1
|
Moquist PN, Zhang X, Leiske CI, Eng-Duncan NML, Zeng W, Bindman NA, Wo SW, Wong A, Henderson CM, Crowder K, Lyon R, Doronina SO, Senter PD, Neff-LaFord HD, Sussman D, Gardai SJ, Levengood MR. Reversible Chemical Modification of Antibody Effector Function Mitigates Unwanted Systemic Immune Activation. Bioconjug Chem 2024; 35:855-866. [PMID: 38789102 PMCID: PMC11191404 DOI: 10.1021/acs.bioconjchem.4c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.
Collapse
Affiliation(s)
- Philip N. Moquist
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Xinqun Zhang
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Chris I. Leiske
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | | | - Weiping Zeng
- ADC
In Vivo Pharmacology, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Noah A. Bindman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Serena W. Wo
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Abbie Wong
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Clark M. Henderson
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Karalyne Crowder
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Robert Lyon
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Svetlana O. Doronina
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Peter D. Senter
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Haley D. Neff-LaFord
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Django Sussman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Shyra J. Gardai
- Immunology, Pfizer,
Inc., 21823 30th Dr.
SE, Bothell, Washington 98021, United States
| | - Matthew R. Levengood
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| |
Collapse
|
2
|
Zhou Y, Richmond A, Yan C. Harnessing the potential of CD40 agonism in cancer therapy. Cytokine Growth Factor Rev 2024; 75:40-56. [PMID: 38102001 PMCID: PMC10922420 DOI: 10.1016/j.cytogfr.2023.11.002] [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: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily of receptors expressed on a variety of cell types. The CD40-CD40L interaction gives rise to many immune events, including the licensing of dendritic cells to activate CD8+ effector T cells, as well as the facilitation of B cell activation, proliferation, and differentiation. In malignant cells, the expression of CD40 varies among cancer types, mediating cellular proliferation, apoptosis, survival and the secretion of cytokines and chemokines. Agonistic human anti-CD40 antibodies are emerging as an option for cancer treatment, and early-phase clinical trials explored its monotherapy or combination with radiotherapy, chemotherapy, immune checkpoint blockade, and other immunomodulatory approaches. In this review, we present the current understanding of the mechanism of action for CD40, along with results from the clinical development of agonistic human CD40 antibodies in cancer treatment (selicrelumab, CDX-1140, APX005M, mitazalimab, 2141-V11, SEA-CD40, LVGN7409, and bispecific antibodies). This review also examines the safety profile of CD40 agonists in both preclinical and clinical settings, highlighting optimized dosage levels, potential adverse effects, and strategies to mitigate them.
Collapse
Affiliation(s)
- Yang Zhou
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Chi Yan
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA.
| |
Collapse
|
3
|
Staniek J, Kalina T, Andrieux G, Boerries M, Janowska I, Fuentes M, Díez P, Bakardjieva M, Stancikova J, Raabe J, Neumann J, Schwenk S, Arpesella L, Stuchly J, Benes V, García Valiente R, Fernández García J, Carsetti R, Piano Mortari E, Catala A, de la Calle O, Sogkas G, Neven B, Rieux-Laucat F, Magerus A, Neth O, Olbrich P, Voll RE, Alsina L, Allende LM, Gonzalez-Granado LI, Böhler C, Thiel J, Venhoff N, Lorenzetti R, Warnatz K, Unger S, Seidl M, Mielenz D, Schneider P, Ehl S, Rensing-Ehl A, Smulski CR, Rizzi M. Non-apoptotic FAS signaling controls mTOR activation and extrafollicular maturation in human B cells. Sci Immunol 2024; 9:eadj5948. [PMID: 38215192 DOI: 10.1126/sciimmunol.adj5948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/08/2023] [Indexed: 01/14/2024]
Abstract
Defective FAS (CD95/Apo-1/TNFRSF6) signaling causes autoimmune lymphoproliferative syndrome (ALPS). Hypergammaglobulinemia is a common feature in ALPS with FAS mutations (ALPS-FAS), but paradoxically, fewer conventional memory cells differentiate from FAS-expressing germinal center (GC) B cells. Resistance to FAS-induced apoptosis does not explain this phenotype. We tested the hypothesis that defective non-apoptotic FAS signaling may contribute to impaired B cell differentiation in ALPS. We analyzed secondary lymphoid organs of patients with ALPS-FAS and found low numbers of memory B cells, fewer GC B cells, and an expanded extrafollicular (EF) B cell response. Enhanced mTOR activity has been shown to favor EF versus GC fate decision, and we found enhanced PI3K/mTOR and BCR signaling in ALPS-FAS splenic B cells. Modeling initial T-dependent B cell activation with CD40L in vitro, we showed that FAS competent cells with transient FAS ligation showed specifically decreased mTOR axis activation without apoptosis. Mechanistically, transient FAS engagement with involvement of caspase-8 induced nuclear exclusion of PTEN, leading to mTOR inhibition. In addition, FASL-dependent PTEN nuclear exclusion and mTOR modulation were defective in patients with ALPS-FAS. In the early phase of activation, FAS stimulation promoted expression of genes related to GC initiation at the expense of processes related to the EF response. Hence, our data suggest that non-apoptotic FAS signaling acts as molecular switch between EF versus GC fate decisions via regulation of the mTOR axis and transcription. The defect of this modulatory circuit may explain the observed hypergammaglobulinemia and low memory B cell numbers in ALPS.
Collapse
Affiliation(s)
- Julian Staniek
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Tomas Kalina
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iga Janowska
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, Proteomics Unit, CIBERONC CB16/12/00400, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Paula Díez
- Department of Medicine and General Cytometry Service-Nucleus, Proteomics Unit, CIBERONC CB16/12/00400, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), Universidad de Salamanca, Salamanca, Spain
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Marina Bakardjieva
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jitka Stancikova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Raabe
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julika Neumann
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Schwenk
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Leonardo Arpesella
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Stuchly
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rodrigo García Valiente
- Department of Medicine and General Cytometry Service-Nucleus, Proteomics Unit, CIBERONC CB16/12/00400, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Jonatan Fernández García
- Department of Medicine and General Cytometry Service-Nucleus, Proteomics Unit, CIBERONC CB16/12/00400, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Rita Carsetti
- B Cell Unit, Immunology Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Eva Piano Mortari
- B Cell Unit, Immunology Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Albert Catala
- Department of Hematology, Institut de Recerca Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Oscar de la Calle
- Immunology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Bénédicte Neven
- Pediatric Hematology-Immunology and Rheumatology Department, University Hospital Necker-Enfants Malades, Paris, France
| | - Frédéric Rieux-Laucat
- Université de Paris, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Aude Magerus
- Université de Paris, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Olaf Neth
- Department of Paediatric Infectious Diseases, Rheumatology and Immunology, Hospital Universitario Virgen del Rocio (HUVR), Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Sevilla, Spain
| | - Peter Olbrich
- Department of Paediatric Infectious Diseases, Rheumatology and Immunology, Hospital Universitario Virgen del Rocio (HUVR), Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla/CSIC, Red de Investigación Traslacional en Infectología Pediátrica RITIP, Sevilla, Spain
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laia Alsina
- Department of Hematology, Institut de Recerca Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Department of Pediatric Allergy and Clinical Immunology, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Luis M Allende
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Luis I Gonzalez-Granado
- Primary Immunodeficiencies Unit, Department of Pediatrics, Research Institute Hospital 12 Octubre (i+12), Madrid, Spain
- School of Medicine, Complutense University, Madrid, Spain
| | - Chiara Böhler
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jens Thiel
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Rheumatology and Clinical Immunology, Medical University Graz, Graz, Austria
| | - Nils Venhoff
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Raquel Lorenzetti
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Rheumatology and Clinical Immunology, Medical University Graz, Graz, Austria
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Susanne Unger
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Seidl
- Department of Pathology, University Medical Center Freiburg, Freiburg, Germany
- Institute of Pathology, Heinrich-Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus Fiebiger Zentrum, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Pascal Schneider
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Stephan Ehl
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Anne Rensing-Ehl
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cristian Roberto Smulski
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - Marta Rizzi
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Pioli KT, Pioli PD. Thymus antibody-secreting cells: once forgotten but not lost. Front Immunol 2023; 14:1170438. [PMID: 37122712 PMCID: PMC10130419 DOI: 10.3389/fimmu.2023.1170438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Antibody-secreting cells are essential contributors to the humoral response. This is due to multiple factors which include: 1) the ability to secrete thousands of antibodies per second, 2) the ability to regulate the immune response and 3) the potential to be long-lived. Not surprisingly, these cells can be found in numerous sites within the body which include organs that directly interface with potential pathogens (e.g., gut) and others that provide long-term survival niches (e.g., bone marrow). Even though antibody-secreting cells were first identified in the thymus of both humans and rodents in the 1960s, if not earlier, only recently has this population begun to be extensively investigated. In this article, we provide an update regarding the current breath of knowledge pertaining to thymus antibody-secreting cells and discuss the potential roles of these cells and their impact on health.
Collapse
|
5
|
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: 2] [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
|
6
|
van Asten SD, Unger PP, Marsman C, Bliss S, Jorritsma T, Thielens NM, van Ham SM, Spaapen RM. Soluble FAS Ligand Enhances Suboptimal CD40L/IL-21-Mediated Human Memory B Cell Differentiation into Antibody-Secreting Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:449-458. [PMID: 34215657 DOI: 10.4049/jimmunol.2001390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/13/2021] [Indexed: 11/19/2022]
Abstract
Differentiation of Ag-specific B cells into class-switched, high-affinity, Ab-secreting cells provides protection against invading pathogens but is undesired when Abs target self-tissues in autoimmunity, beneficial non-self-blood transfusion products, or therapeutic proteins. Essential T cell factors have been uncovered that regulate T cell-dependent B cell differentiation. We performed a screen using a secreted protein library to identify novel factors that promote this process and may be used to combat undesired Ab formation. We tested the differentiating capacity of 756 secreted proteins on human naive or memory B cell differentiation in a setting with suboptimal T cell help in vitro (suboptimal CD40L and IL-21). High-throughput flow cytometry screening and validation revealed that type I IFNs and soluble FAS ligand (sFASL) induce plasmablast differentiation in memory B cells. Furthermore, sFASL induces robust secretion of IgG1 and IgG4 Abs, indicative of functional plasma cell differentiation. Our data suggest a mechanistic connection between elevated sFASL levels and the induction of autoreactive Abs, providing a potential therapeutic target in autoimmunity. Indeed, the modulators identified in this secretome screen are associated with systemic lupus erythematosus and may also be relevant in other autoimmune diseases and allergy.
Collapse
Affiliation(s)
- Saskia D van Asten
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter-Paul Unger
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Casper Marsman
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sophie Bliss
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tineke Jorritsma
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; .,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Enhanced Expression of miR-181b in B Cells of CLL Improves the Anti-Tumor Cytotoxic T Cell Response. Cancers (Basel) 2021; 13:cancers13020257. [PMID: 33445508 PMCID: PMC7826592 DOI: 10.3390/cancers13020257] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
The clinical progression of B cell chronic lymphocytic leukemia (CLL) is associated with immune cell dysfunction and a strong decrease of miR-181b-5p (miR-181b), promoting the death of CLL cells. Here we investigated whether the reduction of miR-181b impairs the immune response in CLL. We demonstrate that activated CD4+ T cells increase miR-181b expression in CLL through CD40-CD40L signaling, which enhances the maturation and activity of cytotoxic T cells and, consequently, the apoptotic response of CLL cells. The cytotoxic response is facilitated by a depletion of the anti-inflammatory cytokine interleukin 10, targeted by miR-181b. In vivo experiments in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice confirmed that miR-181b promotes the apoptotic death of CLL cells only when functional T cells are restored. Overall, our findings suggest that the reinstatement of miR-181b in CLL cells could be an exploitable adjuvant therapeutic option for the treatment of CLL.
Collapse
|
8
|
Sugihara E, Hashimoto N, Osuka S, Shimizu T, Ueno S, Okazaki S, Yaguchi T, Kawakami Y, Kosaki K, Sato TA, Okamoto S, Saya H. The Inhibitor of Apoptosis Protein Livin Confers Resistance to Fas-Mediated Immune Cytotoxicity in Refractory Lymphoma. Cancer Res 2020; 80:4439-4450. [PMID: 32928920 DOI: 10.1158/0008-5472.can-19-3993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 07/09/2020] [Accepted: 08/26/2020] [Indexed: 11/16/2022]
Abstract
Death receptor Fas-mediated apoptosis not only eliminates nonspecific and autoreactive B cells but also plays a major role in antitumor immunity. However, the possible mechanisms underlying impairment of Fas-mediated induction of apoptosis during lymphomagenesis remain unknown. In this study, we employed our developed syngeneic lymphoma model to demonstrate that downregulation of Fas is required for both lymphoma development and lymphoma cell survival to evade immune cytotoxicity. CD40 signal activation significantly restored Fas expression and thereby induced apoptosis after Fas ligand treatment in both mouse and human lymphoma cells. Nevertheless, certain human lymphoma cell lines were found to be resistant to Fas-mediated apoptosis, with Livin (melanoma inhibitor of apoptosis protein; ML-IAP) identified as a driver of such resistance. High expression of Livin and low expression of Fas were associated with poor prognosis in patients with aggressive non-Hodgkin's lymphoma. Livin expression was tightly driven by bromodomain and extraterminal (BET) proteins BRD4 and BRD2, suggesting that Livin expression is epigenetically regulated in refractory lymphoma cells to protect them from Fas-mediated apoptosis. Accordingly, the combination of CD40-mediated Fas restoration with targeting of the BET proteins-Livin axis may serve as a promising immunotherapeutic strategy for refractory B-cell lymphoma. SIGNIFICANCE: These findings yield insights into identifying risk factors in refractory lymphoma and provide a promising therapy for tumors resistant to Fas-mediated antitumor immunity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/20/4439/F1.large.jpg.
Collapse
Affiliation(s)
- Eiji Sugihara
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan. .,Research and Development Center for Precision Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Norisato Hashimoto
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Osuka
- Department of Neurosurgery, Wallace Tumor Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Takatsune Shimizu
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Pathophysiology, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Sayaka Ueno
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Section of Translational Research, Hyogo Cancer Center, Hyogo, Japan
| | - Shogo Okazaki
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Immunology, School of Medicine, International University of Health and Welfare, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Taka-Aki Sato
- Research and Development Center for Precision Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
9
|
Staniek J, Lorenzetti R, Heller B, Janowska I, Schneider P, Unger S, Warnatz K, Seidl M, Venhoff N, Thiel J, Smulski CR, Rizzi M. TRAIL-R1 and TRAIL-R2 Mediate TRAIL-Dependent Apoptosis in Activated Primary Human B Lymphocytes. Front Immunol 2019; 10:951. [PMID: 31114586 PMCID: PMC6503035 DOI: 10.3389/fimmu.2019.00951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/12/2019] [Indexed: 11/28/2022] Open
Abstract
The maintenance of B cell homeostasis requires a tight control of B cell generation, survival, activation, and maturation. In lymphocytes upon activation, increased sensitivity to apoptotic signals helps controlling differentiation and proliferation. The death receptor Fas is important in this context because genetic Fas mutations in humans lead to an autoimmune lymphoproliferative syndrome that is similar to lymphoproliferation observed in Fas-deficient mice. In contrast, the physiological role of TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs) in humans has been poorly studied so far. Indeed, most studies have focused on tumor cell lines and on mouse models whose results are difficult to transpose to primary human B cells. In the present work, the expression of apoptosis-inducing TRAIL-R1 and TRAIL-R2 and of the decoy receptors TRAIL-R3 and TRAIL-R4 was systematically studied in all developmental stages of peripheral B cells isolated from the blood and secondary lymphoid organs. Expression of TRAIL-Rs is modulated along development, with highest levels observed in germinal center B cells. In addition, T-dependent and T-independent signals elicited induction of TRAIL-Rs with distinct kinetics, which differed among B cell subpopulations: switched memory cells rapidly upregulated TRAIL-R1 and -2 upon activation while naïve B cells only reached similar expression levels at later time points in culture. Increased expression of TRAIL-R1 and -2 coincided with a caspase-3-dependent sensitivity to TRAIL-induced apoptosis in activated B cells but not in freshly isolated resting B cells. Finally, both TRAIL-R1 and TRAIL-R2 could signal actively and both contributed to TRAIL-induced apoptosis. In conclusion, this study provides a systematic analysis of the expression of TRAIL-Rs in human primary B cells and of their capacity to signal and induce apoptosis. This dataset forms a basis to further study and understand the dysregulation of TRAIL-Rs and TRAIL expression observed in autoimmune diseases. Additionally, it will be important to foresee potential bystander immunomodulation when TRAIL-R agonists are used in cancer treatment.
Collapse
Affiliation(s)
- Julian Staniek
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Raquel Lorenzetti
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Bianca Heller
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Iga Janowska
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Pascal Schneider
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Susanne Unger
- Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Maximilian Seidl
- Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Nils Venhoff
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Jens Thiel
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Cristian Roberto Smulski
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - Marta Rizzi
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Karnell JL, Rieder SA, Ettinger R, Kolbeck R. Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv Drug Deliv Rev 2019; 141:92-103. [PMID: 30552917 DOI: 10.1016/j.addr.2018.12.005] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/12/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
CD40 is a TNF receptor superfamily member expressed on both immune and non-immune cells. Interactions between B cell-expressed CD40 and its binding partner, CD40L, predominantly expressed on activated CD4+ T cells, play a critical role in promoting germinal center formation and the production of class-switched antibodies. Non-hematopoietic cells expressing CD40 can also engage CD40L and trigger a pro-inflammatory response. This article will highlight what is known about the biology of the CD40-CD40L axis in humans and describe the potential contribution of CD40 signaling on both hematopoietic and non-hematopoietic cells to autoimmune disease pathogenesis. Additionally, novel therapeutic approaches to target this pathway, currently being evaluated in clinical trials, are discussed.
Collapse
|
11
|
Congenital neutropenia and primary immunodeficiency diseases. Crit Rev Oncol Hematol 2019; 133:149-162. [DOI: 10.1016/j.critrevonc.2018.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
|
12
|
Yiwen Z, Shilin G, Yingshi C, Lishi S, Baohong L, Chao L, Linghua L, Ting P, Hui Z. Efficient generation of antigen-specific CTLs by the BAFF-activated human B Lymphocytes as APCs: a novel approach for immunotherapy. Oncotarget 2018; 7:77732-77748. [PMID: 27780916 PMCID: PMC5363617 DOI: 10.18632/oncotarget.12792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023] Open
Abstract
Efficient antigen presentation is indispensable for cytotoxic T lymphocyte (CTL)-mediated immunotherapy. B-lymphocytes propagated with CD40L have been developed as antigen-presenting cells (APCs), but this capacity needs further optimization. Here, we aimed to expand human B-lymphocytes on a large scale while maintaining their antigen-presenting ability by using both CD40L and B-cell activating factor (BAFF). The addition of BAFF enhanced the expansion efficiency and prolonged the culture time without causing apoptosis of the expanded B-cells. This method thus provided an almost unlimited source of cellular adjuvant to achieve sufficient expansion of CTLs in cases where several rounds of stimulation are required. We also showed that the addition of BAFF significantly enhanced the expression of major costimulatory molecules, CD80 and CD86. Subsequently, the antigen-presenting ability of the B-lymphocytes also increased. Consequently, these B-lymphocytes showed robust CTL responses to inhibit tumor growth after tumor-specific peptide pulses. A similar method induced potent antigen-specific CTL responses, which effectively eradicated human immunodeficiency virus type 1 (HIV-1) latency in CD4 T-lymphocytes isolated from patients receiving suppressive anti-retroviral therapy (ART). Together, our findings indicate that potent antigen-specific CTLs can be generated using BAFF-activated B-lymphocytes as APCs ex vivo. This approach can be applied for CTL-mediated immunotherapy in patients with cancers or chronic viral infections.
Collapse
Affiliation(s)
- Zhang Yiwen
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Gao Shilin
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Chen Yingshi
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Su Lishi
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Luo Baohong
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Liu Chao
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Li Linghua
- Department of Infectious Diseases, Guangzhou 8th People's Hospital, Guangzhou, Guangdong, 510080, China
| | - Pan Ting
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhang Hui
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
13
|
Elbers JP, Brown MB, Taylor SS. Identifying genome-wide immune gene variation underlying infectious disease in wildlife populations - a next generation sequencing approach in the gopher tortoise. BMC Genomics 2018; 19:64. [PMID: 29351737 PMCID: PMC5775545 DOI: 10.1186/s12864-018-4452-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 01/15/2018] [Indexed: 11/25/2022] Open
Abstract
Background Infectious disease is the single greatest threat to taxa such as amphibians (chytrid fungus), bats (white nose syndrome), Tasmanian devils (devil facial tumor disease), and black-footed ferrets (canine distemper virus, plague). Although understanding the genetic basis to disease susceptibility is important for the long-term persistence of these groups, most research has been limited to major-histocompatibility and Toll-like receptor genes. To better understand the genetic basis of infectious disease susceptibility in a species of conservation concern, we sequenced all known/predicted immune response genes (i.e., the immunomes) in 16 Florida gopher tortoises, Gopherus polyphemus. All tortoises produced antibodies against Mycoplasma agassizii (an etiologic agent of infectious upper respiratory tract disease; URTD) and, at the time of sampling, either had (n = 10) or lacked (n = 6) clinical signs. Results We found several variants associated with URTD clinical status in complement and lectin genes, which may play a role in Mycoplasma immunity. Thirty-five genes deviated from neutrality according to Tajima’s D. These genes were enriched in functions relating to macromolecule and protein modifications, which are vital to immune system functioning. Conclusions These results are suggestive of genetic differences that might contribute to disease severity, a finding that is consistent with other mycoplasmal diseases. This has implications for management because tortoises across their range may possess genetic variation associated with a more severe response to URTD. More generally: 1) this approach demonstrates that a broader consideration of immune genes is better able to identify important variants, and; 2) this data pipeline can be adopted to identify alleles associated with disease susceptibility or resistance in other taxa, and therefore provide information on a population’s risk of succumbing to disease, inform translocations to increase genetic variation for disease resistance, and help to identify potential treatments. Electronic supplementary material The online version of this article (10.1186/s12864-018-4452-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean P Elbers
- School of Renewable Natural Resources, 227 RNR Bldg., Louisiana State University and AgCenter, Baton Rouge, LA, 70803, USA.
| | - Mary B Brown
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Sabrina S Taylor
- School of Renewable Natural Resources, 227 RNR Bldg., Louisiana State University and AgCenter, Baton Rouge, LA, 70803, USA
| |
Collapse
|
14
|
Activated GL7 + B cells are maintained within the inflamed CNS in the absence of follicle formation during viral encephalomyelitis. Brain Behav Immun 2017; 60:71-83. [PMID: 27658544 PMCID: PMC5215090 DOI: 10.1016/j.bbi.2016.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/13/2016] [Accepted: 09/18/2016] [Indexed: 01/12/2023] Open
Abstract
Central nervous system (CNS) inflammation associated with viral infection and autoimmune disease results in the accumulation of B cells in various differentiation stages. However, the contribution between peripheral and CNS activation remains unclear. During gliatropic coronavirus induced encephalomyelitis, accumulation of protective antibody secreting cells is preceded by infiltration of B cells with a naïve and early differentiation phenotype (Phares et al., 2014). Investigation of the temporal dynamics of B cell activation in draining cervical lymph nodes (CLN) and the CNS revealed that peak CNS infiltration of early activated, unswitched IgD+ and IgM+ B cells coincided with polyclonal activation in CLN. By contrast, isotype-switched IgG+ B cells did not accumulate until peripheral germinal center formation. In the CNS, unswitched B cells were confined to the perivascular space and meninges, with only rare B cell clusters, while isotype-switched B cells localized to parenchymal areas. Although ectopic follicle formation was not observed, more differentiated B cell subsets within the CNS expressed the germinal center marker GL7, albeit at lower levels than CLN counterparts. During chronic infection, CNS IgDint and IgD- B cell subsets further displayed sustained markers of proliferation and CD4 T cell help, which were only transiently expressed in the CLN. A contribution of local CD4 T cell help to sustain B cell activation was supported by occasional B cells adjacent to T cells. The results suggest that accumulation of differentiated B cell subsets within the CNS is largely dictated by peripheral activation, but that local events contribute to their sustained activation independent of ectopic follicle formation.
Collapse
|
15
|
Dauby N, Sartori D, Kummert C, Lecomte S, Haelterman E, Delforge ML, Donner C, Mach M, Marchant A. Limited Effector Memory B-Cell Response to Envelope Glycoprotein B During Primary Human Cytomegalovirus Infection. J Infect Dis 2015; 213:1642-50. [PMID: 26715677 DOI: 10.1093/infdis/jiv769] [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: 09/13/2015] [Accepted: 12/21/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Following primary human cytomegalovirus (HCMV) infection, the production of antibodies against envelope glycoprotein B (gB) is delayed, compared with production of antibodies against tegument proteins, and this likely reduces the control of HCMV dissemination. METHODS The frequency and the phenotype of gB-specific and tegument protein-specific B cells were studied in a cohort of pregnant women with primary HCMV infection. Healthy adults who had chronic HCMV infection or were recently immunized with tetanus toxoid (TT) were included as controls. RESULTS Primary HCMV infection was associated with high and similar frequencies of gB-specific and tegument protein-specific B cells following primary HCMV infection. During primary infection, tegument protein-specific B cells expressed an activated (CD21(low)) memory B-cell (MBC) phenotype. Activated MBCs were also induced by TT booster immunization, indicating that the expansion of this subset is part of the physiological B-cell response to protein antigens. In contrast, gB-specific B cells had a predominant classical (CD21(+)) MBC phenotype during both primary and chronic infections. CONCLUSIONS The delayed production of gB-specific immunoglobulin G (IgG) during primary HCMV infection is associated with a limited induction of MBCs with effector potential. This novel mechanism by which HCMV may interfere with the production of neutralizing antibodies could represent a target for therapeutic immunization.
Collapse
Affiliation(s)
- Nicolas Dauby
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB)
| | - Delphine Sartori
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB)
| | | | - Sandra Lecomte
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB)
| | | | | | - Catherine Donner
- Department of Obstetrics and Gynecology, Erasme Hospital, ULB, Brussels, Belgium
| | - Michael Mach
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) ImmuneHealth, Gosselies
| |
Collapse
|
16
|
Incrocci R, Hussain S, Stone A, Bieging K, Alt LAC, Fay MJ, Swanson-Mungerson M. Epstein-Barr virus Latent Membrane Protein 2A (LMP2A)-mediated changes in Fas expression and Fas-dependent apoptosis: Role of Lyn/Syk activation. Cell Immunol 2015; 297:108-19. [PMID: 26255694 DOI: 10.1016/j.cellimm.2015.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/15/2015] [Accepted: 08/04/2015] [Indexed: 01/01/2023]
Abstract
Epstein-Barr virus Latent Membrane Protein 2A (LMP2A) is expressed in EBV-infected B cells in the germinal center, a site of significant apoptosis induced by engagement of Fas on activated B cells. Signals from the B cell receptor (BCR) protect germinal center B cells from Fas-mediated apoptosis, and since LMP2A is a BCR mimic, we hypothesized that LMP2A would also protect B cells from Fas-mediated apoptosis. Surprisingly, latently-infected human and murine B cell lines expressing LMP2A were more sensitive to Fas-mediated apoptosis, as determined by increases in Annexin-V staining, and cleavage of caspase-8, -3 and PARP. Additional studies show that LMP2A-expressing B cell lines demonstrate a Lyn- and Syk-dependent increase in sensitivity to Fas-mediated apoptosis, due to an LMP2A-dependent enhancement in Fas expression. These findings demonstrate the ability for LMP2A to directly increase a pro-apoptotic molecule and have implications for EBV latency as well as the treatment of EBV-associated malignancies.
Collapse
Affiliation(s)
- Ryan Incrocci
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA
| | - Samira Hussain
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Amanda Stone
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA
| | - Kathryn Bieging
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren A C Alt
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Michael J Fay
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Michelle Swanson-Mungerson
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA.
| |
Collapse
|
17
|
Wallach-Dayan SB, Elkayam L, Golan-Gerstl R, Konikov J, Zisman P, Dayan MR, Arish N, Breuer R. Cutting edge: FasL(+) immune cells promote resolution of fibrosis. J Autoimmun 2015; 59:67-76. [PMID: 25812467 DOI: 10.1016/j.jaut.2015.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 01/26/2015] [Accepted: 02/23/2015] [Indexed: 01/07/2023]
Abstract
Immune cells, particularly those expressing the ligand of the Fas-death receptor (FasL), e.g. cytotoxic T cells, induce apoptosis in 'undesirable' self- and non-self-cells, including lung fibroblasts, thus providing a means of immune surveillance. We aimed to validate this mechanism in resolution of lung fibrosis. In particular, we elucidated whether FasL(+) immune cells possess antifibrotic capabilities by induction of FasL-dependent myofibroblast apoptosis and whether antagonists of membrane (m) and soluble (s) FasL can inhibit these capabilities. Myofibroblast interaction with immune cells and its FasL-dependency, were investigated in vitro in coculture with T cells and in vivo, following transplantation into lungs of immune-deficient syngeneic Rag-/- as well as allogeneic SCID mice, and into lungs and air pouches of FasL-deficient (gld) mice, before and after reconstitution of the mice with wild-type (wt), FasL(+) immune cells. We found that myofibroblasts from lungs resolving fibrosis undergo FasL-dependent T cell-induced apoptosis in vitro and demonstrate susceptibility to in vivo immune surveillance in lungs of reconstituted, immune- and FasL-deficient, mice. However, immune-deficient Rag-/- and SCID mice, and gld-mice with FasL-deficiency, endure the accumulation of transplanted myofibroblasts in their lungs with subsequent development of fibrosis. Concomitantly, gld mice, in contrast to chimeric FasL-deficient mice with wt immune cells, accumulated transplanted myofibroblasts in the air pouch model. In humans we found that myofibroblasts from fibrotic lungs secrete sFasL and resist T cell-induced apoptosis, whereas normal lung myofibroblasts are susceptible to apoptosis but acquire resistance upon addition of anti-s/mFasL to the coculture. Immune surveillance, particularly functional FasL(+) immune cells, may represent an important extrinsic component in myofibroblast apoptosis and serve as a barrier to fibrosis. Factors interfering with Fas/FasL-immune cell-myofibroblast interaction such as sFasL secreted by fibrotic-lung myofibroblasts, may abrogate immune surveillance during fibrosis. Annulling these factors may pave a new direction to control human lung fibrosis.
Collapse
Affiliation(s)
- Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Liron Elkayam
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Regina Golan-Gerstl
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Jenya Konikov
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Philip Zisman
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Mark Richter Dayan
- Department of Emergency Medicine, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Nissim Arish
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | - Raphael Breuer
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel; Department of Pathology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
18
|
Campo S, Allegra A, D'Ascola A, Alonci A, Scuruchi M, Russo S, Avenoso A, Gerace D, Campo GM, Musolino C. MiRNome expression is deregulated in the peripheral lymphoid compartment of multiple myeloma. Br J Haematol 2014; 165:801-13. [PMID: 24620752 DOI: 10.1111/bjh.12828] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/23/2014] [Indexed: 02/05/2023]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs involved in the regulation of gene expression. Selected groups of miRNAs are differentially expressed in various types of cancers. Alterations in miRNAs gene expression have been shown in cells from the B-cell malignancy, multiple myeloma (MM). However, although MM is a disease of plasma cells, abnormalities have been detected in the peripheral blood of the patients. The goal of our study was to analyse the entire miRNome in peripheral lymphocytes of MM patients using reverse transcription quantitative polymerase chain reaction. Using in silica analysis, we also evaluated some of the most interesting and significant pathways. Analysis revealed that MM samples had a distinct miRNA profile compared to the controls. This resulted in the identification of 203 miRNAs, 85 of which were over-expressed and 118 under-expressed. Of these, 184 possessed validated or highly predicted mRNA targets. We identified 12 354 mRNA targets of the transcriptome: 36·4% of the related proteins are involved in death processes while the 21% are required for growth and cell proliferation. We have demonstrated that miRNAs are differentially expressed in the peripheral blood of MM patients compared to controls, affecting some pathways involved in the anti-apoptotic process, cell proliferation and maybe anti-angiogenesis.
Collapse
Affiliation(s)
- Salvatore Campo
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Figgett WA, Fairfax K, Vincent FB, Le Page MA, Katik I, Deliyanti D, Quah PS, Verma P, Grumont R, Gerondakis S, Hertzog P, O'Reilly LA, Strasser A, Mackay F. The TACI receptor regulates T-cell-independent marginal zone B cell responses through innate activation-induced cell death. Immunity 2013; 39:573-83. [PMID: 24012421 DOI: 10.1016/j.immuni.2013.05.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 05/20/2013] [Indexed: 11/30/2022]
Abstract
Activation-induced cell death (AICD) plays a critical role in immune homeostasis and tolerance. In T-cell-dependent humoral responses, AICD of B cells is initiated by Fas ligand (FasL) on T cells, stimulating the Fas receptor on B cells. In contrast, T-cell-independent B cell responses involve innate-type B lymphocytes, such as marginal zone (MZ) B cells, and little is known about the mechanisms that control AICD during innate B cell responses to Toll-like receptor (TLR) activation. Here, we show that MZ B cells undergo AICD in response to TLR4 activation in vivo. The transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor and TLR4 cooperate to upregulate expression of both FasL and Fas on MZ B cells and also to repress inhibitors of Fas-induced apoptosis signaling. These findings demonstrate an unappreciated role for TACI and its ligands in the regulation of AICD during T-cell-independent B cell responses.
Collapse
Affiliation(s)
- William A Figgett
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Dextran sulfate-induced degradation of spontaneously apoptotic B cells. Int Immunopharmacol 2013; 15:581-7. [DOI: 10.1016/j.intimp.2013.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/02/2013] [Accepted: 01/16/2013] [Indexed: 01/01/2023]
|
21
|
Rakhmilevich AL, Alderson KL, Sondel PM. T-cell-independent antitumor effects of CD40 ligation. Int Rev Immunol 2012; 31:267-78. [PMID: 22804571 DOI: 10.3109/08830185.2012.698337] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD40 ligation has been shown to induce antitumor effects in mice and cancer patients. Most of the studies have focused on the ability of an agonistic anti-CD40 mAb to either directly kill CD40-positive tumor cells or activate T-cell immune responses. In this review the authors focus on the ability of CD40 ligation to activate antitumor effector mechanisms of the cells of innate immunity such as macrophages and NK cells.
Collapse
Affiliation(s)
- Alexander L Rakhmilevich
- Department of Human Oncology and Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | | | | |
Collapse
|
22
|
Hancz A, Koncz G, Szili D, Sármay G. TLR9-mediated signals rescue B-cells from Fas-induced apoptosis via inactivation of caspases. Immunol Lett 2012; 143:77-84. [PMID: 22553782 DOI: 10.1016/j.imlet.2012.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The death receptor, CD95/Fas, serves to eliminate potentially dangerous, self-reactive B cells. Engagement of B-cell receptors (BCR) on mature B-cells mediates the escape from cell death resulting in the activation and expansion of antigen specific clones. In addition to the antigen receptors, the receptors of B-cell activating factor belong to the tumor necrosis factor (TNF) family (BAFFR); moreover, the pattern recognition receptor, TLR9 may also deliver survival signals inhibiting Fas-mediated death of B-cells. Our aim was to compare the mechanism of BCR-induced and the BAFFR- or TLR9-stimulated rescue of B-cells from CD95/Fas-mediated apoptosis. We have found that BAFFR and TLR9 collaborate with BCR to protect B-cells from Fas-induced elimination and the rescue is independent of protein synthesis. The results revealed that the TLR9- and BCR-triggered rescue signals are transmitted through partially overlapping pathways; the protein kinase C (PKC) and the abl kinase induced phosphorylation may inactivate caspases in both CpG and anti-IgG stimulated cells. However, PI3-K activation is crucial upon the BCR driven anti-apoptotic effect, while p38 MAPK-mediated inactivation of caspases seems to play essential role in TLR9-mediated protection against Fas-induced programmed cell death.
Collapse
Affiliation(s)
- Anikó Hancz
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | | | | |
Collapse
|
23
|
Koncz G, Hueber AO. The Fas/CD95 Receptor Regulates the Death of Autoreactive B Cells and the Selection of Antigen-Specific B Cells. Front Immunol 2012; 3:207. [PMID: 22848207 PMCID: PMC3404404 DOI: 10.3389/fimmu.2012.00207] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/30/2012] [Indexed: 12/13/2022] Open
Abstract
Cell death receptors have crucial roles in the regulation of immune responses. Here we review recent in vivo data confirming that the Fas death receptor (TNFSR6) on B cells is important for the regulation of autoimmunity since the impairment of only Fas function on B cells results in uncontrolled autoantibody production and autoimmunity. Fas plays a role in the elimination of the non-specific and autoreactive B cells in germinal center, while during the selection of antigen-specific B cells different escape signals ensure the resistance to Fas-mediated apoptosis. Antigen-specific survival such as BCR or MHCII signal or coreceptors (CD19) cooperating with BCR inhibits the formation of death inducing signaling complex. Antigen-specific survival can be reinforced by antigen-independent signals of IL-4 or CD40 overproducing the anti-apoptotic members of the Bcl-2 family proteins.
Collapse
Affiliation(s)
- Gabor Koncz
- Immunology Research Group of the Hungarian Academy of Sciences, University Eötvös Lorand Budapest, Hungary
| | | |
Collapse
|
24
|
Mavroudi I, Papadaki HA. The role of CD40/CD40 ligand interactions in bone marrow granulopoiesis. ScientificWorldJournal 2011; 11:2011-9. [PMID: 22125452 PMCID: PMC3217605 DOI: 10.1100/2011/671453] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 10/05/2011] [Indexed: 12/14/2022] Open
Abstract
The CD40 ligand (CD40L) and CD40 are two molecules belonging to the TNF/TNF receptor superfamily, and their role in adaptive immune system has widely been explored. However, the wide range of expression of these molecules on hematopoietic as well as nonhematopoietic cells has revealed multiple functions of the CD40/CD40L interactions on different cell types and processes such as granulopoiesis. CD40 triggering on stromal cells has been documented to enhance the expression of granulopoiesis growth factors such as granulocyte-colony-stimulating factor (G-CSF) and granulocyte/monocyte-colony-stimulating factor (GM-CSF), and upon disruption of the CD40/CD40L-signaling pathway, as in the case of X-linked hyperimmunoglobulin M (IgM) syndrome (XHIGM), it can lead to neutropenia. In chronic idiopathic neutropenia (CIN) of adults, however, under the influence of an inflammatory microenvironment, CD40L plays a role in granulocytic progenitor cell depletion, providing thus a pathogenetic cause of CIN.
Collapse
Affiliation(s)
- Irene Mavroudi
- Department of Hematology, University of Crete School of Medicine, P.O. Box 1352, 71110 Heraklion, Crete, Greece
| | | |
Collapse
|
25
|
Wong ESW, Papenfuss AT, Belov K. Genomic identification of chemokines and cytokines in opossum. J Interferon Cytokine Res 2011; 31:317-30. [PMID: 21214366 DOI: 10.1089/jir.2010.0045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The cytokine repertoire of marsupials is largely unknown. The sequencing of the opossum genome has expedited the identification of many immune genes. However, many genes have not been identified using automated annotation pipelines because of high levels of sequence divergence. To fill gaps in our knowledge of the cytokine gene complement in marsupials, we searched the genome assembly of the gray short-tailed opossum for chemokine, interleukin, colony-stimulating factor, tumor necrosis factor, and transforming growth factor genes. In particular, we focused on genes that were not previously identified through Ensembl's automatic annotations. We report that the vast majority of cytokines are conserved, with direct orthologs between therian species. The major exceptions are chemokine genes, which show lineage-specific duplication/loss. Thirty-six chemokines were identified in opossum, including a lineage-specific expansion of macrophage inflammatory protein family genes. Divergent cytokines IL7, IL9, IL31, IL33, and CSF2 were identified. This is the first time IL31 and IL33 have been described outside of eutherian species. The high levels of similarities between the cytokine gene repertoires of therians suggest that the marsupial immune response is highly similar to eutherians.
Collapse
Affiliation(s)
- Emily S W Wong
- 1 Faculty of Veterinary Sciences, University of Sydney , Camperdown, Australia
| | | | | |
Collapse
|
26
|
Naïve and memory B cells in the rhesus macaque can be differentiated by surface expression of CD27 and have differential responses to CD40 ligation. J Immunol Methods 2010; 363:166-76. [PMID: 20875419 DOI: 10.1016/j.jim.2010.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 09/10/2010] [Accepted: 09/13/2010] [Indexed: 11/20/2022]
Abstract
The rhesus macaque (RM) model has the potential to be an invaluable tool for studying B cell populations during pathogenic infections, however, to date, there has been no definitive delineation of naïve and memory B cell populations in the RM. This has precluded a rigorous analysis of the generation, persistence and resolution of a pathogen-specific memory B cell response. The present study utilized multiple analyses to demonstrate that CD27 expression on B cells is consistent with a memory phenotype. Compared to CD20+CD27- B cells, CD20+CD27+ B cells were larger in size, and preferentially accumulated at effector sites. Direct sequence analysis revealed that CD20+CD27+ B cells had an increased frequency of point mutations that were consistent with somatic hypermutation and at a functional level, CD40 ligation improved CD20+CD27- but not CD20+CD27+ B cell survival in vitro. These data provide definitive evidence that the naïve and memory B cell populations of the RM can be differentiated using surface expression of CD27.
Collapse
|
27
|
Abstract
IMPORTANCE OF THE FIELD A significant number of patients relapse or do not respond to rituximab due to intrinsic or acquired resistance. Hence, mAbs targeting other cell surface antigens on B-cell lymphomas are being studied. CD80 is a glycoprotein expressed on Hodgkin's lymphoma, mature B-cell lymphomas and immunoeffector cells which may have T-regulatory, in addition to direct antitumor activity. CD80 serves as an attractive target in the continued development of mAbs against lymphoma. AREAS COVERED IN THIS REVIEW Preclinical studies with galiximab, an anti-CD80 primatized mAb, have been encouraging and have demonstrated antitumor activity against various B-cell lymphoma models, both as a single agent as well as in combination with rituximab. Data were reviewed from a PubMed literature search from 1975 to 2009 and also included a review of abstracts from published proceedings of annual meetings from the American Society of Hematology and International Conference of Malignant Lymphoma, Lugano. WHAT THE READER WILL GAIN Readers will gain a better understanding of mechanisms of action (both documented and proposed) of galiximab. An update of currently available clinical data will be presented. TAKE HOME MESSAGE Data from completed clinical trials are promising and galiximab is being studied in both upfront and relapsed settings with the potential of being incorporated into the future treatment of B-cell lymphoma.
Collapse
Affiliation(s)
- Seema Bhat
- Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
28
|
Abstract
BACKGROUND Third-party-specific cytotoxic T lymphocytes (CTL), or veto CTL, are being assessed as a cellular therapeutic for the induction of T-cell tolerance during transplantation. Conceptually, veto cell-expressed antigens (Ags) may induce B-cell immune responses, and this may have deleterious consequences. Whether veto cells induce immunity, tolerance, or are ignored by B lymphocytes has, however, not been addressed. METHODS CTL were retrovirally transduced with a model cell surface Ag to generate veto CTL. The impact of CTL-specific Ag expression on the activation and tolerization of Ag-specific B cells was assessed in vitro and, using adoptive transfer models, in vivo. RESULTS In vitro, CTL-expressed Ag induced an abortive proliferative response in specific B lymphocytes, whereby an initial proliferative burst was followed by cell death. In vivo, the administration of veto CTL also induced B-cell tolerance. Specific immunoglobulin was not detected after subsequent immunization with a veto cell-expressed Ag. Modeling of this effect with Ag-specific B-cell receptor transgenic B lymphocytes demonstrated that Ag-specific B cells were eliminated by the veto CTL; the cell division was accompanied by the exhaustion and depletion of responding cells. Veto-induced B-cell tolerance could be wholly abrogated by treatment with the toll-like receptor ligand lipopolysaccharide, implying that this tolerance resulted from the absence of adequate supplemental signals during antigenic stimulation. CONCLUSIONS Veto CTL are effective promoters of B-cell tolerance. Further assessment of their therapeutic potential in this regard is warranted.
Collapse
|
29
|
Abstract
Normal, bi-directional interactions between CD 40 and its natural ligand CD 154 (CD 40 ligand) are central to the generation of both T cell-dependent, humoral immune responses and cytotoxic T-cell responses. CD 40 is expressed on a broad range of hematological and epithelial malignancies. The development of monoclonal antibodies directed against CD 40 allows effective targeting of malignant cells through multiple mechanisms that include the recruitment of immune effector mechanisms such as complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity, direct anti-proliferative effects on neoplastic cells and, importantly, by the activation of tumor-targeted cellular cytotoxicity. This review provides the background to the early clinical trial data that are now beginning to emerge for this potentially exciting new treatment approach.
Collapse
Affiliation(s)
- Tom Geldart
- Cancer Research UK Oncology Unit, Cancer Sciences Division, School of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | | |
Collapse
|
30
|
Increased expression of CD154 and FAS in SLE patients’ lymphocytes. Rheumatol Int 2009; 30:181-5. [DOI: 10.1007/s00296-009-0933-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Accepted: 03/28/2009] [Indexed: 10/20/2022]
|
31
|
Law CL, Grewal IS. Therapeutic interventions targeting CD40L (CD154) and CD40: the opportunities and challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:8-36. [PMID: 19760064 DOI: 10.1007/978-0-387-89520-8_2] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD40 was originally identified as a receptor on B-cells that delivers contact-dependent T helper signals to B-cells through interaction with CD40 ligand (CD40L, CD154). The pivotal role played by CD40-CD40L interaction is illustrated by the defects in B-lineage cell development and the altered structures of secondary lymphoid tissues in patients and engineered mice deficient in CD40 or CD40L. CD40 signaling also provides critical functions in stimulating antigen presentation, priming of helper and cytotoxic T-cells and a variety of inflammatory reactions. As such, dysregulations in the CD40-CD40L costimulation pathway are prominently featured in human diseases ranging from inflammatory conditions to systemic autoimmunity and tissue-specific autoimmune diseases. Moreover, studies in CD40-expressing cancers have provided convincing evidence that the CD40-CD40L pathway regulates survival of neoplastic cells as well as presentation of tumor-associated antigens to the immune system. Extensive research has been devoted to explore CD40 and CD40L as drug targets. A number of anti-CD40L and anti-CD40 antibodies with diverse biological effects are in clinical development for treatment of cancer and autoimmune diseases. This chapter reviews the role of CD40-CD40L costimulation in disease pathogenesis, the characteristics of therapeutic agents targeting this pathway and status of their clinical development.
Collapse
Affiliation(s)
- Che-Leung Law
- Department of Preclinical Therapeutics, Seattle Genetics Inc., 21823 30th Drive SE, Bothell, Washington, 98021, USA.
| | | |
Collapse
|
32
|
Lugli E, Ferraresi R, Roat E, Troiano L, Pinti M, Nasi M, Nemes E, Bertoncelli L, Gibellini L, Salomoni P, Cooper EL, Cossarizza A. Quercetin inhibits lymphocyte activation and proliferation without inducing apoptosis in peripheral mononuclear cells. Leuk Res 2008; 33:140-50. [PMID: 18774171 DOI: 10.1016/j.leukres.2008.07.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2008] [Revised: 07/11/2008] [Accepted: 07/23/2008] [Indexed: 01/08/2023]
Abstract
Toxicity of chemotherapeutic drugs towards normal cells is a serious side effect of cancer treatment. Thus, finding of molecules with low toxicity for normal cells is crucial. Several natural compounds, such as flavonoid quercertin, are receiving a growing attention as "chemopreventers". Quercetin kills tumour-derived cell lines, but little is known about its effects on normal cells. Here we show that although quercetin exerts a higher apoptotic potential on leukemic cell lines than on peripheral blood mononuclear cells (PBMCs) and does not sensitize PBMCs to CD95-induced apoptosis, it is able to inhibit normal immune functions such as T cell proliferation and activation. Quercetin sensitivity is independent on cell cycle progression since it was not abrogated in serum-starved U937 cells, nor proliferating PBMCs underwent apoptosis after quercetin treatment. However, quercetin prevented PHA-induced PBMC proliferation and SEB-induced upregulation of activation markers. Our data suggest that quercetin, while incapable of inducing apoptosis in normal cells under several conditions, could interfere with effector T cell function.
Collapse
Affiliation(s)
- Enrico Lugli
- Department of Biomedical Sciences, Chair of Immunology, University of Modena and Reggio Emilia, via Campi 287, 41100 Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
High expression of CD40 on B-cell precursor acute lymphoblastic leukemia blasts is an independent risk factor associated with improved survival and enhanced capacity to up-regulate the death receptor CD95. Blood 2008; 112:1028-34. [PMID: 18552209 DOI: 10.1182/blood-2007-11-123315] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD40 and CD27, members of the tumor necrosis factor receptor (TNFR) family, are critical regulators of lymphocyte growth and differentiation. In B-cell precursor acute lymphoblastic leukemia (BCP-ALL), we prospectively assessed the impact of CD40 and CD27 on outcome in 121 children treated according to the CoALL06-97 protocol. Expression of both CD40 and CD27 was found to be significantly higher in low- than in high-risk patients as defined by standard clinical risk parameters such as age and white blood cell count. In addition, in multivariable analysis, a very high percentage of CD40(+) blasts at diagnosis was identified as an independent favorable prognostic factor for relapse-free survival. Of note, high CD40 expression particularly protected against late relapse. In B cells, CD40 is known to enhance both antigen-presenting capacity and sensitivity to proapoptotic signals. Yet, although CD40 ligation does result in significant up-regulation of CD80/CD86 in our cohort, it is up-regulation of the death receptor CD95 that significantly correlates with the percentage of CD40(+) blasts. Thus very high expression of CD40 on BCP-ALL blasts is an independent prognostic marker indicative of superior relapse-free survival that may in part be due to CD40-dependent death receptor up-regulation.
Collapse
|
34
|
Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, Hansen A, Burmester GR, Diamond B, Lipsky PE, Dörner T. Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: Delineation by expression of CD27, IgD, and CD95. ACTA ACUST UNITED AC 2008; 58:1762-73. [DOI: 10.1002/art.23498] [Citation(s) in RCA: 212] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Lee JK, Mathew SO, Vaidya SV, Kumaresan PR, Mathew PA. CS1 (CRACC, CD319) induces proliferation and autocrine cytokine expression on human B lymphocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:4672-8. [PMID: 17878365 DOI: 10.4049/jimmunol.179.7.4672] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CS1 (CRACC, CD319), a member of the CD2 family of cell surface receptors, is implicated in the activation of NK cell-mediated cytotoxicity. Previous studies showed that CS1 is also expressed on activated B cells. However, the functional role of CS1 in human B-lymphocytes is not known. Two isoforms of CS1, CS1-L and CS1-S, are expressed in human NK cells that differentially regulate NK cell function. CS1-L contains immunoreceptor tyrosine-based switch motifs in its cytoplasmic domain whereas CS1-S lacks immunoreceptor tyrosine-based switch motifs. In this study, we show that human B lymphocytes express only the CS1-L isoform, and its expression is up-regulated upon B cell activation with various stimulators. Moreover, anti-CS1 mAb strongly enhanced proliferation of both freshly isolated as well as activated B cells. The enhanced proliferation effects of CS1 were most prominent on B cells activated by anti-CD40 mAbs and/or hrIL-4. The effects of CS1 on B cell proliferation were shown on both naive and memory B cells. Human cytokine microarray and quantitative real-time PCR results indicated that CS1 activation enhanced mRNA transcripts of flt3 ligand, lymphotoxin A, TNF, and IL-14. Neutralizing Abs against lymphotoxin A, TNF-alpha, and/or flt3 ligand abolished the ability of CS1 on the B cell proliferation. These results suggest that activation of B lymphocytes, through surface CS1, may be mediated through secretion of autocrine cytokines and CS1 may play a role in the regulation of B lymphocyte proliferation during immune responses.
Collapse
Affiliation(s)
- Jae Kyung Lee
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
36
|
Bonsignori M, D'Costa S, Surman S, Hurwitz JL. A combination of 5-fluorouracil and membrane-bound antibody inhibits B-cell lymphoma growth in a mouse model system. Leuk Lymphoma 2007; 48:406-9. [PMID: 17325904 DOI: 10.1080/10428190601078738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Benson RJ, Hostager BS, Bishop GA. Rapid CD40-mediated rescue from CD95-induced apoptosis requires TNFR-associated factor-6 and PI3K. Eur J Immunol 2006; 36:2535-43. [PMID: 16897814 DOI: 10.1002/eji.200535483] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The activation molecule CD40 and the death receptor CD95/Fas play important roles in regulating B cells so that effective antimicrobial immunity occurs without autoimmunity. CD40 signaling increases CD95 expression, sensitizing cells to apoptosis, but sustained CD40 signals rescue B cells from CD95 killing. Here we describe a mechanism of early CD40-mediated rescue from CD95-induced apoptosis in B cells. Maximal rescue was achieved when CD40 signals were given within 1-2 h of initiating CD95 apoptosis. CD40 signaling did not block association of Fas-associated death domain-containing protein with CD95, but decreased CD95-induced activation of caspases 3 and 8. Rapid CD40 rescue did not require NF-kappaB activation and was independent of de novo protein synthesis, but was dependent upon active PI3 K. Signaling via a CD40 mutant that does not bind TNFR-associated factor (TRAF)1, TRAF2, and TRAF3 rescued B cells from CD95-induced apoptosis. TRAF1/2/3-independent rescue was confirmed in B cell lines made deficient in these TRAF molecules by gene targeting. In contrast, CD40 rescue was completely abrogated in TRAF6-deficient B cells, which showed reduced activation of Akt in response to CD40 engagement. These results reveal a new rapid mechanism to balance B cell activation and apoptosis.
Collapse
Affiliation(s)
- Rebecca J Benson
- Medical Scientist Training Program and Immunology Graduate Program, University of Iowa and Veterans Affairs Medical Center, Iowa City 52245, USA
| | | | | |
Collapse
|
38
|
Tangye SG, Bryant VL, Cuss AK, Good KL. BAFF, APRIL and human B cell disorders. Semin Immunol 2006; 18:305-17. [PMID: 16916610 DOI: 10.1016/j.smim.2006.04.004] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 04/03/2006] [Indexed: 12/19/2022]
Abstract
B cells require signals from multiple sources for their development from precursor cells, and differentiation into effector cells. BAFF has been identified as a critical regulator of B cell development and differentiation. Defects in the production of BAFF and/or expression of its receptors have been associated with a diverse array of human immunopathologies characterised by perturbed B cell function and behaviour, including autoimmunity, malignancy, and immunodeficiency. This review will discuss the role of BAFF in the pathogenesis of these human immune disorders. It will also highlight relevant differences between the function of BAFF in humans and mice and the impact of this on the therapeutic utility of BAFF antagonists in the treatment of different human diseases.
Collapse
Affiliation(s)
- Stuart G Tangye
- Centenary Institute of Cancer Medicine and Cell Biology, Newtown, NSW, Australia.
| | | | | | | |
Collapse
|
39
|
Salomoni P, Guernah I, Pandolfi PP. The PML-nuclear body associated protein Daxx regulates the cellular response to CD40. Cell Death Differ 2006; 13:672-5. [PMID: 16311507 DOI: 10.1038/sj.cdd.4401820] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
40
|
Crow MK. Modification of accessory molecule signaling. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 2006; 27:409-24. [PMID: 16738953 DOI: 10.1007/s00281-006-0018-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Accepted: 03/30/2006] [Indexed: 01/06/2023]
Abstract
The concept of costimulation, the requirement for an independent accessory cellular activation signal that supplements the signal delivered to a lymphocyte by antigen, has been a focal point of progress in understanding the regulation of the immune system. While considerable attention has been directed to new developments related to the activation of cells of the innate immune system through Toll-like receptors, resulting in the production of soluble mediators, augmented expression of cell surface costimulatory molecules on antigen-presenting cells is arguably the most significant early outcome of immune system activation. It is those cell surface molecules that provide the essential afferent costimulatory signals to T cells of the adaptive immune response. Once fully activated, T cells express their own cell surface accessory molecules that permit those T cells to instruct interacting B cells, macrophages, and dendritic cells to further implement an effective immune response. Significantly for patients with autoimmune diseases, the manipulation of costimulatory signals represents a rational and effective approach to modulating the chronic immune system activation that characterizes those diseases. Further elucidation of the complexities of members of the accessory molecule families and their functions should lead to an ever greater capacity for therapeutic modulation of the immune response in autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Department of Medicine, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA.
| |
Collapse
|
41
|
Yu J, Ren X, Cao S, Zhang W, Hao X. Th1 Polarization and Apoptosis-Inducing Activity of CD4+T -Cells in Cytokine-Induced Killers Might Favor the Antitumor Cytotoxicity of Cytokine-Induced KillersIn Vivo. Cancer Biother Radiopharm 2006; 21:276-84. [PMID: 16918305 DOI: 10.1089/cbr.2006.21.276] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The cytokines induced killers (CIKs) treatment is an emerging adoptive immunotherapy with greater antitumor activity than lymphocyte-activated killers (LAKs). Our previous study suggested that CD4+ T-cells in CIKs (CD4+ CIKs) might contribute to the CIK-mediated therapy in vivo. In this experiment, we studied the mechanisms that might be involved. METHODS Fresh CD4+ CIKs were purified and proportions of Th1- and Th2-type cells were determined by intracellular cytokine staining. Cytokine secretion and mRNA synthesis were measured by enzyme-linked immunosorbent assay (ELISA) and real-time quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), respectively. Direct cytolysis and apoptosis-inducing activity were examined by the lactate dehydrogenase (LDH) method and Annexin-V staining, respectively. RESULTS The Th1 polarization in CD4+ CIKs was identified, characterized with the enhanced frequency of Th1 subset, and a dramatic increase of the levels of interleukin-2 (IL-2) and interferon gamma (IFN-gamma) in the culture supernatant. The elevation in synthesis of Th1-type cytokines could be maintained without any exogenous cytokine supplement, as implied by the results from quantitative RT-PCR. Although no tumor lysis occurred, an early stage of apoptosis was detected in Raji cocultured with CD4+ CIKs after 4 hours of incubation. This apoptosis-inducing activity of CD4+ CIKs elevated along with the incubation time and depended on the cell contact through the Fas/FasL pathway. CONCLUSIONS CD4+ CIK is a subset that might favor the antitumor cytotoxicity of CIKs in vivo by producing an advantageous Th1-dominance microenvironment and inducing tumor apoptosis though the Fas/FasL pathway.
Collapse
Affiliation(s)
- Jinpu Yu
- Department of Immunology, Tianjin Cancer Institute and Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | | | |
Collapse
|
42
|
Voorzanger-Rousselot N, Alberti L, Blay JY. CD40L induces multidrug resistance to apoptosis in breast carcinoma and lymphoma cells through caspase independent and dependent pathways. BMC Cancer 2006; 6:75. [PMID: 16545138 PMCID: PMC1435764 DOI: 10.1186/1471-2407-6-75] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 03/18/2006] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND CD40L was found to reduce doxorubicin-induced apoptosis in non Hodgkin's lymphoma cell lines through caspase-3 dependent mechanism. Whether this represents a general mechanism for other tumor types is unknown. METHODS The resistance induced by CD40L against apoptosis induced by a panel of cytotoxic chemotherapeutic drugs in non Hodgkin's lymphoma and breast carcinoma cell lines was investigated. RESULTS Doxorubicin, cisplatyl, etoposide, vinblastin and paclitaxel increased apoptosis in a dose-dependent manner in breast carcinoma as well as in non Hodgkin's lymphoma cell lines. Co-culture with irradiated L cells expressing CD40L significantly reduced the percentage of apoptotic cells in breast carcinoma and non Hodgkin's lymphoma cell lines treated with these drugs. In breast carcinoma cell lines, these 5 drugs induced an inconsistent increase of caspase-3/7 activity, while in non Hodgkin's lymphoma cell lines all 5 drugs increased caspase-3/7 activity up to 28-fold above baseline. Co-culture with CD40L L cells reduced (-39% to -89%) the activation of caspase-3/7 induced by these agents in all 5 non Hodgkin's lymphoma cell lines, but in none of the 2 breast carcinoma cell lines. Co culture with CD40L L cells also blocked the apoptosis induced by exogenous ceramides in breast carcinoma and non Hodgkin's lymphoma cell lines through a caspase-3-like, 8-like and 9-like dependent pathways. CONCLUSION These results indicate that CD40L expressed on adjacent non tumoral cells induces multidrug resistance to cytotoxic agents and ceramides in both breast carcinoma and non Hodgkin's lymphoma cell lines, albeit through a caspase independent and dependent pathway respectively.
Collapse
Affiliation(s)
| | - Laurent Alberti
- Equipe Cytokines et Cancer, Unité INSERM U590, Centre Léon Bérard, 28 rue Laënnec, 69373 LYON cedex 08, France
| | - Jean-Yves Blay
- Equipe Cytokines et Cancer, Unité INSERM U590, Centre Léon Bérard, 28 rue Laënnec, 69373 LYON cedex 08, France
| |
Collapse
|
43
|
Kojima Y, Tsurumi H, Goto N, Shimizu M, Kasahara S, Yamada T, Kanemura N, Hara T, Sawada M, Saio M, Yamada T, Takahashi T, Tomita E, Takami T, Moriwaki H. Fas and Fas ligand expression on germinal center type-diffuse large B-cell lymphoma is associated with the clinical outcome. Eur J Haematol 2006; 76:465-72. [PMID: 16494623 DOI: 10.1111/j.1600-0609.2006.00631.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In recent years, diffuse large B-cell lymphoma (DLBCL) has been classified by DNA microarray analysis into the germinal center B-cell-like (GC) type, the activated B-cell-like (ABC) type and type 3. The latter two types can be collectively categorized as the non-GC (NGC) type. From the prognostic perspective, the GC type has a favorable clinical outcome when compared with the NGC type. The protein Fas induces apoptosis of lymphocytes by binding with the Fas ligand (FasL), and escape from such apoptosis is considered to lead to malignant transformation of the cells and unrestricted growth of lymphoma. We proposed a hypothesis that Fas/FasL expression could be possibly related with a better survival of GC type DLBCL and evaluated 69 DLBCL cases immunohistochemically with CD10, Bcl-6, MUM1, Fas and FasL. These lymphomas were classified as GC type (positive for CD10 or Bcl-6 and negative for MUM1) or NGC type. The GC type had a better overall survival rate than the NGC type (P = 0.0723). Among markers as given above, positive CD10 was the most significant prognostic factor for overall survival in total DLBCL (P < 0.05). In the GC type, Fas and FasL expressions were significantly associated with a favorable overall survival (Fas: P < 0.005; FasL: P < 0.05). Hence, Fas or FasL expression might contribute to a better prognosis of this type of DLBCL.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis/physiology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Biomarkers, Tumor/blood
- Cyclophosphamide/administration & dosage
- DNA-Binding Proteins/analysis
- Doxorubicin/administration & dosage
- Doxorubicin/analogs & derivatives
- Embryonal Carcinoma Stem Cells
- Fas Ligand Protein
- Female
- Follow-Up Studies
- Germinal Center/pathology
- Humans
- Interferon Regulatory Factors/analysis
- L-Lactate Dehydrogenase/blood
- Life Tables
- Lymphoma, Large B-Cell, Diffuse/classification
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Prednisolone/administration & dosage
- Prognosis
- Proportional Hazards Models
- Proto-Oncogene Proteins c-bcl-6
- Survival Analysis
- Treatment Outcome
- Tumor Necrosis Factors/biosynthesis
- Tumor Necrosis Factors/genetics
- Vincristine/administration & dosage
- fas Receptor/biosynthesis
- fas Receptor/genetics
Collapse
Affiliation(s)
- Yasushi Kojima
- First Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
de Goër de Herve MG, Durali D, Tran TA, Maigné G, Simonetta F, Leclerc P, Delfraissy JF, Taoufik Y. Differential effect of agonistic anti-CD40 on human mature and immature dendritic cells: the Janus face of anti-CD40. Blood 2005; 106:2806-14. [PMID: 15994291 DOI: 10.1182/blood-2004-12-4678] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AbstractAgonistic monoclonal antibodies to CD40 (CD40 mAbs) have a puzzling dual therapeutic effect in experimental animal models. CD40 mAbs induce tumor regression by potentiating antitumoral T-cell responses, yet they also have immunosuppressive activity in chronic autoimmune inflammatory processes. CD40 mAbs are thought to act on antigen presentation by dendritic cells (DCs) to T cells. DCs can be distinguished as either immature or mature by their phenotype and their ability to generate an effective T-cell response. Here we found that, on human cells, although anti-CD40 led immature DCs to mature and became immunogenic, it also reduced the capacity of lipopolysaccharide (LPS) and tumor necrosis factor α (TNF-α)-matured DCs to generate a specific CD4 T-cell response. This inhibitory effect was related to rapid and selective apoptosis of mature DCs. Anti-CD40-mediated apoptosis was due to an indirect mechanism involving cooperation with the death domain-associated receptor Fas, leading to activation of Fas-associated death domain protein (FADD) and caspase-8. On human cells, CD40 activation by such agonists could, therefore, trigger immune responses to antigens presented by immature DCs, which are otherwise nonimmunogenic, by inducing maturation. On the other hand, anti-CD40 mAbs, by rapidly inducing apoptosis, may reduce the capacity of inflammatory signal-matured immunogenic DCs to generate an effective T-cell response. These results call for caution in CD40 mAb-based immunotherapy strategies. (Blood. 2005;106:2806-2814)
Collapse
|
45
|
Lee JK, Seki N, Sayers TJ, Subleski J, Gruys EM, Murphy WJ, Wiltrout RH. Constitutive expression of functional CD40 on mouse renal cancer cells: induction of Fas and Fas-mediated killing by CD40L. Cell Immunol 2005; 235:145-52. [PMID: 16213477 DOI: 10.1016/j.cellimm.2005.08.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2005] [Revised: 08/22/2005] [Accepted: 08/24/2005] [Indexed: 01/08/2023]
Abstract
CD40, a member of the TNF receptor superfamily, is expressed on B cells, dendritic cells, and some tumor cells, including melanoma and bladder carcinoma. In this study, we report that both mouse and human renal carcinoma cells (RCC) also constitutively express functional CD40. Treatment of mouse RCC with CD40L induced strong expression of genes and proteins for ICAM-1 and Fas, and this expression was further enhanced by combining CD40L with IFN-gamma. Similar effects were demonstrated using an agonist anti-CD40 antibody. The increased levels of Fas expression on RCC after treatment with CD40L plus IFN-gamma resulted in potent killing by either FasL-positive effector cells or agonistic anti-Fas antibody. The combination of CD40L plus IFN-gamma also significantly enhanced killing of RCC by tumor-specific CTL lines. Our results demonstrate that constitutively expressed CD40 is functionally active and may provide a molecular target for the development of new approaches to the treatment of RCC.
Collapse
Affiliation(s)
- J-K Lee
- National Genome Research Institute, National Institutes of Health, 5 Nokbun-dong, Eunpyung-ku, Seoul 122-701, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
46
|
Arens R, Baars PA, Jak M, Tesselaar K, van der Valk M, van Oers MHJ, van Lier RAW. Cutting Edge: CD95 Maintains Effector T Cell Homeostasis in Chronic Immune Activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:5915-20. [PMID: 15879081 DOI: 10.4049/jimmunol.174.10.5915] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The elimination of activated T cells is important to maintain homeostasis and avoid immunopathology. CD95 (Fas/APO-1) has been identified as a death mediator for activated T cells in vitro but the function of CD95 in death of mature T cells in vivo is still controversial. Here we show that triggering of the costimulatory TNF receptor family member CD27 sensitized T cells for CD95-induced apoptosis. CD95-deficient (lpr/lpr) T cells massively expanded and differentiated into IFN-gamma-secreting effector cells in transgenic mice that constitutively express the CD27 ligand, CD70. Concomitantly, CD95-deficient CD70 transgenic mice became moribund by 4 wk of age with severe liver pathology and bone marrow failure. These findings establish that CD95 is a critical regulator of effector T cell homeostasis in chronic immune activation.
Collapse
Affiliation(s)
- Ramon Arens
- Departments of Experimental Immunology and Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
47
|
Mizuno T, Rothstein TL. B cell receptor (BCR) cross-talk: CD40 engagement enhances BCR-induced ERK activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:3369-76. [PMID: 15749869 DOI: 10.4049/jimmunol.174.6.3369] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bystander B cells may be initially stimulated through CD40, which enhances susceptibility to Fas-mediated apoptosis, before encountering Ag, which produces Fas resistance. A key issue in this process is to what extent CD40 cross-talk might affect subsequent BCR signaling. It has previously been shown that CD40 engagement bypasses or mitigates the need for Bruton's tyrosine kinase in subsequent BCR signaling for NF-kappaB activation. However, the full extent of the effects of CD40 on BCR signaling has not been delineated. In the present study we evaluated the possibility that CD40-mediated cross-talk also affects another principal outcome of BCR signaling: MAPK activation. We found that prior stimulation of primary murine B cells with CD40L markedly enhanced the level of ERK and JNK (but not p38 MAPK) phosphorylation produced by subsequently added anti-Ig Ab, and much, but not all, of this enhancement was independent of PI3K and phospholipase C. CD40L treatment similarly enhanced BCR-induced MAPK kinase (MEK) phosphorylation, and MEK was required for enhancement of ERK. Although BCR-induced c-Raf phosphorylation was also enhanced by prior CD40L treatment, c-Raf was not required for MEK/ERK phosphorylation. These results identify a novel system of receptor cross-talk between CD40 and BCR and indicate that the effects of CD40 engagement on subsequent BCR stimulation spread beyond NF-kappaB to involve the MAPK pathway.
Collapse
Affiliation(s)
- Takuya Mizuno
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
48
|
Lankoff A, Carmichael WW, Grasman KA, Yuan M. The uptake kinetics and immunotoxic effects of microcystin-LR in human and chicken peripheral blood lymphocytes in vitro. Toxicology 2004; 204:23-40. [PMID: 15369846 DOI: 10.1016/j.tox.2004.05.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Accepted: 05/13/2004] [Indexed: 10/26/2022]
Abstract
Microcystin-LR is a cyanobacterial heptapeptide that presents acute and chronic hazards to animal and human health. We investigated the influence of this toxin on human and chicken immune system modulation in vitro. Peripheral blood lymphocytes were treated with microcystin-LR at environmentally relevant doses of 1, 10 and 25 microg/ml for 12, 24, 48, 72 h (for proliferation assay cells were treated for 72 h). T-cell and B-cell proliferation as well as apoptosis and necrosis were determined in human and chicken samples. IL-2 and IL-6 production by human lymphocytes also was measured. In addition, uptake kinetics of microcystin-LR into human and chicken peripheral blood lymphocytes were calculated by Liquid Chromatography (LS) /Mass Spectrometry (MS) analysis. At the highest dose microcystin-LR decreased T-cell proliferation and all doses of microcystin-LR inhibited B-cell proliferation. The frequency of apoptotic and necrotic cells increased in a dose and time-dependent manner. Human lymphocytes responded to stimulation with microcystin-LR by increased production of IL-6 and decreased production of IL-2. Human lymphocytes were able to uptake from 0.014 to 1.663 microg/ml and chicken lymphocytes from 0.035 to 1.733 microg/ml of the microcystin-LR added to the cultures, depending on the treatment time and dose. In conclusion, microcystin-LR acted as an immunomodulator in cytokine production and down-regulated lymphocyte functions by induction of apoptosis and necrosis. However, further studies dealing with the influence of microcystin-LR on expression cytokine genes and transcription factors are necessary to confirm these hypotheses.
Collapse
Affiliation(s)
- Anna Lankoff
- Department of Radiobiology and Immunology, Institute of Biology, Pedagogical University, Swietokrzyska Academy, 25-406 Kielce, ul. Swietokrzyska, 1525 425, Poland.
| | | | | | | |
Collapse
|
49
|
Basso K, Klein U, Niu H, Stolovitzky GA, Tu Y, Califano A, Cattoretti G, Dalla-Favera R. Tracking CD40 signaling during germinal center development. Blood 2004; 104:4088-96. [PMID: 15331443 DOI: 10.1182/blood-2003-12-4291] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Substantial evidence indicates that signaling through the CD40 receptor (CD40) is required for germinal center (GC) and memory B-cell formation. However, it is not fully understood at which stages of B-cell development the CD40 pathway is activated in vivo. To address this question, we induced CD40 signaling in human transformed GC B cells in vitro and identified a CD40 gene expression signature by DNA microarray analysis. This signature was then investigated in the gene expression profiles of normal B cells and found in pre- and post-GC B cells (naive and memory) but, surprisingly, not in GC B cells. This finding was validated in lymphoid tissues by showing that the nuclear factor-kappaB (NF-kappaB) transcription factors, which translocate to the nucleus upon CD40 stimulation, are retained in the cytoplasm in most GC B cells, indicating the absence of CD40 signaling. Nevertheless, a subset of centrocytes and B cells in the subepithelium showed nuclear staining of multiple NF-kappaB subunits, suggesting that a fraction of naive and memory B cells may be subject to CD40 signaling or to other signals that activate NF-kappaB. Together, these results show that GC expansion occurs in the absence of CD40 signaling, which may act only in the initial and final stages of the GC reaction.
Collapse
Affiliation(s)
- Katia Basso
- Institute for Cancer Genetics, Department of Pathology and Genetics and Development, Joint Centers for Systems Biology, Columbia University, 1150 St Nicholas Ave, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
He B, Chadburn A, Jou E, Schattner EJ, Knowles DM, Cerutti A. Lymphoma B cells evade apoptosis through the TNF family members BAFF/BLyS and APRIL. THE JOURNAL OF IMMUNOLOGY 2004; 172:3268-79. [PMID: 14978135 DOI: 10.4049/jimmunol.172.5.3268] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The mechanisms underlying the autonomous accumulation of malignant B cells remain elusive. We show in this study that non-Hodgkin's lymphoma (NHL) B cells express B cell-activating factor of the TNF family (BAFF) and a proliferation-inducing ligand (APRIL), two powerful B cell-activating molecules usually expressed by myeloid cells. In addition, NHL B cells express BAFF receptor, which binds BAFF, as well as transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) and B cell maturation Ag (BCMA), which bind both BAFF and APRIL. Neutralization of endogenous BAFF and APRIL by soluble TACI and BCMA decoy receptors attenuates the survival of NHL B cells, decreases activation of the prosurvival transcription factor NF-kappaB, down-regulates the antiapoptotic proteins Bcl-2 and Bcl-x(L), and up-regulates the proapoptotic protein Bax. Conversely, exposure of NHL B cells to recombinant or myeloid cell-derived BAFF and APRIL attenuates apoptosis, increases NF-kappaB activation, up-regulates Bcl-2 and Bcl-x(L), and down-regulates Bax. In some NHLs, exogenous BAFF and APRIL up-regulate c-Myc, an inducer of cell proliferation; down-regulate p53, an inhibitor of cell proliferation; and increase Bcl-6, an inhibitor of B cell differentiation. By showing that nonmalignant B cells up-regulate BAFF and APRIL upon stimulation by T cell CD40 ligand, our findings indicate that NHL B cells deregulate an otherwise physiological autocrine survival pathway to evade apoptosis. Thus, neutralization of BAFF and APRIL by soluble TACI and BCMA decoy receptors could be useful to dampen the accumulation of malignant B cells in NHL patients.
Collapse
Affiliation(s)
- Bing He
- Departments of Pathology and Laboratory Medicine, Weill Medical College, Cornell University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|