1
|
Rudd-Schmidt JA, Laine RF, Noori T, Brennan AJ, Voskoboinik I. ALFA-PRF: a novel approach to detect murine perforin release from CTLs into the immune synapse. Front Immunol 2022; 13:931820. [PMID: 36618385 PMCID: PMC9813862 DOI: 10.3389/fimmu.2022.931820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
When killing through the granule exocytosis pathway, cytotoxic lymphocytes release key effector molecules into the immune synapse, perforin and granzymes, to initiate target cell killing. The pore-forming perforin is essential for the function of cytotoxic lymphocytes, as its pores disrupt the target cell membrane and allow diffusion of pro-apoptotic serine proteases, granzyme, into the target cell, where they initiate various cell death cascades. Unlike human perforin, the detection of its murine counterpart in a live cell system has been problematic due its relatively low expression level and the lack of sensitive antibodies. The lack of a suitable methodology to visualise murine perforin secretion into the synapse hinders the study of the cytotoxic lymphocyte secretory machinery in murine models of human disease. Here, we describe a novel recombinant technology, whereby a short ALFA-tag sequence has been fused with the amino-terminus of a mature murine perforin, and this allowed its detection by the highly specific FluoTag®-X2 anti-ALFA nanobodies using both Total Internal Reflection Fluorescence (TIRF) microscopy of an artificial synapse, and confocal microscopy of the physiological immune synapse with a target cell. This methodology can have broad application in the field of cytotoxic lymphocyte biology and for the many models of human disease.
Collapse
Affiliation(s)
- Jesse A. Rudd-Schmidt
- Killer Cell Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,*Correspondence: Ilia Voskoboinik, ; Jesse A. Rudd-Schmidt,
| | - Romain F. Laine
- Medical Research Council (MRC)-Laboratory for Molecular Cell Biology, University College London, London, United Kingdom,The Francis Crick Institute, London, United Kingdom,MicrographiaBio, Translation & Innovation Hub, London, United Kingdom
| | - Tahereh Noori
- Killer Cell Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Amelia J. Brennan
- Killer Cell Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ilia Voskoboinik
- Killer Cell Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia,*Correspondence: Ilia Voskoboinik, ; Jesse A. Rudd-Schmidt,
| |
Collapse
|
2
|
CD46 Genetic Variability and HIV-1 Infection Susceptibility. Cells 2021; 10:cells10113094. [PMID: 34831317 PMCID: PMC8622916 DOI: 10.3390/cells10113094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
CD46 is the main receptor for complement protein C3 and plays an important role in adaptive immune responses. CD46 genetic variants are associated with susceptibility to several infectious and autoimmune diseases. Additionally, CD46 function can be subverted by HIV-1 to evade attack by complement, a strategy shared by viruses of other families. We sought to determine the association between CD46 gene variants and HIV-1 acquired through intravenous drug use (IDU) and sexual routes (n = 823). Study subjects were of European ancestry and were HIV-1 infected (n = 438) or exposed but seronegative (n = 387). Genotyping of the rs2796265 SNP located in the CD46 gene region was done by allele-specific real-time PCR. A meta-analysis merging IDU and sexual cohorts indicates that the minor genotype (CC) was associated with increased resistance to HIV-1 infection OR = 0.2, 95% CI (0.07–0.61), p = 0.004. The HIV-1-protective genotype is correlated with reduced CD46 expression and alterations in the ratio of CD46 mRNA splicing isoforms.
Collapse
|
3
|
Charvet B, Reynaud JM, Gourru-Lesimple G, Perron H, Marche PN, Horvat B. Induction of Proinflammatory Multiple Sclerosis-Associated Retrovirus Envelope Protein by Human Herpesvirus-6A and CD46 Receptor Engagement. Front Immunol 2018; 9:2803. [PMID: 30574140 PMCID: PMC6291489 DOI: 10.3389/fimmu.2018.02803] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022] Open
Abstract
The aberrant expression of human endogenous retrovirus (HERV) elements of the HERV-W family has been associated with different diseases, including multiple sclerosis (MS). In particular, the expression of the envelope protein (Env) from the multiple sclerosis-associated retrovirus (MSRV), a member of HERV-W family and known for its potent proinflammatory activity, is repeatedly detected in the brain lesions and blood of MS patients. Furthermore, human herpesvirus 6 (HHV-6) infection has long been suspected to play a role in the pathogenesis of MS and neuroinflammation. We show here that both HHV-6A and stimulation of its receptor, transmembrane glycoprotein CD46, induce the expression of MSRV-Env. The engagement of extracellular domains SCR3 and SCR4 of CD46-Cyt1 isoform was required for MSRV-env transactivation, limiting thus the MSRV-Env induction to the CD46 ligands binding these domains, including C3b component of complement, specific monoclonal antibodies, and both infectious and UV-inactivated HHV-6A, but neither HHV-6B nor measles virus vaccine strain. Induction of MSRV-Env required CD46 Cyt-1 singling and was abolished by the inhibitors of protein kinase C. Finally, both membrane-expressed and secreted MSRV-Env trigger TLR4 signaling, displaying thus a proinflammatory potential, characteristic for this viral protein. These data expand the specter of HHV-6A effects in the modulation of the immune response and support the hypothesis that cross-talks between exogenous and endogenous viruses may contribute to inflammatory diseases and participate in neuroinflammation. Furthermore, they reveal a new function of CD46, known as an inhibitor of complement activation and receptor for several pathogens, in transactivation of HERV env genes, which may play an important role in the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Benjamin Charvet
- International Centre for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France.,GeNeuro Innovation, Lyon, France
| | - Josephine M Reynaud
- International Centre for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Geraldine Gourru-Lesimple
- International Centre for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | | | - Patrice N Marche
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble-Alpes, IAPC, La Tronche, France
| | - Branka Horvat
- International Centre for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| |
Collapse
|
4
|
Killick J, Morisse G, Sieger D, Astier AL. Complement as a regulator of adaptive immunity. Semin Immunopathol 2018; 40:37-48. [PMID: 28842749 PMCID: PMC5794818 DOI: 10.1007/s00281-017-0644-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
The complement system is an ancient and evolutionarily conserved effector system comprising in mammals over 50 circulating and membrane bound proteins. Complement has long been described as belonging to the innate immune system; however, a number of recent studies have demonstrated its key role in the modulation of the adaptive immune response. This review does not set out to be an exhaustive list of the numerous interactions of the many complement components with adaptive immunity; rather, we will focus more precisely on the role of some complement molecules in the regulation of antigen presenting cells, as well as on their direct effect on the activation of the core adaptive immune cells, B and T lymphocytes. Recent reports on the local production and activation of complement proteins also suggest a major role in the control of effector responses. The crucial role of complement in adaptive immunity is further highlighted by several examples of dysregulation of these pathways in human diseases.
Collapse
Affiliation(s)
- Justin Killick
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Gregoire Morisse
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Dirk Sieger
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Anne L Astier
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK.
- Inserm U1043, CNRS U5282, Université de Toulouse, Centre de Physiopathologie Toulouse-Purpan (CPTP), F-31300, Toulouse, France.
| |
Collapse
|
5
|
Ni Choileain S, Hay J, Thomas J, Williams A, Vermeren MM, Benezech C, Gomez-Salazar M, Hugues OR, Vermeren S, Howie SEM, Dransfield I, Astier AL. TCR-stimulated changes in cell surface CD46 expression generate type 1 regulatory T cells. Sci Signal 2017; 10:10/502/eaah6163. [PMID: 29066539 DOI: 10.1126/scisignal.aah6163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A lack of regulatory T cell function is a critical factor in the pathogenesis of autoimmune diseases, such as multiple sclerosis (MS). Ligation of the complement regulatory protein CD46 facilitates the differentiation of T helper 1 (TH1) effector cells into interleukin-10 (IL-10)-secreting type 1 regulatory T cells (Tr1 cells), and this pathway is defective in MS patients. Cleavage of the ectodomain of CD46, which contains three N-glycosylation sites and multiple O-glycosylation sites, enables CD46 to activate T cells. We found that stimulation of the T cell receptor (TCR)-CD3 complex was associated with a reduction in the apparent molecular mass of CD46 in a manner that depended on O-glycosylation. CD3-stimulated changes in CD46 O-glycosylation status reduced CD46 processing and subsequent T cell signaling. During T cell activation, CD46 was recruited to the immune synapse in a manner that required its serine-, threonine-, and proline-rich (STP) region, which is rich in O-glycosylation sites. Recruitment of CD46 to the immune synapse switched T cells from producing the inflammatory cytokine interferon-γ (IFN-γ) to producing IL-10. Furthermore, CD4+ T cells isolated from MS patients did not exhibit a CD3-stimulated reduction in the mass of CD46 and thus showed increased amounts of cell surface CD46. Together, these data suggest a possible mechanism underlying the regulatory function of CD46 on T cells. Our findings may explain why this pathway is defective in patients with MS and provide insights into MS pathogenesis that could help to design future immunotherapies.
Collapse
Affiliation(s)
- Siobhan Ni Choileain
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Joanne Hay
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Joelle Thomas
- Université Claude Bernard Lyon I, CNRS UMR 5310-INSERM U1217, F-69100 Lyon, France
| | - Anna Williams
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Matthieu M Vermeren
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Cecile Benezech
- UK Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mario Gomez-Salazar
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Owen R Hugues
- Millipore (U.K.) Limited, Croxley Green Business Park, Watford, Hertfordshire WD18 8ZB, UK
| | - Sonja Vermeren
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Sarah E M Howie
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Ian Dransfield
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Anne L Astier
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK. .,Centre de Physiopathologie Toulouse-Purpan, INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse F-31300, France
| |
Collapse
|
6
|
Abstract
Myeloid cells make extensive use of the complement system in the context of recruitment, phagocytosis, and other effector functions. There are several types of complement receptors on myeloid cells, including G protein-coupled receptors for localizing the source of complement activation, and three sets of type I transmembrane proteins that link complement to phagocytosis: complement receptor 1, having an extracellular domain with tandem complement regulatory repeats; complement receptors 3 and 4, which are integrin family receptors comprising heterodimers of type I transmembrane subunits; and VSIG4, a member of the Ig superfamily. This review will focus on the role of the different classes of complement receptors and how their activities are integrated in the setting of immune tolerance and inflammatory responses.
Collapse
|
7
|
Pham K, Shimoni R, Charnley M, Ludford-Menting MJ, Hawkins ED, Ramsbottom K, Oliaro J, Izon D, Ting SB, Reynolds J, Lythe G, Molina-Paris C, Melichar H, Robey E, Humbert PO, Gu M, Russell SM. Asymmetric cell division during T cell development controls downstream fate. J Cell Biol 2015; 210:933-50. [PMID: 26370500 PMCID: PMC4576854 DOI: 10.1083/jcb.201502053] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
T cell precursors undergo asymmetric cell division after T cell receptor genomic recombination, with stromal cell cues controlling the differential inheritance of fate determinants Numb and α-Adaptin by the daughters of a dividing DN3a T cell precursor. During mammalian T cell development, the requirement for expansion of many individual T cell clones, rather than merely expansion of the entire T cell population, suggests a possible role for asymmetric cell division (ACD). We show that ACD of developing T cells controls cell fate through differential inheritance of cell fate determinants Numb and α-Adaptin. ACD occurs specifically during the β-selection stage of T cell development, and subsequent divisions are predominantly symmetric. ACD is controlled by interaction with stromal cells and chemokine receptor signaling and uses a conserved network of polarity regulators. The disruption of polarity by deletion of the polarity regulator, Scribble, or the altered inheritance of fate determinants impacts subsequent fate decisions to influence the numbers of DN4 cells arising after the β-selection checkpoint. These findings indicate that ACD enables the thymic microenvironment to orchestrate fate decisions related to differentiation and self-renewal.
Collapse
Affiliation(s)
- Kim Pham
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Raz Shimoni
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Mirren Charnley
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia Industrial Research Institute Swinburne, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Mandy J Ludford-Menting
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Edwin D Hawkins
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Kelly Ramsbottom
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Jane Oliaro
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David Izon
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Stephen B Ting
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Joseph Reynolds
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Carmen Molina-Paris
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Heather Melichar
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Ellen Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Patrick O Humbert
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Min Gu
- Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Sarah M Russell
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
8
|
Monocyte:T-cell interaction regulates human T-cell activation through a CD28/CD46 crosstalk. Immunol Cell Biol 2015; 93:796-803. [PMID: 25787182 PMCID: PMC4519525 DOI: 10.1038/icb.2015.42] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/20/2015] [Accepted: 03/15/2015] [Indexed: 01/20/2023]
Abstract
T cell activation requires engagement of the T cell receptor and of at least one costimulatory molecule. The key role of CD28 in inducing T cell activation has been reported several decades ago and the molecular mechanisms involved well described. The complement regulator CD46 also acts as a costimulatory molecule for T cells but, in contrast to CD28, has the ability to drive T cell differentiation from producing some IFNγ to secreting some potent anti-inflammatory IL-10, acquiring a so-called Type I regulatory phenotype (Tr1). Proteolytic cleavage of CD46 occurs upon costimulation and is important for T cell activation and IL-10 production. The observation that CD46 cleavage was reduced when PBMC were costimulated compared to purified naive T cells led us to hypothesize that interactions between different cell types within the PBMC were able to modulate the CD46 pathway. We show that CD46 downregulation is also reduced when CD4+ T cells are co-cultured with autologous monocytes. Indeed, monocyte:T cell co-cultures impaired CD46–mediated T cell differentiation and coactivation, by reducing downregulation of surface CD46, lowering induction of the early activation marker CD69, as well as reducing the levels of IL-10 secretion. Blocking of CD86 could partly restore CD69 expression and cytokine secretion, demonstrating that the CD28-CD86 pathway regulates CD46 activation. Direct concomitant ligation of CD28 and CD46 on CD4+ T cells also modulated CD46 expression and regulated cytokine production. These data identify a crosstalk between two main costimulatory pathways and provide novel insights into the regulation of human T cell activation.
Collapse
|
9
|
Hay J, Carter D, Lieber A, Astier AL. Recombinant Ad35 adenoviral proteins as potent modulators of human T cell activation. Immunology 2014; 144:453-460. [PMID: 25251258 PMCID: PMC4557682 DOI: 10.1111/imm.12391] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/03/2014] [Accepted: 09/16/2014] [Indexed: 11/30/2022] Open
Abstract
The protein CD46 protects cells from complement attack by regulating cleavage of C3b and C3d. CD46 also regulates the adaptive immune response by controlling T cell activation and differentiation. Co-engagement of the T cell receptor and CD46 notably drives T cell differentiation by switching production of IFNγ to secretion of anti-inflammatory IL-10. This regulatory pathway is altered in several chronic inflammatory diseases highlighting its key role for immune homeostasis. The manipulation of the CD46 pathway may therefore provide a powerful means to regulate immune responses. Herein, we investigated the effect of recombinant proteins derived from the fiber knob of the adenovirus serotype 35 (Ad35) that uses CD46 as its entry receptor, on human T cell activation. We compared the effects of Ad35K++, engineered to exhibit enhanced affinity to CD46, and of Ad35K-, mutated in the binding site for CD46. Ad35K++ profoundly affects T cell activation by decreasing the levels of CD46 at the surface of primary T cells, and impairing T cell co-activation, shown by decreased CD25 expression, reduced proliferation and lower secretion of IL-10 and IFNγ. In contrast, Ad35K- acts a potent coactivator of T cells, enhancing T cell proliferation and cytokine production. These data show that recombinant Ad35 proteins are potent modulators of human T cell activation, and support their further development as potential drugs targeting T cell responses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Joanne Hay
- MRC Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research InstituteEdinburgh, UK
| | - Darrick Carter
- PAI Life Sciences Inc.Seattle, WA, USA
- Compliment Corp.Seattle, WA, USA
| | - André Lieber
- Department of Medical Genetics, University of WashingtonSeattle, WA, USA
| | - Anne L Astier
- MRC Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research InstituteEdinburgh, UK
| |
Collapse
|
10
|
Ramsbottom KM, Hawkins ED, Shimoni R, McGrath M, Chan CJ, Russell SM, Smyth MJ, Oliaro J. Cutting Edge: DNAX Accessory Molecule 1–Deficient CD8+ T Cells Display Immunological Synapse Defects That Impair Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2013; 192:553-7. [DOI: 10.4049/jimmunol.1302197] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
11
|
Yamamoto H, Fara AF, Dasgupta P, Kemper C. CD46: the 'multitasker' of complement proteins. Int J Biochem Cell Biol 2013; 45:2808-20. [PMID: 24120647 DOI: 10.1016/j.biocel.2013.09.016] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 09/23/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Complement is undeniably quintessential for innate immunity by detecting and eliminating infectious microorganisms. Recent work, however, highlights an equally profound impact of complement on the induction and regulation of a wide range of immune cells. In particular, the complement regulator CD46 emerges as a key sensor of immune activation and a vital modulator of adaptive immunity. In this review, we summarize the current knowledge of CD46-mediated signalling events and their functional consequences on immune-competent cells with a specific focus on those in CD4(+) T cells. We will also discuss the promises and challenges that potential therapeutic modulation of CD46 may hold and pose.
Collapse
Affiliation(s)
- Hidekazu Yamamoto
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, UK; The Urology Centre, Guy's and St. Thomas' NHS Foundations Trust, London SE1 9RT, UK
| | | | | | | |
Collapse
|
12
|
Membrane-bound complement regulatory proteins as biomarkers and potential therapeutic targets for SLE. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:55-81. [PMID: 23402019 DOI: 10.1007/978-1-4614-4118-2_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.
Collapse
|
13
|
Kickler K, Ni Choileain S, Williams A, Richards A, Astier AL. Calcitriol modulates the CD46 pathway in T cells. PLoS One 2012; 7:e48486. [PMID: 23144765 PMCID: PMC3483209 DOI: 10.1371/journal.pone.0048486] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 10/02/2012] [Indexed: 11/19/2022] Open
Abstract
The complement regulator CD46 is a costimulatory molecule for human T cells that induces a regulatory Tr1 phenotype, characterized by large amounts of IL-10 secretion. Secretion of IL-10 upon CD46 costimulation is largely impaired in T cells from patients with multiple sclerosis (MS). Vitamin D can exert a direct effect on T cells, and may be beneficial in several pathologies, including MS. In this pilot study, we examined whether active vitamin D (1,25(OH)(2)D(3) or calcitriol) could modulate the CD46 pathway and restore IL-10 production by CD46-costimulated CD4+ T cells from patients with MS. In healthy T cells, calcitriol profoundly affects the phenotype of CD46-costimulated CD4+ T cells, by increasing the expression of CD28, CD25, CTLA-4 and Foxp3 while it concomitantly decreased CD46 expression. Similar trends were observed in MS CD4+ T cells except for CD25 for which a striking opposite effect was observed: while CD25 was normally induced on MS T cells by CD46 costimulation, addition of calcitriol consistently inhibited its induction. Despite the aberrant effect on CD25 expression, calcitriol increased the IL-10:IFNγ ratio, characteristic of the CD46-induced Tr1 phenotype, in both T cells from healthy donors and patients with MS. Hence, we show that calcitriol affects the CD46 pathway, and that it promotes anti-inflammatory responses mediated by CD46. Moreover, it might be beneficial for T cell responses in MS.
Collapse
Affiliation(s)
- Karoline Kickler
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Siobhan Ni Choileain
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
| | - Anna Williams
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
| | - Anna Richards
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Anne L. Astier
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
14
|
Tsai YG, Niu DM, Yang KD, Hung CH, Yeh YJ, Lee CY, Lin CY. Functional defects of CD46-induced regulatory T cells to suppress airway inflammation in mite allergic asthma. J Transl Med 2012; 92:1260-9. [PMID: 22751347 DOI: 10.1038/labinvest.2012.86] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Defective recruitment of regulatory T cells (Treg) function to the airway is important in the pathogenesis of allergic asthma. Complement regulatory protein (CD46) is a newly defined costimulatory molecule for Treg activation, which together with IL-10/granzyme B production may aid in suppressing asthmatic inflammation. This study examines chemotaxis and adhesion molecule expression on CD3/CD46-activated CD4(+) T cells (Tregs) from patients with and without asthma to suppress mite allergen-induced respiratory epithelial cells inflammation and to elucidate the mechanism of CD46-mediated Treg activation. Diminished IL-10/granzyme B and CCR4 expression from CD3/CD46-activated Tregs appeared in asthmatic subjects. CD3/CD46-activated Tregs from asthma patients co-cultured with BEAS-2B cells suppressed Dermatophagoides pteronyssinus 2 induced nuclear factor-κB/p65 by cell contact inhibition. Decreased interaction of CD3/CD46-mediated Tregs and BEAS-2B cells from asthmatics was associated with downregulated phosphorylation of protein kinase B (AKT) expression. Results provide the first evidence that decreased interaction between CD46-mediated Tregs and lung epithelial cells with less IL-10/granzyme B production may cause airway inflammation in allergic asthma.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- Department of Pediatrics, Changhua Christian Hospital, Changhua, Taiwan
| | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
The interaction between T cells and APCs bearing cognate antigen results in the formation of an immunological synapse (IS). During this process, many receptors and signaling proteins segregate to regions proximal to the synapse. This protein movement is thought to influence T cell function. However, some proteins are transported away from the IS, which is controlled in part by ERM family proteins. Tim-1 is a transmembrane protein with co-stimulatory functions that is found on many immune cells, including T cells. However, the expression pattern of Tim-1 on T cells upon activation by APCs has not been explored. Interestingly, in this study we demonstrate that the majority of Tim-1 on activated T cells is excluded from the IS. Tim-1 predominantly resides outside of the IS, and structure/function studies indicate that the cytoplasmic tail influences Tim-1 polarization. Specifically, a putative ERM binding motif (KRK 244-246) in the Tim-1 cytoplasmic tail appears necessary for proper Tim-1 localization. Furthermore, mutation of the KRK motif results in enhanced early tyrosine phosphorylation downstream of TCR/CD28 stimulation upon ectopic expression of Tim-1. Paradoxically however, the KRK motif is necessary for Tim-1 co-stimulation of NFAT/AP-1 activation and co-stimulation of cytokine production. This work reveals unexpected complexity underlying Tim-1 localization and suggests potentially novel mechanisms by which Tim-1 modulates T cell activity.
Collapse
Affiliation(s)
- Jean Lin
- University of Pittsburgh Medical Scientist Training Program and Graduate Program in Immunology, Pittsburgh, 15261, USA
| | - Leo Chen
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, 15261, USA
| |
Collapse
|
16
|
Kickler K, Maltby K, Ni Choileain S, Stephen J, Wright S, Hafler DA, Jabbour HN, Astier AL. Prostaglandin E2 affects T cell responses through modulation of CD46 expression. THE JOURNAL OF IMMUNOLOGY 2012; 188:5303-10. [PMID: 22544928 DOI: 10.4049/jimmunol.1103090] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The ubiquitous protein CD46, a regulator of complement activity, promotes T cell activation and differentiation toward a regulatory Tr1-like phenotype. The CD46-mediated differentiation pathway is defective in several chronic inflammatory diseases, underlying the importance of CD46 in controlling T cell function and the need to understand its regulatory mechanisms. Using an RNA interference-based screening approach in primary T cells, we have identified that two members of the G protein-coupled receptor kinases were involved in regulating CD46 expression at the surface of activated cells. We have investigated the role of PGE(2), which binds to the E-prostanoid family of G protein-coupled receptors through four subtypes of receptors called EP 1-4, in the regulation of CD46 expression and function. Conflicting roles of PGE(2) in T cell functions have been reported, and the reasons for these apparent discrepancies are not well understood. We show that addition of PGE(2) strongly downregulates CD46 expression in activated T cells. Moreover, PGE(2) differentially affects T cell activation, cytokine production, and phenotype depending on the activation signals received by the T cells. This was correlated with a distinct pattern of the PGE(2) receptors expressed, with EP4 being preferentially induced by CD46 activation. Indeed, addition of an EP4 antagonist could reverse the effects observed on cytokine production after CD46 costimulation. These data demonstrate a novel role of the PGE(2)-EP4 axis in CD46 functions, which might at least partly explain the diverse roles of PGE(2) in T cell functions.
Collapse
Affiliation(s)
- Karoline Kickler
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Adams WC, Berenson RJ, Karlsson Hedestam GB, Lieber A, Koup RA, Loré K. Attenuation of CD4+ T-cell function by human adenovirus type 35 is mediated by the knob protein. J Gen Virol 2012; 93:1339-1344. [PMID: 22357750 DOI: 10.1099/vir.0.039222-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The complement-regulatory protein CD46 is the primary receptor for human adenovirus type 35 (HAdV-35) and can regulate human immune-cell activation. CD4(+) T-cells are critical for initiating and maintaining adaptive immunity elicited by infection or vaccination. It was reported previously that HAdV-35 can bind these cells and suppress their activation. The data reported here demonstrate that recombinant trimeric HAdV-35 knob proteins alone can induce CD46 receptor downregulation and inhibit interleukin-2 production and proliferation of human CD4(+) T-cells in vitro similarly to mAbs specific to the CD46 region bound by HAdV-35 knobs. A mutant knob protein with increased affinity for CD46 compared with the wild-type knob caused equivalent effects. In contrast, a CD46-binding-deficient mutant knob protein did not inhibit T-cell activation. Thus, the capacity of HAdV-35 to attenuate human CD4(+) T-cell activation depends predominantly on knob interactions with CD46 and can occur independently of infection.
Collapse
Affiliation(s)
- William C Adams
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Richard A Koup
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | - Karin Loré
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Choileain SN, Astier AL. CD46 processing: a means of expression. Immunobiology 2012; 217:169-75. [PMID: 21742405 PMCID: PMC4363545 DOI: 10.1016/j.imbio.2011.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/06/2011] [Accepted: 06/13/2011] [Indexed: 12/15/2022]
Abstract
CD46 is a ubiquitously expressed type I transmembrane protein, first identified as a regulator of complement activation, and later as an entry receptor for a variety of pathogens. The last decade has also revealed the role of CD46 in regulating the adaptive immune response, acting as an additional costimulatory molecule for human T cells and inducing their differentiation into Tr1 cells, a subset of regulatory T cells. Interestingly, CD46 regulatory pathways are defective in T cells from patients with multiple sclerosis, asthma and rheumatoid arthritis, illustrating its importance in regulating T cell homeostasis. Indeed, CD46 expression at the cell surface is tightly regulated in many different cell types, highlighting its importance in several biological processes. Notably, CD46 is the target of enzymatic processing, being cleaved by metalloproteinases and by the presenilin/gamma secretase complex. This processing is required for its functions, at least in T cells. This review will summarize the latest updates on the regulation of CD46 expression and on its effects on T cell activation.
Collapse
Affiliation(s)
- Siobhan Ni Choileain
- MRC Centre for Inflammation Research, University of Edinburgh, UK
- Centre for MS Research, University of Edinburgh, UK
| | - Anne L Astier
- MRC Centre for Inflammation Research, University of Edinburgh, UK
- Centre for MS Research, University of Edinburgh, UK
| |
Collapse
|
19
|
Adenovirus type-35 vectors block human CD4+ T-cell activation via CD46 ligation. Proc Natl Acad Sci U S A 2011; 108:7499-504. [PMID: 21502499 DOI: 10.1073/pnas.1017146108] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recombinant adenoviruses (rAds) based on types 5 (rAd5) and 35 (rAd35) have emerged as important vaccine delivery vectors in clinical testing for a variety of pathogens. A major difference between these vectors is their binding to cellular receptors used for infection. Whereas rAd5 binds coxsackie-adenovirus receptor (CAR), rAd35 binds the complement regulatory protein CD46. Although rAd35 infected and phenotypically matured human blood dendritic cells (DCs) more efficiently than rAd5, we show here that rAd35 markedly suppressed DC-induced activation of naive CD4(+) T cells. rAd35 specifically blocked both DCs and anti-CD3/CD28 mAb-induced naive T-cell proliferation and IL-2 production. This effect was also observed in CD4(+) memory T cells but to a lesser extent. The suppression occurred by rAd35 binding to CD46 on T cells and was independent of infection. CD46 engagement with mAb mimicked the effects of rAd35 and also led to deficient NF-κB nuclear translocation. In contrast, rAd5 and rAd35 vectors with ablated CD46 binding did not inhibit T-cell activation. Our findings provide insights into the basic biology of adenoviruses and indicate that CD46 binding may have an impact on the generation of primary CD4(+) T-cell responses by Ad35.
Collapse
|
20
|
Abstract
CD46 was discovered in 1986 during a search for novel C3b-binding proteins. CD46 is expressed ubiquitously and functions as a co-factor in the factor I-mediated proteolytic cleavage of C3b and C4b. Its vital role in preventing complement deposition on host tissue is underpinned by the fact that deficiency of CD46 is a predisposing factor for numerous disease conditions arising from complement-mediated 'self-attack'. However, in the last 10 years, it has become apparent that CD46 is also heavily involved in a new and somewhat surprising functional aspect of the complement system: the down-modulation of adaptive T helper type 1 (Th1) immune responses by regulating the production of interferon (IFN)-γ versus interleukin (IL)-10 within these cells. Specifically, this latter function of CD46 is a tantalizing discovery - it may not only have delivered the explanation as to why so many pathogens use and abuse CD46 as cell entry receptor but clearly has important clinical implications for the better understanding of Th1-mediated disease states and novel therapeutic approaches for their amelioration. Here, we summarize and discuss the current knowledge about CD46 and its expanding roles in the immune system.
Collapse
Affiliation(s)
- J Cardone
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | | | | |
Collapse
|
21
|
Choileain SN, Astier AL. CD46 plasticity and its inflammatory bias in multiple sclerosis. Arch Immunol Ther Exp (Warsz) 2011; 59:49-59. [PMID: 21267793 PMCID: PMC4363543 DOI: 10.1007/s00005-010-0109-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 09/16/2010] [Indexed: 01/13/2023]
Abstract
Known as a link to the adaptive immune system, a complement regulator, a "pathogen magnet" and more recently as an inducer of autophagy, CD46 is the human receptor that refuses to be put in a box. This review summarizes the current roles of CD46 during immune responses and highlights the role of CD46 as both a promoter and attenuator of the immune response. In patients with multiple sclerosis (MS), CD46 responses are overwhelmingly pro-inflammatory with notable defects in cytokine and chemokine production. Understanding the role of CD46 as an inflammatory regulator is a distant goal considering the darkness in which its regulatory mechanisms reside. Further research into the regulation of CD46 expression through its internalization and processing will undoubtedly extend our knowledge of how the balance is tipped in favor of inflammation in MS patients.
Collapse
Affiliation(s)
- Siobhan Ni Choileain
- Institute for Immunology and Infection Research
- MRC Centre for Inflammation Research
- Centre for MS Research, University of Edinburgh, UK
| | - Anne L Astier
- Institute for Immunology and Infection Research
- MRC Centre for Inflammation Research
- Centre for MS Research, University of Edinburgh, UK
| |
Collapse
|
22
|
The Reorientation of T-Cell Polarity and Inhibition of Immunological Synapse Formation by CD46 Involves Its Recruitment to Lipid Rafts. J Lipids 2011; 2011:521863. [PMID: 21490803 PMCID: PMC3067059 DOI: 10.1155/2011/521863] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/01/2010] [Indexed: 12/14/2022] Open
Abstract
Many infectious agents utilize CD46 for infection of human cells, and therapeutic applications of CD46-binding viruses are now being explored. Besides mediating internalization to enable infection, binding to CD46 can directly alter immune function. In particular, ligation of CD46 by antibodies or by measles virus can prevent activation of T cells by altering T-cell polarity and consequently preventing the formation of an immunological synapse. Here, we define a mechanism by which CD46 reorients T-cell polarity to prevent T-cell receptor signaling in response to antigen presentation. We show that CD46 associates with lipid rafts upon ligation, and that this reduces recruitment of both lipid rafts and the microtubule organizing centre to the site of receptor cross-linking. These data combined indicate that polarization of T cells towards the site of CD46 ligation prevents formation of an immunological synapse, and this is associated with the ability of CD46 to recruit lipid rafts away from the site of TCR ligation.
Collapse
|
23
|
Ni Choileain S, Weyand NJ, Neumann C, Thomas J, So M, Astier AL. The dynamic processing of CD46 intracellular domains provides a molecular rheostat for T cell activation. PLoS One 2011; 6:e16287. [PMID: 21283821 PMCID: PMC3023775 DOI: 10.1371/journal.pone.0016287] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 12/10/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Adequate termination of an immune response is as important as the induction of an appropriate response. CD46, a regulator of complement activity, promotes T cell activation and differentiation towards a regulatory Tr1 phenotype. This Tr1 differentiation pathway is defective in patients with MS, asthma and rheumatoid arthritis, underlying its importance in controlling T cell function and the need to understand its regulatory mechanisms. CD46 has two cytoplasmic tails, Cyt1 and Cyt2, derived from alternative splicing, which are co-expressed in all nucleated human cells. The regulation of their expression and precise functions in regulating human T cell activation has not been fully elucidated. METHODOLOGY/PRINCIPAL FINDINGS Here, we first report the novel role of CD46 in terminating T cell activation. Second, we demonstrate that its functions as an activator and inhibitor of T cell responses are mediated through the temporal processing of its cytoplasmic tails. Cyt1 processing is required to turn T cell activation on, while processing of Cyt2 switches T cell activation off, as demonstrated by proliferation, CD25 expression and cytokine secretion. Both tails require processing by Presenilin/γSecretase (P/γS) to exert these functions. This was confirmed by expressing wild-type Cyt1 and Cyt2 tails and uncleavable mutant tails in primary T cells. The role of CD46 tails was also demonstrated with T cells expressing CD19 ectodomain-CD46 C-Terminal Fragment (CTF) fusions, which allowed specific triggering of each tail individually. CONCLUSIONS/SIGNIFICANCE We conclude that CD46 acts as a molecular rheostat to control human T cell activation through the regulation of processing of its cytoplasmic tails.
Collapse
Affiliation(s)
- Siobhan Ni Choileain
- Institute of Immunology and Infection Research, Edinburgh, United Kingdom
- Centre for Inflammation Research, Centre for Multiple Sclerosis Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Nathan J. Weyand
- BIO5 Institute and Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Christian Neumann
- Institute of Immunology and Infection Research, Edinburgh, United Kingdom
- Centre for Inflammation Research, Centre for Multiple Sclerosis Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Joelle Thomas
- Université Lyon 1, Lyon, CNRS, UMR5534, Centre de Génétique Moléculaire et Cellulaire, Villeurbanne, France
| | - Magdalene So
- BIO5 Institute and Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Anne L. Astier
- Institute of Immunology and Infection Research, Edinburgh, United Kingdom
- Centre for Inflammation Research, Centre for Multiple Sclerosis Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
24
|
Hawkins ED, Oliaro J. CD46 signaling in T cells: Linking pathogens with polarity. FEBS Lett 2010; 584:4838-44. [DOI: 10.1016/j.febslet.2010.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 08/19/2010] [Accepted: 09/01/2010] [Indexed: 10/19/2022]
|
25
|
Yao K, Graham J, Akahata Y, Oh U, Jacobson S. Mechanism of neuroinflammation: enhanced cytotoxicity and IL-17 production via CD46 binding. J Neuroimmune Pharmacol 2010; 5:469-78. [PMID: 20661655 PMCID: PMC4758197 DOI: 10.1007/s11481-010-9232-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 06/28/2010] [Indexed: 12/18/2022]
Abstract
The membrane co-factor protein CD46 is the cellular receptor for a number of pathogens including the human herpesvirus 6 (HHV-6). In addition to its function as an inhibitory complement receptor, engagement of CD46 in the context of T-cell receptor (TCR) signaling influences T-cell activation. Simultaneous cross-linking of the CD3/CD46 molecules led to differentiation of a unique population of CD4+ T-cell subset characterized by enhanced expressions of IFN-gamma, IL-10, granzyme B, adhesion molecule MAdCAM-1 (alpha-4-beta-7), surface-bound cytokine LIGHT, and chemokine receptor CCR9. Multiple sclerosis is a chronic inflammatory neurodegenerative disorder of the central nervous system (CNS) with unknown etiology. The HHV-6 is a candidate pathogen in MS and uses the CD46 molecule as its receptor. We hypothesize that binding of the HHV-6 glycoprotein to CD46 may trigger a pro-inflammatory response that could contribute to CNS tissue damage. To address this question, we examined immunological parameters such as proliferation, cytokine production and cytotoxic functions in CD4+ T cells of healthy individuals and MS patients following CD3/CD46 co-engagement by using anti-CD3 and anti-CD46 monoclonal antibodies as surrogates to mimic T-cell receptor and CD46 signaling. Our results demonstrated that CD3/CD46 cross-linking induced expression of IL-1beta and IL-17A in multiple sclerosis patient T cells. Additionally, increase in transient surface expression of lysosomal associated protein CD107a suggested enhanced CD4+ T-cell cytotoxic functions following CD3/CD46 co-stimulation. Collectively, this study demonstrated evidence to suggest a potential mechanism of virus-induced neuroinflammation that may be involved in MS disease pathogenesis.
Collapse
Affiliation(s)
- Karen Yao
- Viral Immunology Section, NINDS, NIH, Bethesda, MD 20892, USA, Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jhanelle Graham
- Viral Immunology Section, NINDS, NIH, Bethesda, MD 20892, USA
| | - Yoshimi Akahata
- Viral Immunology Section, NINDS, NIH, Bethesda, MD 20892, USA
| | - Unsong Oh
- Viral Immunology Section, NINDS, NIH, Bethesda, MD 20892, USA
| | - Steven Jacobson
- Viral Immunology Section, NINDS, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Macropinocytotic uptake and infection of human epithelial cells with species B2 adenovirus type 35. J Virol 2010; 84:5336-50. [PMID: 20237079 DOI: 10.1128/jvi.02494-09] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human adenovirus serotype 35 (HAdV-35; here referred to as Ad35) causes kidney and urinary tract infections and infects respiratory organs of immunocompromised individuals. Unlike other adenoviruses, Ad35 has a low seroprevalence, which makes Ad35-based vectors promising candidates for gene therapy. Ad35 utilizes CD46 and integrins as receptors for infection of epithelial and hematopoietic cells. Here we show that infectious entry of Ad35 into HeLa cells, human kidney HK-2 cells, and normal human lung fibroblasts strongly depended on CD46 and integrins but not heparan sulfate and variably required the large GTPase dynamin. Ad35 infections were independent of expression of the carboxy-terminal domain of AP180, which effectively blocks clathrin-mediated uptake. Ad35 infections were inhibited by small chemicals against serine/threonine kinase Pak1 (p21-activated kinase), protein kinase C (PKC), sodium-proton exchangers, actin, and acidic organelles. Remarkably, the F-actin inhibitor jasplakinolide, the Pak1 inhibitor IPA-3, or the sodium-proton exchange inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) blocked endocytic uptake of Ad35. Dominant-negative proteins or small interfering RNAs against factors driving macropinocytosis, including the small GTPase Rac1, Pak1, or the Pak1 effector C-terminal binding protein 1 (CtBP1), potently inhibited Ad35 infection. Confocal laser scanning microscopy, electron microscopy, and live cell imaging showed that Ad35 colocalized with fluid-phase markers in large endocytic structures that were positive for CD46, alphanu integrins, and also CtBP1. Our results extend earlier observations with HAdV-3 (Ad3) and establish macropinocytosis as an infectious pathway for species B human adenoviruses in epithelial and hematopoietic cells.
Collapse
|
27
|
Weyand NJ, Calton CM, Higashi DL, Kanack KJ, So M. Presenilin/gamma-secretase cleaves CD46 in response to Neisseria infection. THE JOURNAL OF IMMUNOLOGY 2009; 184:694-701. [PMID: 20018629 DOI: 10.4049/jimmunol.0900522] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD46 is a type I transmembrane protein with complement and T cell regulatory functions in human cells. CD46 has signaling and receptor properties in immune and nonimmune cells, many of which are dependent on the expression of cytoplasmic tail (cyt) isoforms cyt1 or cyt2. Little is known about how cyt1 and cyt2 mediate cellular responses. We show that CD46-cyt1 and CD46-cyt2 are substrates for presenilin/gamma-secretase (PS/gammaS), an endogenous protease complex that regulates many important signaling proteins through proteolytic processing. PS/gammaS processing of CD46 releases immunoprecipitable cyt1 and cyt2 tail peptides into the cell, is blocked by chemical inhibitors, and is prevented in dominant negative presenilin mutant cell lines. Two human pathogens, Neisseria gonorrhoeae and Neisseria meningitidis, stimulate PS/gammaS processing of CD46-cyt1 and CD46-cyt2. This stimulation requires type IV pili and PilT, the type IV pilus retraction motor, implying that mechanotransduction plays a role in this event. We present a model for PS/gammaS processing of CD46 that provides a mechanism by which signals are transduced via the cyt1 and cyt2 tails to regulate CD46-dependent cellular responses. Our findings have broad implications for understanding the full range of CD46 functions in infection and noninfection situations.
Collapse
Affiliation(s)
- Nathan J Weyand
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
| | | | | | | | | |
Collapse
|
28
|
Hawkins ED, Russell SM. Upsides and downsides to polarity and asymmetric cell division in leukemia. Oncogene 2009; 27:7003-17. [PMID: 19029941 DOI: 10.1038/onc.2008.350] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The notion that polarity regulators can act as tumor suppressors in epithelial cells is now well accepted. The function of these proteins in lymphocytes is less well explored, and their possible function as suppressors of leukemia has had little attention so far. We review the literature on lymphocyte polarity and the growing recognition that polarity proteins have an important function in lymphocyte function. We then describe molecular relationships between the polarity network and signaling pathways that have been implicated in leukemogenesis, which suggest mechanisms by which the polarity network might impact on leukemogenesis. We particularly focus on the possibility that disruption of polarity might alter asymmetric cell division (ACD), and that this might be a leukemia-initiating event. We also explore the converse possibility that leukemic stem cells might be produced or maintained by ACD, and therefore that Dlg, Scribble and Lgl might be important regulators of this process.
Collapse
Affiliation(s)
- E D Hawkins
- Immune Signalling Laboratory, Cancer Immunology, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | |
Collapse
|
29
|
Abstract
Measles virus (MV) was isolated in 1954 (Enders and Peeble 1954). It is among the most contagious of viruses and a leading cause of mortality in children in developing countries (Murray and Lopez 1997; Griffin 2001; Bryce et al. 2005). Despite intense research over decades on the biology and pathogenesis of the virus and the successful development in 1963 of an effective MV vaccine (Cutts and Markowitz 1994), cell entry receptor(s) for MV remained unidentified until 1993. Two independent studies showed that transfection of nonsusceptible rodent cells with human CD46 renders these cells permissive to infection with the Edmonston and Halle vaccine strains of measles virus (Dorig et al. 1993; Naniche et al. 1993). A key finding in these investigations was that MV binding and infection was inhibited by monoclonal and polyclonal antibodies to CD46. These reports established CD46 as a MV cell entry receptor. This chapter summarizes the role of CD46 in measles virus infection.
Collapse
Affiliation(s)
- C Kemper
- Division of Rheumatology, St. Louis, MO 63110, USA
| | | |
Collapse
|
30
|
Abstract
T cell cytoarchitecture differs dramatically depending on whether the cell is circulating within the bloodstream, migrating through tissues, or interacting with antigen-presenting cells. The transition between these states requires important signaling-dependent changes in actin cytoskeletal dynamics. Recently, analysis of actin-regulatory proteins associated with T cell activation has provided new insights into how T cells control actin dynamics in response to external stimuli and how actin facilitates downstream signaling events and effector functions. Among the actin-regulatory proteins that have been identified are nucleation-promoting factors such as WASp, WAVE2, and HS1; severing proteins such as cofilin; motor proteins such as myosin II; and linker proteins such as ezrin and moesin. We review the current literature on how signaling pathways leading from diverse cell surface receptors regulate the coordinated activity of these and other actin-regulatory proteins and how these proteins control T cell function.
Collapse
Affiliation(s)
- Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
31
|
Abstract
CD46 is a complement regulatory molecule expressed on every cell type, except for erythrocytes. While initially described as a regulator of complement activity, it later became a 'magnet for pathogens', binding to several viruses and bacteria. More recently, an alternative role for such complement molecules has emerged: they do regulate T-cell immunity, affecting T-cell proliferation and differentiation. In particular, CD46 stimulation induces Tr1 cells, regulatory T cells characterized by massive production of interleukin-10 (IL-10), a potent anti-inflammatory cytokine. Hence, CD46 is likely to control inflammation. Indeed, data from CD46 transgenic mice highlight a role for CD46 in inflammation, with antagonist roles depending on the cytoplasmic tail being expressed. Furthermore, recent data have shown that CD46 is defective in multiple sclerosis, IL-10 production being severely impaired in these patients. This lack of IL-10 production probably participates in the inflammation observed in patients with multiple sclerosis. This review will summarize the data on CD46 and T cells, and how CD46 is likely involved in multiple sclerosis.
Collapse
Affiliation(s)
- Anne L Astier
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
32
|
Subversion of CtBP1-controlled macropinocytosis by human adenovirus serotype 3. EMBO J 2008; 27:956-69. [PMID: 18323776 DOI: 10.1038/emboj.2008.38] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 02/13/2008] [Indexed: 12/22/2022] Open
Abstract
Endocytosis supports cell communication, growth, and pathogen infection. The species B human adenovirus serotype 3 (Ad3) is associated with epidemic conjunctivitis, and fatal respiratory and systemic disease. Here we show that Ad3 uses dynamin-independent endocytosis for rapid infectious entry into epithelial and haematopoietic cells. Unlike Ad5, which uses dynamin-dependent endocytosis, Ad3 endocytosis spatially and temporally coincided with enhanced fluid-phase uptake. It was sensitive to macropinocytosis inhibitors targeting F-actin, protein kinase C, the sodium-proton exchanger, and Rac1 but not Cdc42. Infectious Ad3 macropinocytosis required viral activation of p21-activated kinase 1 (PAK1) and the C-terminal binding protein 1 of E1A (CtBP1), recruited to macropinosomes. These macropinosomes also contained the Ad3 receptors CD46 and alpha v integrins. CtBP1 is a phosphorylation target of PAK1, and is bifunctionally involved in membrane traffic and transcriptional repression of cell cycle, cancer, and innate immunity pathways. Phosphorylation-defective S147A-CtBP1 blocked Ad3 but not Ad5 infection, providing a direct link between PAK1 and CtBP1. The data show that viruses induce macropinocytosis for infectious entry, a pathway used in antigen presentation and cell migration.
Collapse
|
33
|
Abstract
The differentiation, activation and expansion of T cells are dictated by their integrated response to a complex array of extracellular signals. Recent studies provide insight into how these signals are integrated and demonstrate a key role for cell shape in many aspects of T-cell signalling. T cells polarise during migration, antigen presentation and cell division to give rise to daughter cells that can have different cell fates. In each case, the polarity of the T cell facilitates this activity. This raises the possibility that adoption of a polarised state acts as a positive feedback mechanism to enhance responses to specific signals. Similarly, in asymmetric division of other cell types, the distribution of different molecules into each daughter can have profound consequences for proliferation, death and differentiation. The mechanisms of polarity regulation are far better understood in cells such as epithelial cells, neurons and neuronal precursors, and the fertilised zygote. With the emerging parallels between polarity in these cells and T cells, we should now be able to elucidate how polarity affects signalling and cell fate determination in T cells.
Collapse
Affiliation(s)
- Sarah Russell
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 2002, Australia and Center for MicroPhotonics, Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Victoria 3122, Australia
| |
Collapse
|
34
|
Shingai M, Ebihara T, Begum NA, Kato A, Honma T, Matsumoto K, Saito H, Ogura H, Matsumoto M, Seya T. Differential type I IFN-inducing abilities of wild-type versus vaccine strains of measles virus. THE JOURNAL OF IMMUNOLOGY 2007; 179:6123-33. [PMID: 17947687 DOI: 10.4049/jimmunol.179.9.6123] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Laboratory adapted and vaccine strains of measles virus (MV) induced type I IFN in infected cells. The wild-type strains in contrast induced it to a far lesser extent. We have investigated the mechanism for this differential type I IFN induction in monocyte-derived dendritic cells infected with representative MV strains. Laboratory adapted strains Nagahata and Edmonston infected monocyte-derived dendritic cells and activated IRF-3 followed by IFN-beta production, while wild-type MS failed to activate IRF-3. The viral IRF-3 activation is induced within 2 h, an early response occurring before protein synthesis. Receptor usage of CD46 or CD150 and nucleocapsid (N) protein variations barely affected the strain-to-strain difference in IFN-inducing abilities. Strikingly, most of the IFN-inducing strains possessed defective interference (DI) RNAs of varying sizes. In addition, an artificially produced DI RNA consisting of stem (the leader and trailer of MV) and loop (the GFP sequence) exhibited potential IFN-inducing ability. In this case, however, cytoplasmic introduction was needed for DI RNA to induce type I IFN in target cells. By gene-silencing analysis, DI RNA activated the RIG-I/MDA5-mitochondria antiviral signaling pathway, but not the TLR3-TICAM-1 pathway. DI RNA-containing strains induced IFN-beta mRNA within 2 h while the same recombinant strains with no DI RNA required >12 h postinfection to attain similar levels of IFN-beta mRNA. Thus, the stem-loop structure, rather than full genome replication or specific internal sequences of the MV genome, is required for an early phase of type I IFN induction by MV in host cells.
Collapse
Affiliation(s)
- Masashi Shingai
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). In the recent years, accumulating evidence has supported an immunosuppressive role for regulatory T cells (Tregs). Most studies in the context of autoimmunity have focused on the defects of the CD4+CD25 high Tregs. However, we recently demonstrated an altered function of Tr1 Treg cells in MS, characterized by a lack of IL-10 secretion. Therefore, several major regulatory T cell defects are involved in human autoimmune disease. Hence, the induction of Tregs or the stimulation of Treg activity may be beneficial for the treatment of such diseases.
Collapse
Affiliation(s)
- Anne L Astier
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
36
|
Abstract
The complement system is an important component of the innate immune system and a modulator of adaptive immunity. The entire complement system is focused on C3 and C5. Thus, there are proteins that activate C3 and C5, those that regulate this activation, and those that transduce the effects of C3 and C5 activation products; each can affect the kidney in renal injury. The normal kidney has the inherent capacity to protect itself from complement activation through cellular expression of decay-accelerating factor, membrane cofactor protein (in human beings), and Crry (in rodents). In addition, plasma factor H protects vascular spaces in the kidney. Although the main function of these proteins is to limit complement activation, there is now considerable evidence that they can transduce signals on engagement in immune cells. The G-protein-coupled 7-span transmembrane receptors for C3a and C5a, and the integral membrane complement receptors (CR) for C3b, iC3b, and C3dg, are expressed outside the kidney, particularly in cells of hematopoietic and immune lineage. These are important in renal injury through their infiltration of the kidney and/or by affecting kidney-directed immune responses. There is mounting evidence that intrinsic glomerular and tubular cell C3aR and C5aR expression and activation also can affect renal injury. CR1 on podocytes and the beta2 integrins CR3 and CR4 in kidney dendritic cells have functions that remain poorly defined. Cells of the kidney also have the capacity to produce and activate their own complement proteins. Thus, intrinsic renal cells express decay-accelerating factor, membrane cofactor protein, Crry, C3aR, C5aR, CR1, CR3, and CR4. These can be engaged by C3 and C5 activation products derived from systemic and local pools in renal injury. Given their capacity to provide signals that influence kidney cellular behavior, their activation can have substantial effects in renal injury. Defining these in a cell- and disease-specific fashion is an exciting challenge for future research.
Collapse
Affiliation(s)
- Tipu S Puri
- Section of Nephrology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
37
|
Membrane cofactor protein (MCP, CD46) binding to clinical isolates of Streptococcus pyogenes: binding to M type 18 strains is independent of Emm or Enn proteins. Mol Immunol 2007; 44:3571-9. [PMID: 17467801 DOI: 10.1016/j.molimm.2007.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Accepted: 03/11/2007] [Indexed: 01/20/2023]
Abstract
The complement regulatory protein CD46 (MCP, membrane cofactor protein) is used as a cell receptor by a number of bacterial and viral pathogens, including Streptococcus pyogenes (Group A Streptococci). The highly variable M (Emm) proteins are virulence factors of S. pyogenes, and Emm proteins of serotypes 5, 6 or 22 are able of binding to CD46, thus mediating the binding of Streptococci to human cells. In this work, using a soluble construction encompassing the extracellular domain of human CD46, we have analyzed its binding to clinical isolates of S. pyogenes, including isolates of the M types 1, 3 and 18 that are frequently found in invasive infections or rheumatic fever. Our data show a strong binding of CD46 to bacteria of M types 1, 3, 8, 18, 24, 28, 29, 31 and 78; weak binding to M6 and M29 and no binding to M types 11, 12, M27 or M30. Surprisingly, CD46 bound to isogenic mutants of one clinical M18 isolate lacking the Emm protein or Emm and the Emm-related protein Enn, regardless of having capsule or not. In addition, these isogenic mutants bound to keratinocytes in a CD46-dependent manner, confirming the role of CD46 as one of the cell receptors for Group A Streptococci. Furthermore, CD46 did not bind to a recombinant Emm 18 construct, confirming that Emm is not involved in CD46 binding to M18 bacteria. Emm-dependent and -independent CD46 binding of clinical isolates of Streptococci confirms the importance of CD46 as a cell target that might confer pathogens some biological advantages over the host.
Collapse
|
38
|
Sakurai F, Akitomo K, Kawabata K, Hayakawa T, Mizuguchi H. Downregulation of human CD46 by adenovirus serotype 35 vectors. Gene Ther 2007; 14:912-9. [PMID: 17377598 DOI: 10.1038/sj.gt.3302946] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human CD46 (membrane cofactor protein), which serves as a receptor for a variety of pathogens, including strains of measles virus, human herpesvirus type 6 and Neisseria, is rapidly downregulated from the cell surface following infection by these pathogens. Here, we report that replication-incompetent adenovirus (Ad) serotype 35 (Ad35) vectors, which belong to subgroup B and recognize human CD46 as a receptor, downregulate CD46 following infection. A decline in the surface expression of CD46 in human peripheral blood mononuclear cells was detectable 6 h after infection, and reached maximum (72%) 12 h after infection. Ad35 vector-induced downregulation of surface CD46 levels gradually recovered after the removal of Ad35 vectors, however, complete recovery of CD46 expression was not observed even at 96 h after removal. The surface expression of CD46 was also reduced after incubation with fiber-substituted Ad serotype 5 (Ad5) vectors bearing Ad35 fiber proteins, ultraviolet-irradiated Ad35, vectors and recombinant Ad35 fiber knob proteins; in contrast, conventional Ad5 vectors did not induce surface CD46 downregulation, suggesting that the fiber knob protein of Ad35 plays a crucial role in the downregulation of surface CD46 density. These results have important implications for gene therapy using CD46-utilizing Ad vectors and for the pathogenesis of Ads that interact with CD46.
Collapse
Affiliation(s)
- F Sakurai
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | |
Collapse
|
39
|
Research Highlights. Nat Immunol 2007. [DOI: 10.1038/ni0107-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Leavy O. Which signal to respond to? Nat Rev Immunol 2007. [DOI: 10.1038/nri2007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|