroda K, Fukano Y, Yamagishi K, Tsuboi E, Hashimoto S. Gene expression analysis in airway-secreting extracellular vesicles upon house dust mite exposure. Allergol Int 2016; 65 Suppl:S53-5. [PMID: 27184857 DOI: 10.1016/j.alit.2016.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/26/2016] [Accepted: 04/08/2016] [Indexed: 11/17/2022] Open
Affiliation(s)
- Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan.
| | - Toshio Inoue
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenji Mizumura
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kazumichi Kuroda
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshihito Fukano
- Department of Chemical Biology and Applied Chemistry, College of Engineering, Nihon University, Fukushima, Japan
| | - Kenji Yamagishi
- Department of Chemical Biology and Applied Chemistry, College of Engineering, Nihon University, Fukushima, Japan
| | - Eriko Tsuboi
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shu Hashimoto
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Kratzer B, Pickl WF. Years in Review: Recent Progress in Cellular Allergology. Int Arch Allergy Immunol 2016; 169:1-12. [PMID: 26953825 PMCID: PMC7058417 DOI: 10.1159/000444753] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This review highlights the recent key advances in the biology of CD4+ effector T cells, antigen-presenting cells, Th17 and T regulatory cells, as well as immediate effector cells, such as mast cells, basophils and eosinophils, which are critically contributing to the better understanding of the pathophysiology of allergic diseases and are helping to improve their diagnosis and therapy. Some of the key advances with a direct impact on allergic asthma research and treatment are summarized.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
23
|
Jabara HH, Boyden SE, Chou J, Ramesh N, Massaad MJ, Benson H, Bainter W, Fraulino D, Rahimov F, Sieff C, Liu ZJ, Alshemmari SH, Al-Ramadi BK, Al-Dhekri H, Arnaout R, Abu-Shukair M, Vatsayan A, Silver E, Ahuja S, Davies EG, Sola-Visner M, Ohsumi TK, Andrews NC, Notarangelo LD, Fleming MD, Al-Herz W, Kunkel LM, Geha RS. A missense mutation in TFRC, encoding transferrin receptor 1, causes combined immunodeficiency. Nat Genet 2016; 48:74-8. [PMID: 26642240 PMCID: PMC4696875 DOI: 10.1038/ng.3465] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 11/13/2015] [Indexed: 02/05/2023]
Abstract
Patients with a combined immunodeficiency characterized by normal numbers but impaired function of T and B cells had a homozygous p.Tyr20His substitution in transferrin receptor 1 (TfR1), encoded by TFRC. The substitution disrupts the TfR1 internalization motif, resulting in defective receptor endocytosis and markedly increased TfR1 expression on the cell surface. Iron citrate rescued the lymphocyte defects, and expression of wild-type but not mutant TfR1 rescued impaired transferrin uptake in patient-derived fibroblasts. Tfrc(Y20H/Y20H) mice recapitulated the immunological defects of patients. Despite the critical role of TfR1 in erythrocyte development and function, patients had only mild anemia and only slightly increased TfR1 expression in erythroid precursors. We show that STEAP3, a metalloreductase expressed in erythroblasts, associates with TfR1 and partially rescues transferrin uptake in patient-derived fibroblasts, suggesting that STEAP3 may provide an accessory TfR1 endocytosis signal that spares patients from severe anemia. These findings demonstrate the importance of TfR1 in adaptive immunity.
Collapse
MESH Headings
- Adaptive Immunity/genetics
- Anemia/genetics
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Cycle Proteins
- Cells, Cultured
- Endocytosis
- Female
- Fibroblasts/physiology
- Humans
- Immunologic Deficiency Syndromes/genetics
- Male
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Mutation, Missense
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Oxidoreductases
- Pedigree
- Receptors, Transferrin/genetics
- Receptors, Transferrin/immunology
- Receptors, Transferrin/metabolism
Collapse
Affiliation(s)
- Haifa H Jabara
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven E Boyden
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Narayanaswamy Ramesh
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Halli Benson
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Wayne Bainter
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David Fraulino
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Fedik Rahimov
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Colin Sieff
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhi-Jian Liu
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Neonatology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Salem H Alshemmari
- Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Hasan Al-Dhekri
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rand Arnaout
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohammad Abu-Shukair
- Immunology, Allergy and Rheumatology Division, Queen Rania Hospital for Children, Amman, Jordan
| | - Anant Vatsayan
- Department of Pediatric Hematology/Oncology, University Hospitals Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - Eli Silver
- Division of Pulmonology and Sleep Medicine, University Hospitals Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sanjay Ahuja
- Department of Pediatric Hematology/Oncology, University Hospitals Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - E Graham Davies
- Centre for Immunodeficiency, Great Ormond Street Hospital and Institute of Child Health, London, UK
| | - Martha Sola-Visner
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Neonatology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Toshiro K Ohsumi
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nancy C Andrews
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Luigi D Notarangelo
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Louis M Kunkel
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Generation of a soluble recombinant trimeric form of bovine CD40L and its potential use as a vaccine adjuvant in cows. Vet Immunol Immunopathol 2015; 168:1-13. [PMID: 26553560 DOI: 10.1016/j.vetimm.2015.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/11/2015] [Accepted: 09/22/2015] [Indexed: 11/24/2022]
Abstract
Vaccination is the most cost-effective way to control infectious diseases in cattle. However, many infectious diseases leading to severe economical losses worldwide still remain for which a really effective and safe vaccine is not available. These diseases are most often due to intracellular pathogens such as bacteria or viruses, which are, by their localization, protected from antibiotics and/or CD4(+) T cell-dependent humoral responses. We therefore postulated that strategies leading to induction of not only CD4(+) T cell responses but also CD8(+) cytotoxic T lymphocyte (CTL) responses against infected cells should be privileged in the development of new vaccines against problematic intracellular pathogens in bovines. CD40 signaling in antigen-presenting cells may lead to the induction of robust CD4-independent CTL responses and several studies, especially in mice, have used CD40 stimulation to promote CD8(+) T cell-mediated immunity. For example, we have recently shown that immunization of mice with heat-killed Staphylococcus aureus (HKSA) and agonistic anti-CD40 monoclonal antibodies elicits strong CTL responses capable of protecting mice from subsequent staphylococcal mastitis. Unfortunately, there is at present no tool available to efficiently stimulate CD40 in cattle. In this study, we therefore first produced a soluble recombinant trimeric form of the natural bovine CD40 ligand (sboCD40LT). We then observed that sboCD40LT was able to potently stimulate bovine cells in vitro. Finally, we provide evidence that immunization of cows with sboCD40LT combined with HKSA was able to significantly increase the number of both HKSA-specific CD4(+) and CD8(+) T cells in the draining lymph nodes. In conclusion, we suggest that this new molecular tool could help in the development of vaccine strategies against bovine diseases caused by intracellular pathogens.
Collapse
|
25
|
Clark EA. A Short History of the B-Cell-Associated Surface Molecule CD40. Front Immunol 2014; 5:472. [PMID: 25324844 PMCID: PMC4179537 DOI: 10.3389/fimmu.2014.00472] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/14/2014] [Indexed: 12/28/2022] Open
Abstract
This perspective traces developments using monoclonal antibody technology that led to the discovery of CD40, a receptor that on B cells mediates “T cell help” and on dendritic cells helps to program CD8 T cell responses. I discuss some things that we got right during the path of discovery and some things we missed. Immunotherapies that block or stimulate the CD40 pathway hold great promise for treatment of autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Edward A Clark
- Department of Immunology, University of Washington , Seattle, WA , USA
| |
Collapse
|
26
|
Na K, Yoo HS, Zhang YX, Choi MS, Lee K, Yi TG, Song SU, Jeon MS. Bone marrow-derived clonal mesenchymal stem cells inhibit ovalbumin-induced atopic dermatitis. Cell Death Dis 2014; 5:e1345. [PMID: 25032868 PMCID: PMC4123091 DOI: 10.1038/cddis.2014.299] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 05/20/2014] [Accepted: 06/11/2014] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) possess immunomodulatory activities, including suppression of T- and B-cell activation. However, their effects on atopic dermatitis (AD) have not yet been studied. Using an ovalbumin-induced AD mouse model, we investigated whether MSCs can be used as therapeutics in AD. We isolated both allogeneic and syngeneic clonal MSCs (cMSCs) from mouse bone marrow according to the subfractionation culturing method. Our cMSCs suppressed both T- and B-cell activation. T-cell proliferation and cytokine production, including interferon (IFN)-γ and interleukin (IL)-4, were suppressed by inhibition of transcription factors, such as T-bet, GATA-3, and c-Maf. Those transcription factors were nitric oxide dependent. Immunoglobulin E (IgE) suppression occurred through downregulation of AID and BLIMP-1, important regulators for isotype class switch and B-cell differentiation. The cMSCs were injected intravenously into ovalbumin-induced AD mouse model, and the therapeutic effects were analyzed. Injection of both allogeneic and syngeneic cMSCs in an AD mouse model inhibited cell infiltration in skin lesions and decreased the serum level of IgE. IL-4 expression was also suppressed by cMSCs in both the lymph node and skin. The cMSCs migrated to skin lesions and draining lymph nodes. Taken together, these data demonstrated that cMSCs, which suppressed T- and B-cell functions, can be used for the treatment of AD in mice.
Collapse
Affiliation(s)
- K Na
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea
| | - H S Yoo
- Department of Drug Development, Inha University School of Medicine, Incheon, Republic of Korea
| | - Y X Zhang
- Department of Molecular Biomedicine, Inha University School of Medicine, Incheon, Republic of Korea
| | - M-S Choi
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea
| | - K Lee
- Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea
| | - T G Yi
- 1] Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea [2] SCM Lifescience Co. Ltd, Incheon, Republic of Korea [3] Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon, Republic of Korea
| | - S U Song
- 1] Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea [2] Department of Drug Development, Inha University School of Medicine, Incheon, Republic of Korea [3] SCM Lifescience Co. Ltd, Incheon, Republic of Korea
| | - M-S Jeon
- 1] Translational Research Center, Inha University School of Medicine, Incheon, Republic of Korea [2] Department of Molecular Biomedicine, Inha University School of Medicine, Incheon, Republic of Korea [3] Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
27
|
Kim EY, Sturgill JL, Hait NC, Avni D, Valencia EC, Maceyka M, Lima S, Allegood J, Huang WC, Zhang S, Milstien S, Conrad D, Spiegel S. Role of sphingosine kinase 1 and sphingosine-1-phosphate in CD40 signaling and IgE class switching. FASEB J 2014; 28:4347-58. [PMID: 25002116 DOI: 10.1096/fj.14-251611] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The tumor necrosis factor (TNF) receptor family member CD40 plays an essential role in the activation of antigen-presenting cells, B cell maturation, and immunoglobulin (Ig) class switching critical for adaptive immunity. Although the bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) and the kinase that produces it, sphingosine kinase 1 (SphK1), have long been implicated in the actions of TNF mediated by engagement of TNFR1, nothing is yet known of their role in CD40-mediated events. We have now found that ligation of CD40 activates and translocates SphK1 to the plasma membrane, leading to generation of S1P. SphK1 inhibition in human tonsil B cells, as well as inhibition or deletion of SphK1 in mouse splenic B cells, significantly reduced CD40-mediated Ig class switching and plasma cell differentiation ex vivo. Optimal activation of downstream CD40 signaling pathways, including NF-κB, p38, and JNK, also required SphK1. In mice treated with a SphK1 inhibitor or in SphK1(-/-) mice, isotype switching to antigen-specific IgE was decreased in vivo by 70 and 55%, respectively. Our results indicate that SphK1 is important for CD40-mediated B cell activation and regulation of humoral responses and suggest that targeting SphK1 might be a useful therapeutic approach to control antigen-specific IgE production.
Collapse
Affiliation(s)
- Eugene Y Kim
- Department of Biochemistry and Molecular Biology
| | | | - Nitai C Hait
- Department of Biochemistry and Molecular Biology
| | - Dorit Avni
- Department of Biochemistry and Molecular Biology
| | | | | | | | | | | | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | |
Collapse
|
28
|
Genome-wide expression profiling of B lymphocytes reveals IL4R increase in allergic asthma. J Allergy Clin Immunol 2014; 134:972-5. [PMID: 24975796 DOI: 10.1016/j.jaci.2014.05.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/21/2014] [Accepted: 05/01/2014] [Indexed: 11/23/2022]
|
29
|
Neunkirchner A, Schmetterer KG, Pickl WF. Lymphocyte-based model systems for allergy research: a historic overview. Int Arch Allergy Immunol 2014; 163:259-91. [PMID: 24777172 PMCID: PMC7617143 DOI: 10.1159/000360163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During the last decades, a multitude of studies applying distinct in vitro and in vivo model systems have contributed greatly to our better understanding of the initiation and regulation of inflammatory processes leading to allergic diseases. Over the years, it has become evident that among lymphocytes, not only IgE-producing B cells and allergy-orchestrating CD4(+) helper cells but also cytotoxic CD8(+) T cells, γδ-T cells and innate lymphoid cells, as well as regulatory lymphocytes, might critically shape the immune response towards usually innocuous allergens. In this review, we provide a historic overview of pioneering work leading to the establishment of important lymphocyte-based model systems for allergy research. Moreover, we contrast the original findings with our currently more refined knowledge to appreciate the actual validity of the respective models and to reassess the conclusions obtained from them. Conflicting studies and interpretations are identified and discussed. The tables are intended to provide an easy overview of the field not only for scientists newly entering the field but also for the broader readership interested in updating their knowledge. Along those lines, herein we discuss in vitro and in vivo approaches to the investigation of lymphocyte effector cell activation, polarization and regulation, and describe depletion and adoptive transfer models along with gene knockout and transgenic (tg) methodologies. In addition, novel attempts to establish humanized T cell antigen receptor tg mouse models for allergy research are described and discussed.
Collapse
Affiliation(s)
- Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
30
|
Mast cells, basophils and B cell connection network. Mol Immunol 2014; 63:94-103. [PMID: 24671125 DOI: 10.1016/j.molimm.2014.02.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/25/2014] [Accepted: 02/25/2014] [Indexed: 12/27/2022]
Abstract
It has been proven that both resting and activated mast cells (MCs) and basophils are able to induce a significant increase in proliferation and survival of naïve and activated B cells, and their differentiation into antibody-producing cells. The immunological context in which this regulation occurs is of particular interest and the idea that these innate cells induce antibody class switching and production is increasingly gaining ground. This direct role of MCs and basophils in acquired immunity requires cell to cell contact as well as soluble factors and exosomes. Here, we review our current understanding of the interaction between B cells and MCs or basophils as well as the evidence supporting B lymphocyte-MC/basophil crosstalk in pathological settings. Furthermore, we underline the obscure aspects of this interaction that could serve as important starting points for future research in the field of MC and basophil biology in the peculiar context of the connection between innate and adaptive immunity.
Collapse
|
31
|
Eckl-Dorna J, Niederberger V. What is the source of serum allergen-specific IgE? Curr Allergy Asthma Rep 2013; 13:281-7. [PMID: 23585215 DOI: 10.1007/s11882-013-0348-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Immunoglobulin E (IgE), the key effector element in the induction and propagation of allergic diseases, is the least abundant antibody class. In allergic patients, class switch recombination to IgE in B cells is induced by allergen contact in conjunction with T cell interaction and a Th2 cytokine environment. With regard to future therapeutic approaches, the sites of IgE production in human subjects and the nature and characteristics of IgE-producing cells are of great interest. In this context, it has been shown that allergen-specific IgE levels can be boosted by contact with allergens via the respiratory mucosa of the nose. Also, it has been proposed that allergy effector organs (e.g., the nasal mucosa and the lung) may be important sites of IgE production in allergic patients. IgE-producing cells have also been found in the blood, but their numbers are extremely low. Transfer of specific sensitization during bone marrow transplantation indicates the presence of IgE-producing B memory cells or plasma cells also in the bone marrow. This review summarizes data on the induction of IgE production, IgE memory and the sites of IgE production in human allergic patients.
Collapse
Affiliation(s)
- Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, General Hospital Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | |
Collapse
|
32
|
Nedbal J, Hobson PS, Fear DJ, Heintzmann R, Gould HJ. Comprehensive FISH probe design tool applied to imaging human immunoglobulin class switch recombination. PLoS One 2012; 7:e51675. [PMID: 23272136 PMCID: PMC3522715 DOI: 10.1371/journal.pone.0051675] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/02/2012] [Indexed: 12/22/2022] Open
Abstract
We present a web engine boosted fluorescence in-situ hybridization (webFISH) algorithm using a genome-wide sequence similarity search to design target-specific single-copy and repetitive DNA FISH probes. The webFISH algorithm featuring a user-friendly interface (http://www.webfish2.org/) maximizes the coverage of the examined sequences with FISH probes by considering locally repetitive sequences absent from the remainder of the genome. The highly repetitive human immunoglobulin heavy chain sequence was analyzed using webFISH to design three sets of FISH probes. These allowed direct simultaneous detection of class switch recombination in both immunoglobulin-heavy chain alleles in single cells from a population of cultured primary B cells. It directly demonstrated asynchrony of the class switch recombination in the two alleles in structurally preserved nuclei while permitting parallel readout of protein expression by immunofluorescence staining. This novel technique offers the possibility of gaining unprecedented insight into the molecular mechanisms involved in class switch recombination.
Collapse
Affiliation(s)
- Jakub Nedbal
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, United Kingdom
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Guy’s Hospital, London, United Kingdom
- * E-mail: (JN); (HJG)
| | - Philip S. Hobson
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, United Kingdom
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Guy’s Hospital, London, United Kingdom
| | - David J. Fear
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Guy’s Hospital, London, United Kingdom
- Division of Asthma, Allergy and Lung Biology, Guy’s Hospital, London, United Kingdom
| | - Rainer Heintzmann
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, United Kingdom
- Institute of Photonics Technology, Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University, Jena, Germany
| | - Hannah J Gould
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, United Kingdom
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Guy’s Hospital, London, United Kingdom
- * E-mail: (JN); (HJG)
| |
Collapse
|
33
|
|
34
|
|
35
|
May RD, Monk PD, Cohen ES, Manuel D, Dempsey F, Davis NHE, Dodd AJ, Corkill DJ, Woods J, Joberty-Candotti C, Conroy LA, Koentgen F, Martin EC, Wilson R, Brennan N, Powell J, Anderson IK. Preclinical development of CAT-354, an IL-13 neutralizing antibody, for the treatment of severe uncontrolled asthma. Br J Pharmacol 2012; 166:177-93. [PMID: 21895629 PMCID: PMC3415647 DOI: 10.1111/j.1476-5381.2011.01659.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE IL-13 is a pleiotropic Th2 cytokine considered likely to play a pivotal role in asthma. Here we describe the preclinical in vitro and in vivo characterization of CAT-354, an IL-13-neutralizing IgG4 monoclonal antibody (mAb), currently in clinical development. EXPERIMENTAL APPROACH In vitro the potency, specificity and species selectivity of CAT-354 was assayed in TF-1 cells, human umbilical vein endothelial cells and HDLM-2 cells. The ability of CAT-354 to modulate disease-relevant mechanisms was tested in human cells measuring bronchial smooth muscle calcium flux induced by histamine, eotaxin generation by normal lung fibroblasts, CD23 upregulation in peripheral blood mononuclear cells and IgE production by B cells. In vivo CAT-354 was tested on human IL-13-induced air pouch inflammation in mice, ovalbumin-sensitization and challenge in IL-13 humanized mice and antigen challenge in cynomolgus monkeys. KEY RESULTS CAT-354 has a 165 pM affinity for human IL-13 and functionally neutralized human, human variant associated with asthma and atopy (R130Q) and cynomolgus monkey, but not mouse, IL-13. CAT-354 did not neutralize human IL-4. In vitro CAT-354 functionally inhibited IL-13-induced eotaxin production, an analogue of smooth muscle airways hyperresponsiveness, CD23 upregulation and IgE production. In vivo in humanized mouse and cynomolgus monkey antigen challenge models CAT-354 inhibited airways hyperresponsiveness and bronchoalveolar lavage eosinophilia. CONCLUSIONS AND IMPLICATIONS CAT-354 is a potent and selective IL-13-neutralizing IgG4 mAb. The preclinical data presented here support the trialling of this mAb in patients with moderate to severe uncontrolled asthma.
Collapse
Affiliation(s)
- R D May
- MedImmune, Abington, Cambridge, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
n-3 Fatty acids inhibit transcription of human IL-13: implications for development of T helper type 2 immune responses. Br J Nutr 2012; 109:990-1000. [PMID: 22849952 DOI: 10.1017/s0007114512002917] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Fish oil supplementation during pregnancy has been associated with lower levels of cord blood IL-13, suggesting that the administration of n-3 fatty acids may attenuate the development of allergic disease. The present study aimed to investigate the mechanism by which n-3 fatty acid administration influences the production of IL-13. Pregnant BALB/c mice were fed nutritionally complete high-fat diets (15 %, w/w) with an n-3 fatty acid-enriched (DHA 1 %, w/w) or control diet (0 % DHA) immediately following delivery. Pups were exposed during suckling and weaned to the maternal diet for the remainder of the study. The production of IL-13, IL-4, IL-10 and interferon-γ from the splenocytes of ovalbumin (ova)-sensitised animals was assessed following in vitro ova stimulation or unstimulated conditions. Human T helper type 2 (Th2) cells were mitogen-stimulated in the presence or absence of DHA (10 μM) and assessed for IL-13 and IL-4 expression using intracellular flow cytometry. The influence on transcriptional activation was studied using a human IL-13 promoter reporter construct and electromobility shift assay. Ova-activated splenocytes from DHA-fed mice produced less IL-13 (57.2 (se 21.7) pg/ml) and IL-4 (7.33 (SE 3.4) pg/ml) compared with cells from the animals fed the control diet (161.5 (SE 45.0), P< 0.05; 33.2 (SE 11.8), P< 0.05). In vitro, DHA inhibited the expression of IL-13 protein from human Th2 cells as well as transcriptional activation and binding of the transcription factors cyclic AMP response element binding and activating transcription factor 2 to the human IL-13 promoter. These data indicate the potential of n-3 fatty acids to attenuate IL-13 expression, and suggest that they may subsequently reduce allergic sensitisation and the development of allergic disease.
Collapse
|
37
|
Jabara HH, McDonald DR, Janssen E, Massaad MJ, Ramesh N, Borzutzky A, Rauter I, Benson H, Schneider L, Baxi S, Recher M, Notarangelo LD, Wakim R, Dbaibo G, Dasouki M, Al-Herz W, Barlan I, Baris S, Kutukculer N, Ochs HD, Plebani A, Kanariou M, Lefranc G, Reisli I, Fitzgerald KA, Golenbock D, Manis J, Keles S, Ceja R, Chatila TA, Geha RS. DOCK8 functions as an adaptor that links TLR-MyD88 signaling to B cell activation. Nat Immunol 2012; 13:612-20. [PMID: 22581261 PMCID: PMC3362684 DOI: 10.1038/ni.2305] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 04/11/2012] [Indexed: 12/13/2022]
Abstract
The adaptors DOCK8 and MyD88 have been linked to serological memory. Here we report that DOCK8-deficient patients had impaired antibody responses and considerably fewer CD27(+) memory B cells. B cell proliferation and immunoglobulin production driven by Toll-like receptor 9 (TLR9) were considerably lower in DOCK8-deficient B cells, but those driven by the costimulatory molecule CD40 were not. In contrast, TLR9-driven expression of AICDA (which encodes the cytidine deaminase AID), the immunoglobulin receptor CD23 and the costimulatory molecule CD86 and activation of the transcription factor NF-κB, the kinase p38 and the GTPase Rac1 were intact. DOCK8 associated constitutively with MyD88 and the tyrosine kinase Pyk2 in normal B cells. After ligation of TLR9, DOCK8 became tyrosine-phosphorylated by Pyk2, bound the Src-family kinase Lyn and linked TLR9 to a Src-kinase Syk-transcription factor STAT3 cascade essential for TLR9-driven B cell proliferation and differentiation. Thus, DOCK8 functions as an adaptor in a TLR9-MyD88 signaling pathway in B cells.
Collapse
Affiliation(s)
- Haifa H Jabara
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wallemacq H, Bedoret D, Pujol J, Desmet C, Drion PV, Farnir F, Mainil J, Lekeux P, Bureau F, Fiévez L. CD40 triggering induces strong cytotoxic T lymphocyte responses to heat-killed Staphylococcus aureus immunization in mice: A new vaccine strategy for staphylococcal mastitis. Vaccine 2012; 30:2116-24. [DOI: 10.1016/j.vaccine.2012.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 12/21/2011] [Accepted: 01/15/2012] [Indexed: 11/27/2022]
|
39
|
Godava M, Kopriva F, Bohmova J, Vodicka R, Dusek L, Cvanova M, Muzik J, Markova M, Schneiderova E, Vrtel R. Association of STAT6 and ADAM33 single nucleotide polymorphisms with asthma bronchiale and IgE level and its possible epigenetic background. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 156:236-47. [PMID: 22660217 DOI: 10.5507/bp.2012.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 01/10/2010] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND ADAM33 and STAT6 belong to the candidate genes that have been commonly associated with asthma, bronchial hyperresponsiveness or IgE levels. Our objective was to assess the association of 11 SNPs of the ADAM33 and 6 of the STAT6 and their haplotypes with IgE levels and asthma. We also evaluated the possible role of parental origin of haplotypes on IgE levels. METHODS We enrolled 109 children with asthma and 45 healthy controls. Genotyping was performed by TaqMan probes and confirmed by sequencing. Haplotype construction was based on the knowledge of parental genotypes and also inferred by using the EM algorithm and Bayes' theorem. RESULTS None of the SNPs were associated with elevated IgE level or asthma. We found that the most frequent STAT6 haplotype ATTCAA (built from rs324012, rs324011, rs841718, rs3024974, rs3024974, rs4559 SNPs, respectively) was associated with elevated total IgE levels (P=0.01) and this haplotype was predominantly transmitted paternally (P<0.001). We compared our results with those of studies performed on German and Australian Caucasian populations and found that rs324011, rs3024974 and rs4559 SNPs in STAT6 should have a major effect on IgE levels. Therefore, we suggest the TCA haplotype alone (built from rs324011, rs3024974 and rs4559 SNPs, respectively) in STAT6 is associated with total IgE elevation. CONCLUSIONS The influence of paternal origin of the STAT6 haplotype on IgE levels is surprising but the exact role of possible paternal imprinting in STAT6 regulation should be investigated and confirmed in future studies.
Collapse
Affiliation(s)
- Marek Godava
- Department of Medical Genetics and Fetal Medicine, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chu SY, Horton HM, Pong E, Leung IWL, Chen H, Nguyen DH, Bautista C, Muchhal US, Bernett MJ, Moore GL, Szymkowski DE, Desjarlais JR. Reduction of total IgE by targeted coengagement of IgE B-cell receptor and FcγRIIb with Fc-engineered antibody. J Allergy Clin Immunol 2012; 129:1102-15. [PMID: 22257644 DOI: 10.1016/j.jaci.2011.11.029] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 11/16/2011] [Accepted: 11/18/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND Sequestration of IgE to prevent its binding to high-affinity IgE receptor FcεRI on basophils and mast cells is an effective therapy for allergic asthma. IgE production requires differentiation of activated IgE(+) B cells into plasma cells upon allergen sensitization. B-cell receptor signaling is suppressed by the inhibitory IgG Fc receptor FcγRIIb; therefore, we reasoned that a therapeutic antibody that coengages FcγRIIb and IgE B-cell receptor would not only sequester IgE but also suppress its production by blocking IgE(+) B-cell activation and differentiation to IgE-secreting plasma cells. OBJECTIVE To explore the effects of IgE sequestration versus IgE suppression by comparing omalizumab to FcγRIIb-optimized anti-IgE antibodies in humanized mouse models of immunoglobulin production. METHODS By using a murine anti-IgE antibody as a template, we humanized, increased IgE binding, and modified its Fc domain to increase affinity for FcγRIIb. We next compared effects of this antibody (XmAb7195) versus omalizumab on the secretion of IgE and other isotypes in human PBMC cultures and in PBMC-engrafted severe combined immunodeficiency mice. RESULTS Relative to omalizumab, XmAb7195 has a 5-fold higher affinity for human IgE and more than 400-fold higher affinity for FcγRIIb. In addition to sequestering soluble IgE, XmAb7195 inhibited plasma cell differentiation and consequent human IgE production through coengagement of IgE B-cell receptor with FcγRIIb. In PBMC-engrafted mice, XmAb7195 reduced total human IgE (but not IgG or IgM) levels by up to 40-fold relative to omalizumab. CONCLUSION XmAb7195 acts by IgE sequestration coupled with an FcγRIIb-mediated inhibitory mechanism to suppress the formation of IgE-secreting plasma cells and reduce both free and total IgE levels.
Collapse
|
41
|
|
42
|
Esen M, Forster J, Ajua A, Spänkuch I, Paparoupa M, Mordmüller B, Kremsner PG. Effect of IL-15 on IgG versus IgE antibody-secreting cells in vitro. J Immunol Methods 2011; 375:7-13. [PMID: 21945396 DOI: 10.1016/j.jim.2011.08.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 08/24/2011] [Accepted: 08/24/2011] [Indexed: 10/17/2022]
Abstract
Immunoglobulin E (IgE) antibodies are major contributors to the pathology of atopic and allergic diseases as well as to immune response to helminth infections. Development of an adequate immunoglobulin G (IgG) immune response against infectious agents and vaccine antigens is considered in most cases as crucial for protection from disease. In vivo and in vitro production of IgE and IgG depends on cytokines and other soluble factors. Recently it has been shown that IgG antibody secreting cells (ASCs) can be generated by in vitro maturation of blood cells with Interleukin- (IL-)15 and CpG DNA or other stimulation cocktails, while IgE-ASCs develop upon cultivation with anti-CD40 and IL-4. In the present study we employed an enzyme linked immunospot assay (ELISPOT) to assess the capacity of individuals to develop into either IgE-ASCs or IgG-ASCs upon stimulation with different combinations of stimulation cocktails in order to investigate the influence of cytokines that are dysregulated in IgE-mediated immune reactions on ASC generation. Furthermore, we modified the method to assess IgG- and IgE-ASCs specific for two model antigens causing allergic rhinitis in humans. We demonstrate that IL-15, which is important for development of IgG-ASCs, decreases the number of IgE-ASCs when added to media commonly used for in vitro development of IgE-ASCs. We show that our method is suitable for the detection of specific and non-specific IgE-ASCs and IgG-ASCs and allows the investigation of the interplay between IgG-ASCs and IgE-ASCs in different populations.
Collapse
Affiliation(s)
- Meral Esen
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstraße 27, D-72074 Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
43
|
HOIL-1L interacting protein (HOIP) is essential for CD40 signaling. PLoS One 2011; 6:e23061. [PMID: 21829693 PMCID: PMC3148254 DOI: 10.1371/journal.pone.0023061] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 07/12/2011] [Indexed: 11/19/2022] Open
Abstract
CD40 is a cell surface receptor important in the activation of antigen-presenting cells during immune responses. In macrophages and dendritic cells, engagement of CD40 by its ligand CD154 provides signals critical for anti-microbial and T cell-mediated immune responses, respectively. In B cells, CD40 signaling has a major role in regulating cell proliferation, antibody production, and memory B cell development. CD40 engagement results in the formation of a receptor-associated complex that mediates activation of NF-κB, stress-activated protein kinases, and other signaling molecules. However, the mechanisms that link CD40 to these signaling events have been only partially characterized. Known components of the CD40 signaling complex include members of the TNF receptor-associated factor (TRAF) family of proteins. We previously showed that the TRAF family member TRAF2 mediates recruitment of HOIL-1L-interacting protein (HOIP) to the cytoplasmic domain of CD40, suggesting that HOIP has a role in the CD40 signaling pathway. To determine the role of HOIP in CD40 signaling, we used somatic cell gene targeting to generate mouse B cell lines deficient in HOIP. We found that the CD40-induced upregulation of CD80 and activation of germline immunoglobulin epsilon transcription were defective in HOIP-deficient cells. We also found that the CD40-mediated activation of NF-κB and c-Jun kinase was impaired. Recruitment of IκB kinase proteins to the CD40 signaling complex was undetectable in HOIP-deficient cells, potentially explaining the defect in NF-κB activation. Restoration of HOIP expression reversed the defects in cellular activation and signaling. These results reveal HOIP as a key component of the CD40 signaling pathway.
Collapse
|
44
|
Joks R, Durkin HG. Non-antibiotic properties of tetracyclines as anti-allergy and asthma drugs. Pharmacol Res 2011; 64:602-9. [PMID: 21501686 DOI: 10.1016/j.phrs.2011.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
All available therapies for human allergic disease target IgE mediated pathologic responses after IgE has been produced. We are developing tetracyclines as anti-allergy drugs to prevent IgE production, based on our findings that minocycline or doxycycline treatment of allergic asthmatic humans significantly improves their asthma symptoms, reduces their oral steroid requirements, and strongly suppresses their ongoing IgE responses (ELISA, mast cell mediated cutaneous late phase responses); the tetracyclines also strongly suppress peak IgE responses of BPO-KLH sensitized mice (ELISPOT assay, ELISA, skin tests). The antibiotic activity of the tetracyclines is not required for suppression of IgE responses; inclusion of minocycline or doxycycline in sterile culture prevents anti-CD40/IL-4 mediated induction of memory IgE responses by PBMC of allergic asthmatic patients (ELISA), and induction of specific memory IgE responses by spleen cells of BPO-KLH sensitized mice (ELISPOT assay, ELISA). The tetracyclines affect an epsilon specific pathway because IgM, IgG and IgA responses did not decrease. Further, in humans, DTH responses to recall antigens did not decrease. In related studies, we found that two distinct T cell subsets: CD4+CD60 negative and CD8+CD60+ (CD60 is a ganglioside) (humans) and CD4+ Asialo GM1 ganglioside negative and CD8+Asialo GM1 ganglioside+ (mice), both are required for induction of memory IgE responses. Phosphorylated (phos) p38 MAP kinase, but not phos ERK or phos JNK expression by CD4+ and CD8+, including CD8+CD60+, T cells is increased in allergic asthmatic humans, as is IL-4 and IL-10 production. The tetracyclines appear to target T cell pathways to induce suppression of IgE responses because they suppress phos p38 MAP kinase expression by both CD4+ and CD8+, including CD8+CD60+, T cell subsets, and IL-4 and IL-10, while upregulating IL-2 and IFN gamma, and suppressing IgE responses. Our finding that tetracyclines do not require antibiotic activity to suppress IgE responses opens the door to development of new tetracycline-based and other therapeutics for human allergic disease.
Collapse
Affiliation(s)
- Rauno Joks
- Department of Medicine, Center for Allergy and Asthma Research at SUNY Downstate, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA.
| | | |
Collapse
|
45
|
Tuning of CD40–CD154 Interactions in Human B-Lymphocyte Activation: A Broad Array of In Vitro Models for a Complex In Vivo Situation. Arch Immunol Ther Exp (Warsz) 2011; 59:25-40. [DOI: 10.1007/s00005-010-0108-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 08/19/2010] [Indexed: 12/13/2022]
|
46
|
Lisowska KA, Debska-Slizien A, Radzka M, Witkowski JM, Rutkowski B, Bryl E. Recombinant human erythropoietin treatment of chronic renal failure patients normalizes altered phenotype and proliferation of CD4-positive T lymphocytes. Artif Organs 2010; 34:E77-84. [PMID: 20447038 DOI: 10.1111/j.1525-1594.2009.00942.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Patients with chronic renal failure (CRF) receive recombinant human erythropoietin (rhEPO) for the correction of anemia. However, rhEPO also has an immunomodulatory effect. Detailed changes of phenotype and function of CD4(+) T lymphocytes in CRF patients receiving rhEPO have not been reported yet; their study may bring insight into understanding of this immunomodulatory action of rhEPO. Two groups of CRF patients were included into the study: those treated; and those not receiving rhEPO. The expression of activation markers on CD4(+) lymphocytes was measured with flow cytometry, both ex vivo and in vitro. The kinetics of CD4(+) T lymphocytes proliferation was calculated using a dividing cells tracing method and numerical approach. Significantly higher percentages of CD4(+)CD95(+), CD4(+)HLA-DR(+) cells, and lower percentages of CD4(+)CD69(+) and CD4(+)CD28(+) cells were observed in both rhEPO-treated and untreated patients when compared with healthy controls. Changes in the proportions of CD4(+)CD28(+) and CD4(+)HLA-DR(+) subpopulations were dependent on the type of rhEPO, being more pronounced for rhEPObeta. CD4(+) lymphocytes from untreated patients exhibited decreased expression of CD28 and CD69 after stimulation in vitro, whereas the expression of these antigens on lymphocytes of rhEPO-treated patients was similar to that observed in healthy controls. Fewer CD4(+)CD28(+) T lymphocytes of untreated patients proliferated in vitro; these cells had longer G0-->G1 time, which negatively correlated with surface expression of CD28. Our study confirms that rhEPO treatment normalizes activation parameters of CD4(+) T lymphocytes and their proliferative capacity, which could explain earlier described immunomodulatory effects of rhEPO in patients suffering from CRF.
Collapse
Affiliation(s)
- Katarzyna A Lisowska
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | | | | | | |
Collapse
|
47
|
Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev 2009; 229:152-72. [PMID: 19426221 DOI: 10.1111/j.1600-065x.2009.00782.x] [Citation(s) in RCA: 1119] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SUMMARY During the generation of a successful adaptive immune response, multiple molecular signals are required. A primary signal is the binding of cognate antigen to an antigen receptor expressed by T and B lymphocytes. Multiple secondary signals involve the engagement of costimulatory molecules expressed by T and B lymphocytes with their respective ligands. Because of its essential role in immunity, one of the best characterized of the costimulatory molecules is the receptor CD40. This receptor, a member of the tumor necrosis factor receptor family, is expressed by B cells, professional antigen-presenting cells, as well as non-immune cells and tumors. CD40 binds its ligand CD40L, which is transiently expressed on T cells and other non-immune cells under inflammatory conditions. A wide spectrum of molecular and cellular processes is regulated by CD40 engagement including the initiation and progression of cellular and humoral adaptive immunity. In this review, we describe the downstream signaling pathways initiated by CD40 and overview how CD40 engagement or antagonism modulates humoral and cellular immunity. Lastly, we discuss the role of CD40 as a target in harnessing anti-tumor immunity. This review underscores the essential role CD40 plays in adaptive immunity.
Collapse
Affiliation(s)
- Raul Elgueta
- Department of Microbiology and Immunology, Dartmouth Medical School and The Norris Cotton Cancer Center, Lebanon, NH 03756, USA
| | | | | | | | | | | |
Collapse
|
48
|
Heine G, Dahten A, Hilt K, Ernst D, Milovanovic M, Hartmann B, Worm M. Liver X Receptors Control IgE Expression in B Cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:5276-82. [DOI: 10.4049/jimmunol.0801804] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
49
|
Malerba M, Mennuni L, Piepoli T, Caselli G, Makovec F, Rovati LC, D' Amato M, Ferrari F. Andolast acts at different cellular levels to inhibit immunoglobulin E synthesis. Int J Immunopathol Pharmacol 2009; 22:85-94. [PMID: 19309555 DOI: 10.1177/039463200902200110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The anti-asthmatic agent andolast is thought to inhibit the release of allergic mediators, but its mechanism of action is not fully understood. We investigated whether the compound inhibits immunoglobulin E (IgE) synthesis and tested the hypothesis that andolast affects immunoglobulin class switching. Interleukin (IL)-4 and the interaction of CD40 expressed on B cells with its ligand on T cells are necessary for IgE synthesis. Thus, peripheral blood mononuclear cells (PBMCs) from 40 asthmatic, 16 non-asthmatic allergic, and 9 normal donors were stimulated with IL-4 and/or anti-CD40 antibody. T cells from 9 additional allergic donors were activated with anti-CD3/CD28 antibodies to express IL-4 mRNA. After incubation in the absence or presence of test compounds, immunoglobulin concentrations were measured by enzyme immunoassay, and mRNA levels were analyzed by RT-PCR. Andolast significantly inhibited IgE synthesis by stimulated PBMCs from both asthma patients and combined allergic/normal donors. In mechanistic studies, andolast was found to act at different cellular levels. Firstly, it reduced by about 45 percent (p<0.05) the levels of IL-4 mRNA in T cells stimulated with anti-CD3/CD28. Secondly, andolast reduced by about 36 percent (p<0.05) the expression of epsilon germline transcripts in PBMCs stimulated with IL-4/anti-CD40. Thirdly, the effect of andolast on immunoglobulin synthesis was selective in that the production of IgG4 antibodies was not significantly inhibited. Our findings, while supporting the evidence that andolast is effective for the treatment of asthma, provide new insights into its mechanism of action.
Collapse
Affiliation(s)
- M Malerba
- Department of Internal Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Dultz G, Dittmar M, Kahaly GJ. Genetik der Schilddrüsenautoimmunität – Update und klinische Relevanz. ACTA ACUST UNITED AC 2009; 104:210-9. [DOI: 10.1007/s00063-009-1034-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 01/17/2009] [Indexed: 12/12/2022]
|