1
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
2
|
Pöhnl M, Trollmann MFW, Böckmann RA. Nonuniversal impact of cholesterol on membranes mobility, curvature sensing and elasticity. Nat Commun 2023; 14:8038. [PMID: 38081812 PMCID: PMC10713574 DOI: 10.1038/s41467-023-43892-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Biological membranes, composed mainly of phospholipids and cholesterol, play a vital role as cellular barriers. They undergo localized reshaping in response to environmental cues and protein interactions, with the energetics of deformations crucial for exerting biological functions. This study investigates the non-universal role of cholesterol on the structure and elasticity of saturated and unsaturated lipid membranes. Our study uncovers a highly cooperative relationship between thermal membrane bending and local cholesterol redistribution, with cholesterol showing a strong preference for the compressed membrane leaflet. Remarkably, in unsaturated membranes, increased cholesterol mobility enhances cooperativity, resulting in membrane softening despite membrane thickening and lipid compression caused by cholesterol. These findings elucidate the intricate interplay between thermodynamic forces and local molecular interactions that govern collective properties of membranes.
Collapse
Affiliation(s)
- Matthias Pöhnl
- Computational Biology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marius F W Trollmann
- Computational Biology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Erlangen National High Perfomance Computing Center (NHR@FAU), Erlangen, Germany
| | - Rainer A Böckmann
- Computational Biology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- Erlangen National High Perfomance Computing Center (NHR@FAU), Erlangen, Germany.
| |
Collapse
|
3
|
Spiegel F, Trollmann MFW, Kara S, Pöhnl M, Brandner AF, Nimmerjahn F, Lux A, Böckmann RA. Role of lipid nanodomains for inhibitory FcγRIIb function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540011. [PMID: 37214871 PMCID: PMC10197649 DOI: 10.1101/2023.05.09.540011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The inhibitory Fcγ receptor FcγRIIb is involved in immune regulation and is known to localize to specific regions of the plasma membrane called lipid rafts. Previous studies suggested a link between the altered lateral receptor localization within the plasma membrane and the functional impairment of the FcγRIIb-I232T variant that is associated with systemic lupus erythematosus. Here, we conducted microsecond all-atom molecular dynamics simulations and IgG binding assays to investigate the lipid nano-environment of FcγRIIb monomers and of the FcγRIIb-I232T mutant within a plasma membrane model, the orientation of the FcγRIIb ectodomain, and its accessibility to IgG ligands. In contrast to previously proposed models, our simulations indicated that FcγRIIb does not favor a cholesterol- or a sphingolipid-enriched lipid environment. Interestingly, cholesterol was depleted for all studied FcγRIIb variants within a 2-3 nm environment of the receptor, counteracting the usage of raft terminology for models on receptor functionality. Instead, the receptor interacts with lipids that have poly-unsaturated fatty acyl chains and with (poly-) anionic lipids within the cytosolic membrane leaflet. We also found that FcγRIIb monomers adopt a conformation that is not suitable for binding to its IgG ligand, consistent with a lack of detectable binding of monomeric IgG in experiments on primary immune cells. However, our results propose that multivalent IgG complexes might stabilize FcγRIIb in a binding-competent conformation. We suggest differences in receptor complex formation within the membrane as a plausible cause of the altered membrane localization or clustering and the altered suppressive function of the FcγRIIb-I232T variant.
Collapse
Affiliation(s)
- Franziska Spiegel
- Computational Biology, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
| | - Marius F W Trollmann
- Computational Biology, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
- Erlangen National High-Performance Computing Center (NHR@FAU)
| | - Sibel Kara
- Institute of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
| | - Matthias Pöhnl
- Computational Biology, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
| | - Astrid F Brandner
- Computational Biology, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
- Current address: Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
| | - Anja Lux
- Institute of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
| | - Rainer A Böckmann
- Computational Biology, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91058, Germany
- Erlangen National High-Performance Computing Center (NHR@FAU)
| |
Collapse
|
4
|
Summerfield A, Gerber H, Schmitt R, Liniger M, Grazioli S, Brocchi E. Relationship between neutralizing and opsonizing monoclonal antibodies against foot-and-mouth disease virus. Front Vet Sci 2022; 9:1033276. [PMID: 36311653 PMCID: PMC9597200 DOI: 10.3389/fvets.2022.1033276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/04/2022] Open
Abstract
Previous studies demonstrated that polyclonal antibodies against foot-and-mouth disease virus (FMDV) generated by vaccination can mediate immune functions not only through virus neutralization but also through promoting virus uptake by macrophages and dendritic cells that are otherwise resistant to FMDV infection. This causes abortive infections resulting in activation, enhanced antigen presentation but also cell death. Here we report the use of RAW264.7 cells representing a murine macrophage cells line to characterize opsonizing functions of a collection of monoclonal antibodies (mAbs) against FMDV O and A serotypes. We demonstrate that all neutralizing immunoglobulin G isotype mAbs are able to opsonize FMDV resulting in increased cell death of RAW264.7 cells. In contrast, neutralizing IgM antibodies did not possess this activity. Opsonization was observed with broader reactivity within the serotype when compared to neutralization. Importantly, the anti-O serotype D9 mAb reacting with the continuous epitope within the G-H loop of VP1 that contains the RGD binding site of FMDV, opsonized several FMDV serotypes despite its restricted neutralizing activity within the O serotype. Furthermore, by generating RAW264.7 cells expressing bovine CD32, an easy-to-use cell-based assay system to test for bovine antibody-dependent enhanced infection of FMDV was generated and tested with a collection of sera. The data indicate that opsonizing titers correlated better with vaccine dose when compared to neutralizing titers. On the other hand, neutralization and opsonization titers were similar predictive of protection. We conclude that low avidity interactions are sufficient to mediate Fcγ receptor-mediated immune functions that could contribute to protective immune responses against FMDV.
Collapse
Affiliation(s)
- Artur Summerfield
- Institute of Virology and Immunology, Köniz, Switzerland,Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland,*Correspondence: Artur Summerfield
| | - Heidi Gerber
- Institute of Virology and Immunology, Köniz, Switzerland
| | - Rebeka Schmitt
- Institute of Virology and Immunology, Köniz, Switzerland
| | | | - Santina Grazioli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Emiliana Brocchi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| |
Collapse
|
5
|
Wang J, Li Z, Xu L, Yang H, Liu W. Transmembrane domain dependent inhibitory function of FcγRIIB. Protein Cell 2018; 9:1004-1012. [PMID: 29497990 PMCID: PMC6251803 DOI: 10.1007/s13238-018-0509-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/22/2017] [Indexed: 01/26/2023] Open
Abstract
FcγRIIB, the only inhibitory IgG Fc receptor, functions to suppress the hyper-activation of immune cells. Numerous studies have illustrated its inhibitory function through the ITIM motif in the cytoplasmic tail of FcγRIIB. However, later studies revealed that in addition to the ITIM, the transmembrane (TM) domain of FcγRIIB is also indispensable for its inhibitory function. Indeed, recent epidemiological studies revealed that a non-synonymous single nucleotide polymorphism (rs1050501) within the TM domain of FcγRIIB, responsible for the I232T substitution, is associated with the susceptibility to systemic lupus erythematosus (SLE). In this review, we will summarize these epidemiological and functional studies of FcγRIIB-I232T in the past few years, and will further discuss the mechanisms accounting for the functional loss of FcγRIIB-I232T. Our review will help the reader gain a deeper understanding of the importance of the TM domain in mediating the inhibitory function of FcγRIIB and may provide insights to a new therapeutic target for the associated diseases.
Collapse
Affiliation(s)
- Junyi Wang
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zongyu Li
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Liling Xu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, 400 Technology Square, Cambridge, MA, 02139, USA.
| | - Hengwen Yang
- The First Affiliate Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, 510632, China.
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Zwozdesky MA, Fei C, Lillico DME, Stafford JL. Imaging flow cytometry and GST pulldown assays provide new insights into channel catfish leukocyte immune-type receptor-mediated phagocytic pathways. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:126-138. [PMID: 27984101 DOI: 10.1016/j.dci.2016.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 06/06/2023]
Abstract
Channel catfish (Ictalurus punctatus) leukocyte immune-type receptors (IpLITRs) control various innate immune cell effector responses including the phagocytic process. This large immunoregulatory receptor family also consists of multiple receptor-types with variable signaling abilities that is dependent on their inherent or acquired tyrosine-containing cytoplasmic tail (CYT) regions. For example, IpLITR 2.6b associates with the immunoreceptor tyrosine-based activation motif (ITAM)-containing adaptor molecule IpFcRγ-L, and when expressed in mammalian cells it activates phagocytosis using a similar profile of intracellular signaling mediators that also regulate the prototypical mammalian Fc receptor (FcR) phagocytic pathway. Alternatively, IpLITR 1.1b contains a long tyrosine-containing CYT with multifunctional capabilities including both inhibitory and stimulatory actions. Recently, we demonstrated that IpLITR 1.1b activates a unique phagocytic pathway involving the generation of multiple plasma membrane extensions that rapidly capture extracellular targets and secure them on the cell surface in phagocytic cup-like structures. Occasionally, these captured targets are completely engulfed albeit at a significantly lower rate than what was observed for IpLITR 2.6b. While this novel IpLITR 1.1b phagocytic activity is insensitive to classical blockers of phagocytosis, its distinct target capture and engulfment actions depend on the engagement of the actin polymerization machinery. However, it is not known how this protein translates target recognition into intracellular signaling events during this atypical mode of phagocytosis. Using imaging flow cytometry and GST pulldown assays, the aims of this study were to specifically examine what regions of the IpLITR 1.1b CYT trigger phagocytosis and to establish what profile of intracellular signaling molecules likely participate in its actions. Our results show that in stably transfected AD293 cells, the membrane proximal and distal CYT segments of IpLITR 1.1b independently regulate its phagocytic activities. These CYT regions were also shown to differentially recruit various SH2 domain-containing intracellular mediators, which provides new information about the dynamic immunoregulatory abilities of IpLITR 1.1b. Overall, this work further advances our understanding of how certain immunoregulatory receptor-types link extracellular target binding events to the actin polymerization machinery during a non-classical mode of phagocytosis.
Collapse
Affiliation(s)
- Myron A Zwozdesky
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Chenjie Fei
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Dustin M E Lillico
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
FcγRIIa requires lipid rafts, but not co-localization into rafts, for effector function. Inflamm Res 2012; 62:37-43. [PMID: 22945762 DOI: 10.1007/s00011-012-0548-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/23/2012] [Accepted: 08/16/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE To determine if receptor localization into lipid rafts, or the lipid rafts themselves, are important for FcγRIIa effector functions. MATERIAL Wild-type FcγRIIa or mutant FcγRIIa(C208A) that does not translocate to lipid rafts were transfected into Chinese hamster ovary (CHO) cells which have been shown to be reliable cells for studying FcγR function. TREATMENT Cells were treated with buffer or methyl-β-cyclodextrin (MβCD) to deplete cholesterol and dissolve the structure of lipid rafts. METHODS To evaluate lipid raft association, transfected CHO cells were lysed and centrifuged over a sucrose gradient. Fractions were run on SDS-PAGE and blotted for FcγRIIa or sphingolipid GM1 to illustrate the lipid raft fractions. Lateral mobility of GFP-tagged wild-type or mutant FcγRIIa was assessed using fluorescence recovery after photobleaching (FRAP) microscopy. Internalization of IgG-opsonized erythrocytes was assessed by fluorescence microscopy and uptake of heat-aggregated IgG (haIgG) was measured using flow cytometry. RESULTS We observed that FcγRIIa(C208A) did not localize into lipid rafts. However, the mutant FcγRIIa retained lateral mobility and effector function similar to wild-type FcγRIIa. However, mutant FcγRIIa function was abolished upon treatment with MβCD. CONCLUSIONS Lipid rafts provide an essential component required for effector activities independent of receptor localization.
Collapse
|
8
|
Gibson AW, Li X, Wu J, Baskin JG, Raman C, Edberg JC, Kimberly RP. Serine phosphorylation of FcγRI cytoplasmic domain directs lipid raft localization and interaction with protein 4.1G. J Leukoc Biol 2011; 91:97-103. [PMID: 22003208 DOI: 10.1189/jlb.0711368] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The high-affinity IgG receptor (CD64, FcγRI) has several special capacities, including the receptor-stimulated cleavage of the cell surface B cell-activating factor of the TNF superfamily (TNFSF13B). With the use of the yeast two-hybrid system, we and others have shown that FcγRI interacts with protein 4.1G (EPB41L2). Our mutational analyses identified two required 4.1G-interacting regions in the FcγRI CY and one FcγRI-interacting site in the C-terminus of protein 4.1G. Herein, we explore mechanism(s) that may regulate the interaction between protein 4.1G and FcγRI CY and influence FcγRI membrane mobility and function. We show that FcγRI CY interacts with protein 4.1G in vitro and that FcγRI coimmunoprecipitates protein 4.1G in freshly isolated human PBMC. With the use of immunostaining, we show that FcγRI colocalizes with protein 4.1G in unstimulated U937 cells, in which the FcγRI CY is constitutively serine-phosphorylated, but significant uncoupling occurs following FcγRI cross-linking, suggesting phosphoserine-regulated interaction. In vitro, protein 4.1G interacted preferentially with CK2-phosphorylated FcγRI CY, and compared with WT FcγRI, a nonphosphorylatable FcγRI mutant receptor was excluded from lipid rafts, suggesting a key role for protein 4.1G in targeting phosphorylated FcγRI to rafts. These data are consistent with a phosphoserine-dependent tethering role for protein 4.1G in maintaining FcγRI in lipid rafts and provide insight into the unique phosphoserine-based regulation of receptor signaling by FcγRI CY.
Collapse
Affiliation(s)
- Andrew W Gibson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | | | | | | | |
Collapse
|
9
|
Jin S, Zhou F, Katirai F, Li PL. Lipid raft redox signaling: molecular mechanisms in health and disease. Antioxid Redox Signal 2011; 15:1043-83. [PMID: 21294649 PMCID: PMC3135227 DOI: 10.1089/ars.2010.3619] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lipid rafts, the sphingolipid and cholesterol-enriched membrane microdomains, are able to form different membrane macrodomains or platforms upon stimulations, including redox signaling platforms, which serve as a critical signaling mechanism to mediate or regulate cellular activities or functions. In particular, this raft platform formation provides an important driving force for the assembling of NADPH oxidase subunits and the recruitment of other related receptors, effectors, and regulatory components, resulting, in turn, in the activation of NADPH oxidase and downstream redox regulation of cell functions. This comprehensive review attempts to summarize all basic and advanced information about the formation, regulation, and functions of lipid raft redox signaling platforms as well as their physiological and pathophysiological relevance. Several molecular mechanisms involving the formation of lipid raft redox signaling platforms and the related therapeutic strategies targeting them are discussed. It is hoped that all information and thoughts included in this review could provide more comprehensive insights into the understanding of lipid raft redox signaling, in particular, of their molecular mechanisms, spatial-temporal regulations, and physiological, pathophysiological relevances to human health and diseases.
Collapse
Affiliation(s)
- Si Jin
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | | | | | | |
Collapse
|
10
|
Vieth JA, Kim MK, Pan XQ, Schreiber AD, Worth RG. Differential requirement of lipid rafts for FcγRIIA mediated effector activities. Cell Immunol 2010; 265:111-9. [PMID: 20728077 PMCID: PMC2975250 DOI: 10.1016/j.cellimm.2010.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 05/11/2010] [Accepted: 07/28/2010] [Indexed: 01/18/2023]
Abstract
Immunoglobulin G (IgG) dependent activities are important in host defense and autoimmune diseases. Various cell types including macrophages and neutrophils contribute to pathogen destruction and tissue damage through binding of IgG to Fcγ receptors (FcγR). One member of this family, FcγRIIA, is a transmembrane glycoprotein known to mediate binding and internalization of IgG-containing targets. FcγRIIA has been observed to translocate into lipids rafts upon binding IgG-containing targets. We hypothesize that lipid rafts participate to different extents in binding and internalizing targets of different sizes. We demonstrate that disruption of lipid rafts with 8mM methyl-β-cyclodextrin (MβCD) nearly abolishes binding (91% reduction) and phagocytosis (60% reduction) of large IgG-coated targets. Conversely, binding and internalization of small IgG-complexes is less dependent on lipid rafts (49% and 17% inhibition at 8mM MβCD, respectively). These observations suggest that differences between phagocytosis and endocytosis may arise as early as the initial stages of ligand recognition.
Collapse
Affiliation(s)
- Joshua A. Vieth
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine, Toledo, OH 43614
| | - Moo-kyung Kim
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Xiao Qing Pan
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Alan D. Schreiber
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Randall G. Worth
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine, Toledo, OH 43614
| |
Collapse
|
11
|
Honda ZI, Suzuki T, Honda H. Identification of CENP-V as a novel microtubule-associating molecule that activates Src family kinases through SH3 domain interaction. Genes Cells 2009; 14:1383-94. [DOI: 10.1111/j.1365-2443.2009.01355.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
12
|
Puerta-Guardo H, Mosso C, Medina F, Liprandi F, Ludert JE, del Angel RM. Antibody-dependent enhancement of dengue virus infection in U937 cells requires cholesterol-rich membrane microdomains. J Gen Virol 2009; 91:394-403. [DOI: 10.1099/vir.0.015420-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
13
|
FcgammaRI ligation leads to a complex with BLT1 in lipid rafts that enhances rat lung macrophage antimicrobial functions. Blood 2009; 114:3316-24. [PMID: 19657115 PMCID: PMC2759654 DOI: 10.1182/blood-2009-01-199919] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukotriene (LT) B(4) is generated in response to engagement of the Fc gamma receptor (Fc gamma R) and potently contributes to Fc gamma R-mediated antimicrobial functions in pulmonary alveolar macrophages. In this study, we report that the LTB(4) receptor leukotriene B(4) receptor 1 (BLT1) redistributes from nonlipid raft (LR) to LR membrane microdomains upon immunoglobulin G-red blood cell, but not LTB(4), challenge. Cholesterol depletion to disrupt LRs abolished LTB(4)-induced enhancement of phagocytosis, microbicidal activity, and signaling. The dependence on LR integrity for BLT1 signaling correlated with formation of a complex consisting of BLT1, its primary coupled G protein G alpha i3, Src kinase, and Fc gamma RI within LRs. This association was dependent on Src-mediated phosphorylation of BLT1. These data identify a novel form of regulation in which engagement of a macrophage immunoreceptor recruits a stimulatory G protein-coupled receptor into a LR microdomain with resultant enhanced antimicrobial signaling.
Collapse
|
14
|
Serre-Beinier V, Bosco D, Zulianello L, Charollais A, Caille D, Charpantier E, Gauthier BR, Diaferia GR, Giepmans BN, Lupi R, Marchetti P, Deng S, Buhler L, Berney T, Cirulli V, Meda P. Cx36 makes channels coupling human pancreatic beta-cells, and correlates with insulin expression. Hum Mol Genet 2009; 18:428-39. [PMID: 19000992 PMCID: PMC2638800 DOI: 10.1093/hmg/ddn370] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previous studies have documented that the insulin-producing beta-cells of laboratory rodents are coupled by gap junction channels made solely of the connexin36 (Cx36) protein, and have shown that loss of this protein desynchronizes beta-cells, leading to secretory defects reminiscent of those observed in type 2 diabetes. Since human islets differ in several respects from those of laboratory rodents, we have now screened human pancreas, and islets isolated thereof, for expression of a variety of connexin genes, tested whether the cognate proteins form functional channels for islet cell exchanges, and assessed whether this expression changes with beta-cell function in islets of control and type 2 diabetics. Here, we show that (i) different connexin isoforms are differentially distributed in the exocrine and endocrine parts of the human pancreas; (ii) human islets express at the transcript level different connexin isoforms; (iii) the membrane of beta-cells harbors detectable levels of gap junctions made of Cx36; (iv) this protein is concentrated in lipid raft domains of the beta-cell membrane where it forms gap junctions; (v) Cx36 channels allow for the preferential exchange of cationic molecules between human beta-cells; (vi) the levels of Cx36 mRNA correlated with the expression of the insulin gene in the islets of both control and type 2 diabetics. The data show that Cx36 is a native protein of human pancreatic islets, which mediates the coupling of the insulin-producing beta-cells, and contributes to control beta-cell function by modulating gene expression.
Collapse
Affiliation(s)
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Laurence Zulianello
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Anne Charollais
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Dorothée Caille
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Eric Charpantier
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Benoit R. Gauthier
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Giuseppe R. Diaferia
- Islet Research Laboratory, The Whittier Institute for Diabetes, University of California San Diego, La Jolla, CA, USA
| | - Ben N. Giepmans
- Department of Cell Biology, University of Groningen, Groningen, The Netherlands
| | - Roberto Lupi
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Shaoping Deng
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Léo Buhler
- Surgical Research Unit, Department of Surgery
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Vincenzo Cirulli
- Islet Research Laboratory, The Whittier Institute for Diabetes, University of California San Diego, La Jolla, CA, USA
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| |
Collapse
|
15
|
Huynh KK, Gershenzon E, Grinstein S. Cholesterol accumulation by macrophages impairs phagosome maturation. J Biol Chem 2008; 283:35745-55. [PMID: 18955491 DOI: 10.1074/jbc.m806232200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Macrophages are key to the pathogenesis of atherosclerosis. They take up and store excessive amounts of cholesterol associated with modified low density lipoprotein, eventually becoming foam cells that display altered immune responsiveness. We studied the effects of cholesterol accumulation on phagosome formation and maturation, using lipid transport antagonists and cholesterol transport-deficient mutants. In macrophages treated with U18666A, a transport antagonist that prevents cholesterol exit from late endosomes/lysosomes, the early stages of maturation proceeded normally; phagosomes acquired Rab5, phosphatidylinositol 3-phosphate, and EEA1 and merged with LAMP-containing vesicles. However, fusion with lysosomes was impaired. Rab7, which is required for phagolysosome formation, was acquired by phagosomes but remained inactive. Maturation was also studied in fibroblasts from Niemann-Pick type C individuals that have defective cholesterol transport. Transfection of FcgammaIIA receptors was used to confer phagocytic capability to these fibroblasts. Niemann-Pick type C phagosomes failed to fuse with lysosomes, whereas wild type fibroblasts formed normal phagolysosomes. These findings indicate that cholesterol accumulation can have a detrimental effect on phagosome maturation by impairing the activation of Rab7, sequestering it and its effectors in cholesterol-enriched multilamellar compartments.
Collapse
Affiliation(s)
- Kassidy K Huynh
- Programme in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | |
Collapse
|
16
|
Beekman JM, van der Linden JA, van de Winkel JGJ, Leusen JHW. FcgammaRI (CD64) resides constitutively in lipid rafts. Immunol Lett 2008; 116:149-55. [PMID: 18207250 DOI: 10.1016/j.imlet.2007.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 12/05/2007] [Accepted: 12/06/2007] [Indexed: 01/24/2023]
Abstract
Cellular membranes contain microdomains known as 'lipid rafts' or detergent-insoluble microdomains (DRM), enriched in cholesterol and sphingolipids. DRM can play an important role in many cellular processes, including signal transduction, cytoskeletal organization, and pathogen entry. Many receptors like T cell receptors, B cell receptors and IgE receptors have been shown to reside in DRM. The majority of these receptors depend on multivalent ligand interaction to associate with these microdomains. We, here, study association between the high affinity IgG receptor, FcgammaRI (CD64), and membrane microdomains. FcgammaRI is a 72kDa type I glycoprotein that can mediate phagocytosis of opsonized pathogens, but can also effectively capture small immune complexes, and facilitates antigen presentation. We found FcgammaRI to predominantly reside within detergent-insoluble buoyant membranes, together with FcRgamma-chain, but independent of cross-linking ligand. With the use of confocal imaging, FcgammaRI was found to co-patch with GM1, a microdomain-enriched glycolipid. Depletion of cellular cholesterol, furthermore, modulated FcgammaRI-ligand interactions. These data indicated FcgammaRI to reside within lipid rafts without prior triggering of the receptor.
Collapse
Affiliation(s)
- Jeffrey M Beekman
- Immunotherapy Laboratory, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
17
|
Sohn HW, Pierce SK, Tzeng SJ. Live Cell Imaging Reveals that the Inhibitory FcγRIIB Destabilizes B Cell Receptor Membrane-Lipid Interactions and Blocks Immune Synapse Formation. THE JOURNAL OF IMMUNOLOGY 2008; 180:793-9. [DOI: 10.4049/jimmunol.180.2.793] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Colocalization of the IL-12 receptor and FcgammaRIIIa to natural killer cell lipid rafts leads to activation of ERK and enhanced production of interferon-gamma. Blood 2008; 111:4173-83. [PMID: 18174382 DOI: 10.1182/blood-2007-01-068908] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cells express an activating receptor for the Fc portion of IgG (FcgammaRIIIa) that mediates interferon (IFN)-gamma production in response to antibody (Ab)-coated targets. We have previously demonstrated that NK cells activated with interleukin-12 (IL-12) in the presence of immobilized IgG secrete 10-fold or more higher levels of IFN-gamma as compared with stimulation with either agent alone. We examined the intracellular signaling pathways responsible for this synergistic IFN-gamma production. NK cells costimulated via the FcR and the IL-12 receptor (IL-12R) exhibited enhanced levels of activated STAT4 and Syk as compared with NK cells stimulated through either receptor alone. Extracellular signal-regulated kinase (ERK) was also synergistically activated under these conditions. Studies with specific chemical inhibitors revealed that the activation of ERK was dependent on the activation of PI3-K, whose activation was dependent on Syk, and that sequential activation of these molecules was required for NK cell IFN-gamma production in response to FcR and IL-12 stimulation. Retroviral transfection of ERK1 into primary human NK cells substantially increased IFN-gamma production in response to immobilized IgG and IL-12, while transfection of human NK cells with a dominant-negative ERK1 abrogated IFN-gamma production. Confocal microscopy and cellular fractionation experiments revealed that FcgammaRIIIa and the IL-12R colocalized to areas of lipid raft microdomains in response to costimulation with IgG and IL-12. Chemical disruption of lipid rafts inhibited ERK signaling in response to costimulation and significantly inhibited IFN-gamma production. These data suggest that dual recruitment of FcgammaRIIIa and the IL-12R to lipid raft microdomains allows for enhanced activation of downstream signaling events that lead to IFN-gamma production.
Collapse
|
19
|
Nguyen HTT, Charrier-Hisamuddin L, Dalmasso G, Hiol A, Sitaraman S, Merlin D. Association of PepT1 with lipid rafts differently modulates its transport activity in polarized and nonpolarized cells. Am J Physiol Gastrointest Liver Physiol 2007; 293:G1155-65. [PMID: 17932227 DOI: 10.1152/ajpgi.00334.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The transporter PepT1, apically expressed in intestinal epithelial cells, is responsible for the uptake of di/tripeptides. PepT1 is also expressed in nonpolarized immune cells. Here we investigated the localization of PepT1 in lipid rafts in small intestinal brush border membranes (BBMs) and polarized and nonpolarized cells, as well as functional consequences of the association of PepT1 with lipid rafts. Immunoblot analysis showed the presence of PepT1 in low-density fractions isolated from mouse intestinal BBMs, polarized intestinal Caco2-BBE cells, and nonpolarized Jurkat cells by solubilization in ice-cold 0.5% Triton X-100 and sucrose gradient fractionation. PepT1 colocalized with lipid raft markers GM1 and N-aminopeptidase in intestinal BBMs and Caco2-BBE cell membranes. Disruption of lipid rafts with methyl-beta-cyclodextrin (MbetaCD) shifted PepT1 from low- to high-density fractions. Remarkably, we found that MbetaCD treatment increased PepT1 transport activity in polarized intestinal epithelia but decreased that in intestinal BBM vesicles and nonpolarized immune cells. Mutational analysis showed that phenylalanine 293, phenylalanine 297, and threonine 281 in transmembrane segment 7 of the human di/tripeptide transporter, hPepT1, are important for the targeting to lipid rafts and transport activity of hPepT1. In conclusion, the association of PepT1 with lipid rafts differently modulates its transport activity in polarized and nonpolarized cells.
Collapse
Affiliation(s)
- Hang Thi Thu Nguyen
- Dept. of Medicine, Division of Digestive Diseases, Emory Univ. School of Medicine, 615 Michael St., Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Mousavi SA, Sporstøl M, Fladeby C, Kjeken R, Barois N, Berg T. Receptor-mediated endocytosis of immune complexes in rat liver sinusoidal endothelial cells is mediated by FcgammaRIIb2. Hepatology 2007; 46:871-84. [PMID: 17680646 DOI: 10.1002/hep.21748] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Liver sinusoidal endothelial cells (LSECs) display a number of receptors for efficient uptake of potentially injurious molecules. The receptors for the Fc portion of immunoglobulin G (IgG) antibodies (FcgammaRs) regulate a number of physiological and pathophysiological events. We used reverse transcription polymerase chain reaction (RT-PCR) and Western blotting to determine the expression of different types of FcgammaRs in LSECs. Biochemical approaches and immunofluorescence microscopy were used to characterize the FcgammaR-mediated endocytosis of immune complexes (ICs). FcgammaRIIb2 was identified as the main receptor for the efficient uptake of ICs in LSECs. The receptor was shown to use the clathrin pathway for IC uptake; however, the association with lipid rafts may slow the rate of its internalization. Moreover, despite trafficking through lysosomal integral membrane protein-II (LIMP-II)-containing compartments, the receptor was not degraded. Finally, it was shown that the receptor recycles to the cell surface both with and without IC. CONCLUSION FcgammaRIIb2 is the main receptor for endocytosis of ICs in rat LSECs. Internalized ICs are degraded with slow kinetics, and IC internalization is not linked to receptor downregulation. After internalization, the receptor recycles to the cell surface both with and without ICs. Thus, FcgammaRIIb2 in rat LSECs is used as both a recycling receptor and a receptor for efficient IC clearance.
Collapse
|
22
|
Abstract
Oxidant stress, induced under a variety of conditions, is known to lead to the molecular reprogramming of the tissue-fixed macrophage. This reprogramming is associated with an altered response to subsequent inflammatory stimuli, such as lipopolysaccharide (LPS), leading to enhanced liberation of proinflammatory chemokines and cytokines. Due to this altered response, dysregulated immunity ensues, leading to the development of clinical syndromes such as multiple organ dysfunction syndrome (MODS). Although the mechanisms responsible for this altered macrophage activity by oxidant stress remains complex and poorly elucidated, it appears, based on recent research, that early and direct alterations within lipid rafts are responsible. This early and direct interaction with lipid rafts by oxidants leads to the mobilization of annexin VI from lipid raft constructs, leading to the release of calcium. This increased cytosolic concentration of this secondary messenger, in turn, results in the activation of calcium-dependent kinases, leading to further alterations in lipid raft lipids and eventually lipid raft proteins. Due to these lipid raft compositional changes, preassembly of receptor complexes occur, leading to enhanced proinflammatory activation. Within this review, the complexity of oxidant-induced reprogramming within the tissue fixed macrophage as currently understood is explained.
Collapse
Affiliation(s)
- Joseph Cuschieri
- University of Washington, Department of Surgery, Seattle, Washington 98104, USA.
| | | |
Collapse
|
23
|
Szabo G, Dolganiuc A, Dai Q, Pruett SB. TLR4, ethanol, and lipid rafts: a new mechanism of ethanol action with implications for other receptor-mediated effects. THE JOURNAL OF IMMUNOLOGY 2007; 178:1243-9. [PMID: 17237368 DOI: 10.4049/jimmunol.178.3.1243] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ethanol (EtOH) is the most widely abused substance in the United States, and it contributes to well-documented harmful (at high dosages) and beneficial (at low dosages) changes in inflammatory and immune responses. Lipid rafts have been implicated in the regulation and activation of several important receptor complexes in the immune system, including the TLR4 complex. Many questions remain about the precise mechanisms by which rafts regulate the assembly of these receptor complexes. Results summarized in this review indicate that EtOH acts by altering the LPS-induced redistribution of components of the TLR4 complex within the lipid raft and that this is related to changes in actin cytoskeleton rearrangement, receptor clustering, and subsequent signaling. EtOH provides an example of an immunomodulatory drug that acts at least in part by modifying lipid rafts, and it could represent a model to probe the relationships between rafts, receptor complexes, and signaling.
Collapse
Affiliation(s)
- Gyongyi Szabo
- University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | | | | | |
Collapse
|
24
|
García-García E, Brown EJ, Rosales C. Transmembrane Mutations to FcγRIIA Alter Its Association with Lipid Rafts: Implications for Receptor Signaling. THE JOURNAL OF IMMUNOLOGY 2007; 178:3048-58. [PMID: 17312151 DOI: 10.4049/jimmunol.178.5.3048] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Many immunoreceptors have been reported to associate with lipid rafts upon ligand binding. The way in which this association is regulated is still obscure. We investigated the roles for various domains of the human immunoreceptor FcgammaRIIA in regulating its association with lipid rafts by determining the resistance of unligated, or ligated and cross-linked, receptors to solubilization by the nonionic detergent Triton X-100, when expressed in RBL-2H3 cells. Deletion of the cytoplasmic domain, or destruction of the cytoplasmic palmitoylation site, had no effect on the association of the receptor with lipid rafts. A transmembrane mutant, A224S, lost the ability to associate with lipid rafts upon receptor cross-linking, whereas transmembrane mutants VA231-2MM and VVAL234-7GISF showed constitutive lipid raft association. Wild-type (WT) FcgammaRIIA and all transmembrane mutants activated Syk, regardless of their association with lipid rafts. WT FcgammaRIIA and mutants that associated with lipid rafts efficiently activated NF-kappaB, in an ERK-dependent manner. In contrast, WT FcgammaRIIA and the A224S mutant both presented efficient phagocytosis, while VA231-2MM and VVAL234-7GISF mutants presented lower phagocytosis, suggesting that phagocytosis may proceed independently of lipid raft association. These data identify the transmembrane domain of FcgammaRIIA as responsible for regulating its inducible association with lipid rafts and suggest that FcgammaRIIA-mediated responses, like NF-kappaB activation or phagocytosis, can be modulated by lipid raft association of the ligated receptor.
Collapse
Affiliation(s)
- Erick García-García
- Immunology Department, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City D.F.-04510, Mexico
| | | | | |
Collapse
|
25
|
Menzies D, Nair A, Meldrum KT, Fleming D, Barnes M, Lipworth BJ. Simvastatin does not exhibit therapeutic anti-inflammatory effects in asthma. J Allergy Clin Immunol 2006; 119:328-35. [PMID: 17141851 DOI: 10.1016/j.jaci.2006.10.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 10/08/2006] [Accepted: 10/11/2006] [Indexed: 01/27/2023]
Abstract
BACKGROUND Statins lower cholesterol and also exhibit anti-inflammatory properties. In vitro and animal studies have suggested they may be useful for the treatment of a number of inflammatory conditions. OBJECTIVE To evaluate the in vivo therapeutic potential of simvastatin as an anti-inflammatory agent in patients with asthma. METHODS Potential signal from treatment effect was optimized by withdrawing all anti-inflammatory treatment for the duration of the study. Participants received 1 month of daily simvastatin and 1 month of daily placebo in a randomized, double-blind crossover trial. A total of 16 patients completed per protocol. Asthmatic inflammation was evaluated by measuring exhaled tidal nitric oxide, alveolar nitric oxide, sputum and peripheral eosinophil count, methacholine hyperresponsiveness, salivary eosinophilic cationic protein, and C-reactive protein. Measurements of dynamic and static lung volumes and of cholesterol were also made. RESULTS After initial withdrawal of usual asthma medication, there was a 1.43 geometric mean fold increase (ie, 43% difference) in fraction of exhaled nitric oxide (95% CI, 1.15 to 1.78; P = .004). Compared with placebo, simvastatin led to a 0.86 geometric mean fold decrease (95% CI, 0.7 to 1.04; P = .15) in exhaled nitric oxide (ie, a 14% difference), and a -0.18 doubling dilution shift (95% CI, -1.90 to 1.55; P = 1.0) in methacholine hyperresponsiveness. There were no significant differences in other inflammatory outcomes, lung volumes, or airway resistance between simvastatin and placebo. Treatment with simvastatin led to a significant reduction (P < .005) of total and low-density lipoprotein cholesterol. CONCLUSION There is no evidence to suggest simvastatin has anti-inflammatory activity in patients with asthma. CLINICAL IMPLICATIONS Simvastatin is not useful for the treatment of asthma.
Collapse
Affiliation(s)
- Daniel Menzies
- Asthma and Allergy Research Group, Ninewells Hospital and Medical School, Dundee, UK
| | | | | | | | | | | |
Collapse
|
26
|
Honda ZI. Fcε- and Fcγ-receptor signaling in diseases. ACTA ACUST UNITED AC 2006; 28:365-75. [PMID: 17106671 DOI: 10.1007/s00281-006-0051-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 09/27/2006] [Indexed: 12/14/2022]
Abstract
It has become increasingly clear that receptors for the immunoglobulin Fc region play pivotal roles in immune homeostasis and disease. This review describes the fine regulation of the high-affinity IgE-receptor (FcepsilonRI) signaling, especially focusing on the early events that are coordinately regulated by Src family protein tyrosine kinases (PTKs), FcepsilonRI beta-subunit, and membrane lipid rafts. Because allergen-mediated FcepsilonRI cross-linking leads to the synthesis and release of a variety of proinflammatory mediators and cytokines, the duration and amplitude of the signal need to be strictly controlled, and the counterbalancing signaling is provided by specialized inhibitory receptors and molecules. However, recent work have revealed that Src family PTKs and FcepsilonRI beta-subunit transduce both positive and negative signaling with unexpectedly complex mechanisms. FcgammaRIIB exerts a unique inhibitory function on cell activation processes after the engagement of Fcgamma, FcepsilonRI and B cell receptors. Recent work has shown that FcgammaRIIB polymorphisms are associated with systemic lupus erythematosus, and that a transmembrane polymorphism in FcgammaRIIB results in an impaired distribution to lipid rafts and a reduced inhibitory function. Studies addressing the functions of disease-associated polymorphisms in the FcepsilonRI beta-subunit and low-affinity FcgammaRs are also considered.
Collapse
Affiliation(s)
- Zen-Ichiro Honda
- Department of Allergy and Rheumatology, Faculty of Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan,
| |
Collapse
|
27
|
Tsuchiya N, Honda ZI, Tokunaga K. Role of B cell inhibitory receptor polymorphisms in systemic lupus erythematosus: a negative times a negative makes a positive. J Hum Genet 2006; 51:741-750. [PMID: 16946996 DOI: 10.1007/s10038-006-0030-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 06/09/2006] [Indexed: 10/24/2022]
Abstract
B lymphocytes play a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). Here, we will review our studies on the role of polymorphisms of two genes coding for B cell inhibitory receptors, FCGR2B and CD72. In FCGR2B, a single nucleotide polymorphism leading to a nonsynonymous substitution, Ile232Thr, within the transmembrane domain was identified, and a significant association of the 232Thr/Thr genotype with SLE was observed in Japanese, Thai and Chinese populations, while this allele was found to be rare in Caucasians. On the other hand, the association of FCGR2B promoter polymorphism with SLE in Caucasians has been reported by two independent groups, but this allele was not found to be present in Japanese. These observations demonstrate that the association of FCGR2B polymorphisms with SLE is common to multiple populations, but the alleles associated with SLE depend upon the genetic background of each population. Functional analyses using a human B cell line lacking endogenous FcgammaRIIb revealed that SLE-associated 232Thr allele product was partially excluded from membrane lipid rafts under resting conditions and after coligation with B cell receptor, and was significantly less potent at inhibiting B cell activation. Two haplotypes were identified in CD72, one of which was associated with increased production of an alternative splicing isoform that substantially alters the extracellular region of CD72. Interestingly, the presence of the haplotype significantly decreased the risk of SLE conferred by FCGR2B-232Thr in an epistatic manner. These observations emphasize the need to understand human immune system diversity if we are to improve our understanding of the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Naoyuki Tsuchiya
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Zen-Ichiro Honda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
28
|
Yeung T, Ozdamar B, Paroutis P, Grinstein S. Lipid metabolism and dynamics during phagocytosis. Curr Opin Cell Biol 2006; 18:429-37. [PMID: 16781133 DOI: 10.1016/j.ceb.2006.06.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2006] [Accepted: 06/06/2006] [Indexed: 12/24/2022]
Abstract
Phagocytosis, the engulfment of particles, mediates the elimination of invading pathogens as well as the clearance of apoptotic cells. Ingested particles reside within a vacuole or phagosome, where they are eventually destroyed and digested. The phagosomal lumen acquires microbicidal and digestive properties through interaction with various components of the endocytic pathway, a process known as maturation. Lipids are known to have numerous roles in phagosome formation and maturation; recent developments in the design of lipid-specific probes and in high-resolution imaging have revealed that lipids, notably phosphoinositides, are involved in signaling, actin assembly and the recruitment of molecular motors to sites of ingestion. In addition, phosphoinositides and other lipids also regulate multiple membrane budding, fission and fusion events required for maturation.
Collapse
Affiliation(s)
- Tony Yeung
- Division of Cell Biology, The Hospital for Sick Children, Institute of Medical Sciences, University of Toronto, Toronto, M5S 1A8, Canada
| | | | | | | |
Collapse
|
29
|
Hillyard DZ, Nutt CD, Thomson J, McDonald KJ, Wan RK, Cameron AJM, Mark PB, Jardine AG. Statins inhibit NK cell cytotoxicity by membrane raft depletion rather than inhibition of isoprenylation. Atherosclerosis 2006; 191:319-25. [PMID: 16814295 DOI: 10.1016/j.atherosclerosis.2006.05.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 05/19/2006] [Accepted: 05/22/2006] [Indexed: 11/19/2022]
Abstract
To investigate the potential determinants of the pleiotropic effects of statins, we measured NK cell cytotoxicity in samples from normal subjects and patients, including patients receiving statin therapy. In a multivariate analysis, NK cell cytotoxicity was related to total plasma cholesterol concentration rather than statin use. In vitro, we investigated the role of lipid modification, specifically the effects on membrane rafts and raft-dependent signal transduction. We demonstrate that statins reduce NK cell cytotoxicity and that membrane cholesterol depletion by cyclodextrins has a similar effect. In contrast, isoprenyl transferase inhibitors had little or no effect on NK cell function. We hypothesise that the pleiotropic effects of statins reflect changes in membrane cholesterol and, specifically, the density of membrane rafts. Moreover, there is likely to be a relationship between membrane cholesterol, membrane rafts and cell function that may be involved in the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Dianne Z Hillyard
- Renal Research Group, BHF Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Cell activation results from the transient displacement of an active balance between positive and negative signaling. This displacement depends in part on the engagement of cell surface receptors by extracellular ligands. Among these are receptors for the Fc portion of immunoglobulins (FcRs). FcRs are widely expressed by cells of hematopoietic origin. When binding antibodies, FcRs provide these cells with immunoreceptors capable of triggering numerous biological responses in response to a specific antigen. FcR-dependent cell activation is regulated by negative signals which are generated together with positive signals within signalosomes that form upon FcR engagement. Many molecules involved in positive signaling, including the FcRbeta subunit, the src kinase lyn, the cytosolic adapter Grb2, and the transmembrane adapters LAT and NTAL, are indeed also involved in negative signaling. A major player in negative regulation of FcR signaling is the inositol 5-phosphatase SHIP1. Several layers of negative regulation operate sequentially as FcRs are engaged by extracellular ligands with an increasing valency. A background protein tyrosine phosphatase-dependent negative regulation maintains cells in a "resting" state. SHIP1-dependent negative regulation can be detected as soon as high-affinity FcRs are occupied by antibodies in the absence of antigen. It increases when activating FcRs are engaged by multivalent ligands and, further, when FcR aggregation increases, accounting for the bell-shaped dose-response curve observed in excess of ligand. Finally, F-actin skeleton-associated high-molecular weight SHIP1, recruited to phosphorylated ITIMs, concentrates in signaling complexes when activating FcRs are coengaged with inhibitory FcRs by immune complexes. Based on these data, activating and inhibitory FcRs could be used for new therapeutic approaches to immune disorders.
Collapse
Affiliation(s)
- Marc Daëron
- Unité d'Allergologie Moléculaire et Cellulaire, Département d'Immunologie, Institut Pasteur, Paris, France
| | | |
Collapse
|
31
|
Maurice P, Waeckel L, Pires V, Sonnet P, Lemesle M, Arbeille B, Vassy J, Rochette J, Legrand C, Fauvel-Lafève F. The platelet receptor for type III collagen (TIIICBP) is present in platelet membrane lipid microdomains (rafts). Histochem Cell Biol 2005; 125:407-17. [PMID: 16205938 DOI: 10.1007/s00418-005-0076-y] [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] [Accepted: 09/08/2005] [Indexed: 02/02/2023]
Abstract
Platelet interactions with collagen are orchestrated by the presence or the migration of platelet receptor(s) for collagen into lipid rafts, which are specialized lipid microdomains from the platelet plasma membrane enriched in signalling proteins. Electron microscopy shows that in resting platelets, TIIICBP, a receptor specific for type III collagen, is present on the platelet membrane and associated with the open canalicular system, and redistributes to the platelet membrane upon platelet activation. After platelet lysis by 1% Triton X-100 and the separation of lipid rafts on a discontinuous sucrose gradient, TIIICBP is recovered in lipid raft-containing fractions and Triton X-100 insoluble fractions enriched in cytoskeleton proteins. Platelet aggregation, induced by type III collagen, was inhibited after disruption of the lipid rafts by cholesterol depletion, whereas platelet adhesion under static conditions did not require lipid raft integrity. These results indicate that TIIICBP, a platelet receptor involved in platelet interaction with type III collagen, is localized within platelet lipid rafts where it could interact with other platelet receptors for collagen (GP VI and alpha2beta1 integrin) for efficient platelet activation.
Collapse
Affiliation(s)
- Pascal Maurice
- INSERM, U 553, IFR 105, Institut d'Hématologie, Université Paris VII Denis Diderot, 75475, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kono H, Kyogoku C, Suzuki T, Tsuchiya N, Honda H, Yamamoto K, Tokunaga K, Honda ZI. FcγRIIB Ile232Thr transmembrane polymorphism associated with human systemic lupus erythematosus decreases affinity to lipid rafts and attenuates inhibitory effects on B cell receptor signaling. Hum Mol Genet 2005; 14:2881-92. [PMID: 16115811 DOI: 10.1093/hmg/ddi320] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The B cell inhibitory receptor FcgammaRIIB plays crucial roles in the maintenance of self-tolerance. We have identified a polymorphism FCGR2B c.695T>C that results in the non-conservative replacement of 232Ile at the transmembrane helix to Thr and demonstrated the association of the polymorphism with susceptibility to systemic lupus erythematosus (SLE) in Asians. In this study, we examined the impact of FCGR2B c.695T>C on the functional properties of FcgammaRIIB by expressing each allele product in a human B cell line ST486 lacking endogenous FcgammaRIIB. FcgammaRIIB 232Thr was found to be significantly less potent than wild-type 232Ile in inhibiting B cell receptor (BCR)-mediated phosphatidylinositol-3,4,5-trisphosphate accumulation, Akt and PLCgamma2 activation and calcium mobilization, and to display decreased levels of tyrosine phosphorylation and SH2-containing 5'-inositolphosphate phosphatase recruitment compared with 232Ile after IgG Fc-mediated coligation with BCR. Notably, a quantitative analysis of the subcellular distribution of FcgammaRIIB using 125I-labeled anti-FcgammaRIIB revealed that FcgammaRIIB 232Thr is less effectively distributed to detergent-insoluble lipid rafts than 232Ile, findings in accordance with the importance of the transmembrane amino acid residues, in particular large hydrophobic amino acids including Ile, in the association of membrane proteins with lipid rafts. Given the crucial roles of lipid rafts in integrating BCR signaling, decreased association of FcgammaRIIB 232Thr could contribute to its impaired inhibitory potential. Collectively, the present findings indicate that the Ile232Thr substitution affects the localization and function of FcgammaRIIB and that the molecular mechanism may link the polymorphism and susceptibility to SLE.
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acid Substitution
- Antigens, CD/analysis
- Antigens, CD/genetics
- B-Lymphocytes/immunology
- Calcium Signaling
- Cells, Cultured
- Genetic Predisposition to Disease
- Humans
- Isoleucine/genetics
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Membrane Microdomains/immunology
- Molecular Sequence Data
- Phosphatidylinositol Phosphates/metabolism
- Polymorphism, Genetic
- Receptors, Antigen, B-Cell/antagonists & inhibitors
- Receptors, Antigen, B-Cell/metabolism
- Receptors, IgG/analysis
- Receptors, IgG/genetics
- Signal Transduction
- Threonine/genetics
Collapse
Affiliation(s)
- Hajime Kono
- Department of Allergy and Rheumatology, Faculty of Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Dai Q, Zhang J, Pruett SB. Ethanol alters cellular activation and CD14 partitioning in lipid rafts. Biochem Biophys Res Commun 2005; 332:37-42. [PMID: 15896296 DOI: 10.1016/j.bbrc.2005.04.088] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 04/18/2005] [Indexed: 12/11/2022]
Abstract
Alcohol consumption interferes with innate immunity. In vivo EtOH administration suppresses cytokine responses induced through Toll-like receptor 4 (TLR4) and inhibits TLR4 signaling. Actually, EtOH exhibits a generalized suppressive effect on signaling and cytokine responses induced by through most TLRs. However, the underlying mechanism remains unknown. RAW264.7 cells were treated with LPS or co-treated with EtOH or with lipid raft-disrupting drugs. TNF-alpha production, IRAK-1 activation, and CD14 partition were evaluated. EtOH or nystatin, a lipid raft-disrupting drug, suppressed LPS-induced production of TNF-alpha. The suppressive effect of EtOH on LPS-induced TNF-alpha production was additive with that of methyl-beta-cyclodextrin (MCD), another lipid raft-disrupting drug. EtOH interfered with IRAK-1 activation, an early TLR4 intracellular signaling event. Cell fractionation analyses show that acute EtOH altered LPS-related partition of CD14, a critical component of the LPS receptor complex. These results suggest a novel mechanism of EtOH action that involves interference with lipid raft clustering induced by LPS. This membrane action of EtOH might be one of the mechanisms by which EtOH acts as a generalized suppressor for TLR signaling.
Collapse
Affiliation(s)
- Qun Dai
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | |
Collapse
|
34
|
Riemann D, Tcherkes A, Hansen GH, Wulfaenger J, Blosz T, Danielsen EM. Functional co-localization of monocytic aminopeptidase N/CD13 with the Fcγ receptors CD32 and CD64. Biochem Biophys Res Commun 2005; 331:1408-12. [PMID: 15883031 DOI: 10.1016/j.bbrc.2005.04.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Indexed: 02/02/2023]
Abstract
Information about the function of aminopeptidase N/CD13 on monocytes is limited. In order to gain more insight into its interaction with other proteins, we have identified molecules that co-localize with the membrane ectoenzyme at the cell surface of monocytes. Using laser scanning and electron microscopy as well as fluorescence resonance energy transfer (FRET) measured by flow cytometry we show that monocytic CD13 co-localized with the Fc gamma receptor II/CD32 after Fc receptor ligation by a CD32-specific antibody. FRET was also observed between CD13 and the Fc gamma receptor I/CD64, but not with the myeloid marker CD33 representing a member of the sialoadhesin family. Our results imply a novel functional role of CD13 and Fc gamma receptors as members of a multimeric receptor complex. Further studies have to be done to elucidate common signaling pathways of these molecules.
Collapse
Affiliation(s)
- Dagmar Riemann
- Institute of Medical Immunology, Martin Luther University Halle/Wittenberg, Magdeburger Strasse 2, D-06097 Halle, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Gregor Rothe
- Bremer Zentrum für Laboratoriumsmedizin GmbH, D-28205 Bremen, Germany
| | | |
Collapse
|
36
|
Lesourne R, Fridman WH, Daëron M. Dynamic interactions of Fc gamma receptor IIB with filamin-bound SHIP1 amplify filamentous actin-dependent negative regulation of Fc epsilon receptor I signaling. THE JOURNAL OF IMMUNOLOGY 2005; 174:1365-73. [PMID: 15661894 DOI: 10.4049/jimmunol.174.3.1365] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The engagement of high affinity receptors for IgE (FcepsilonRI) generates both positive and negative signals whose integration determines the intensity of mast cell responses. FcepsilonRI-positive signals are also negatively regulated by low affinity receptors for IgG (FcgammaRIIB). Although the constitutive negative regulation of FcepsilonRI signaling was shown to depend on the submembranous F-actin skeleton, the role of this compartment in FcgammaRIIB-dependent inhibition is unknown. We show in this study that the F-actin skeleton is essential for FcgammaRIIB-dependent negative regulation. It contains SHIP1, the phosphatase responsible for inhibition, which is constitutively associated with the actin-binding protein, filamin-1. After coaggregation, FcgammaRIIB and FcepsilonRI rapidly interact with the F-actin skeleton and engage SHIP1 and filamin-1. Later, filamin-1 and F-actin dissociate from FcR complexes, whereas SHIP1 remains associated with FcgammaRIIB. Based on these results, we propose a dynamic model in which the submembranous F-actin skeleton forms an inhibitory compartment where filamin-1 functions as a donor of SHIP1 for FcgammaRIIB, which concentrate this phosphatase in the vicinity of FcepsilonRI and thereby extinguish activation signals.
Collapse
MESH Headings
- Actins/antagonists & inhibitors
- Actins/metabolism
- Actins/physiology
- Animals
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Contractile Proteins/metabolism
- Down-Regulation/immunology
- Filamins
- Immunoglobulin E/physiology
- Inositol Polyphosphate 5-Phosphatases
- Mast Cells/drug effects
- Mast Cells/enzymology
- Mast Cells/metabolism
- Membrane Microdomains/metabolism
- Mice
- Microfilament Proteins/metabolism
- Molecular Weight
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases
- Phosphoric Monoester Hydrolases/metabolism
- Phosphoric Monoester Hydrolases/physiology
- Protein Binding/immunology
- Protein Isoforms/metabolism
- Rats
- Receptor Aggregation/immunology
- Receptors, IgE/antagonists & inhibitors
- Receptors, IgE/metabolism
- Receptors, IgE/physiology
- Receptors, IgG/antagonists & inhibitors
- Receptors, IgG/metabolism
- Receptors, IgG/physiology
- Resting Phase, Cell Cycle/immunology
- Signal Transduction/immunology
- Thiazoles/pharmacology
- Thiazolidines
- Time Factors
Collapse
Affiliation(s)
- Renaud Lesourne
- Laboratoire d'Immunologie Cellulaire et Clinique, Institut National de la Santé et de la Recherche Médicale, Unité 255, Institut Biomédical des Cordeliers, Paris, France
| | | | | |
Collapse
|
37
|
Mina‐Osorio P, Ortega E. Aminopeptidase N (CD13) functionally interacts with FcgammaRs in human monocytes. J Leukoc Biol 2005; 77:1008-17. [PMID: 15758076 PMCID: PMC7167080 DOI: 10.1189/jlb.1204714] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Aminopeptidase N (E.C. 3.4.11.2) is a membrane-bound metalloproteinase expressed in many tissues. Although its cytoplasmic portion has only eight amino acids, cross-linking of CD13 by monoclonal antibodies (mAb) has been shown to trigger intracellular signaling. A functional association between CD13 and receptors for immunoglobulin G (FcgammaRs) has been proposed. In this work, we evaluated possible functional interactions between CD13 and FcgammaRs in human peripheral blood monocytes and in U-937 promonocytic cells. Our results show that during FcgammaR-mediated phagocytosis, CD13 redistributes to the phagocytic cup and is internalized into the phagosomes. Moreover, modified erythrocytes that interact with the monocytic cell membrane through FcgammaRI and CD13 are ingested simultaneously, more efficiently than those that interact through the FcgammaRI only. Also, co-cross-linking of CD13 with FcgammaRI by specific mAbs increases the level and duration of Syk phosphorylation induced by FcgammaRI cross-linking. Finally, FcgammaRI and CD13 colocalize in zones of cellular polarization and coredistribute after aggregation of either of them. These results demonstrate that CD13 and FcgammaRI can functionally interact on the monocytic cell membrane and suggest that CD13 may act as a signal regulator of FcgammaR function.
Collapse
Affiliation(s)
- Paola Mina‐Osorio
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México
| | - Enrique Ortega
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México
| |
Collapse
|
38
|
Rollet-Labelle E, Marois S, Barbeau K, Malawista SE, Naccache PH. Recruitment of the cross-linked opsonic receptor CD32A (FcgammaRIIA) to high-density detergent-resistant membrane domains in human neutrophils. Biochem J 2004; 381:919-28. [PMID: 15130090 PMCID: PMC1133904 DOI: 10.1042/bj20031808] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 04/05/2004] [Accepted: 05/06/2004] [Indexed: 02/02/2023]
Abstract
We have previously shown that CD32A (or FcgammaRIIA), one of the main opsonin receptors, was rapidly insolubilized and degraded in intact neutrophils after its cross-linking. In view of these experimental difficulties, the early signalling steps in response to CD32A activation were studied in purified plasma membranes of neutrophils. After CD32A cross-linking in these fractions, the tyrosine phosphorylation of two major substrates, the receptor itself and the tyrosine kinase Syk, was observed. Phosphorylation of these two proteins was observed only in the presence of orthovanadate, indicating the presence, in the membranes, of one or more tyrosine phosphatases that maintain CD32A dephosphorylation. The tyrosine phosphorylation of these two proteins was inhibited by the Src kinase inhibitor, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). The ligation of CD32A led to its recruitment to a previously uncharacterized subset of high-density flotillin-1-positive DRMs (detergent-resistant membranes). The changes in the solubility properties of CD32A were observed in the absence of added ATP; therefore, they were probably not secondary to the tyrosine phosphorylation of the receptor, rather they preceded it. Src kinases as well as Syk were constitutively present in DRMs of high and low density and no evident changes in their distribution were detected after cross-linking of CD32A. Pretreatment of plasma membranes with methyl-beta-cyclodextrin did not inhibit the recruitment of CD32A to DRMs, although it led to the loss of the Src kinase Lyn from these fractions. In addition, methyl-beta-cyclodextrin inhibited the tyrosine phosphorylation of CD32A and Syk induced by cross-linking of CD32A. This membrane model allowed us to observe a movement of CD32A from detergent-soluble regions of the membranes to DRMs, where it joined Src kinases and Syk and became tyrosine-phosphorylated.
Collapse
Affiliation(s)
- Emmanuelle Rollet-Labelle
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUL and Department of Medicine, Faculty of Medicine, Laval University, Sainte-Foy, QC, Canada G1V 4G2.
| | | | | | | | | |
Collapse
|
39
|
Swanson JA, Hoppe AD. The coordination of signaling during Fc receptor-mediated phagocytosis. J Leukoc Biol 2004; 76:1093-103. [PMID: 15466916 DOI: 10.1189/jlb.0804439] [Citation(s) in RCA: 224] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Phagocytosis by macrophages can be initiated by Fcgamma receptors (FcR) in membranes that bind to Fc regions of immunoglobulin G (IgG). Activated FcR transduce signals to cytoplasm, which regulate the internalization of IgG-coated particles into plasma membrane-derived vacuoles, phagosomes. Particles internalized by phagocytosis are much larger than FcR, which prompts questions of if and how the receptors are coordinated with each other. FcR-mediated signal transduction entails recruitment of proteins from cytoplasm to the receptor, largely via protein phosphorylation. These FcR signaling complexes then activate proteins that regulate actin, myosin, membrane fusion, and the production of reactive oxygen intermediates. Recent fluorescence microscopic studies of phagocytosis in macrophages indicate that signaling by FcR occurs as a sequence of distinct stages, evident in the spatial and temporal patterns of phosphoinositides, protein kinase C, and Rho-family GTPase activation on forming phagosomes. The coordination of these stages may be regulated by lipids or lipid-anchored proteins, which diffuse away from FcR complexes. Lateral diffusion of FcR-derived signals could integrate FcR-dependent responses over large areas of membrane in the forming phagosome.
Collapse
Affiliation(s)
- Joel A Swanson
- University of Michigan Medical School, 1335 Catherine Street, Med Sci II, Rm. 5608, Ann Arbor, MI 48109-0620, USA.
| | | |
Collapse
|
40
|
Lucero HA, Robbins PW. Lipid rafts-protein association and the regulation of protein activity. Arch Biochem Biophys 2004; 426:208-24. [PMID: 15158671 DOI: 10.1016/j.abb.2004.03.020] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2004] [Revised: 03/22/2004] [Indexed: 11/18/2022]
Abstract
Lipid rafts are membrane microdomains enriched in saturated phospholipids, sphingolipids, and cholesterol. They have a varied but distinct protein composition and have been implicated in diverse cellular processes including polarized traffic, signal transduction, endo- and exo-cytoses, entrance of obligate intracellular pathogens, and generation of pathological forms of proteins associated with Alzheimer's and prion diseases. Raft proteins can be permanently or temporarily associated to lipid rafts. Here, we review recent advances on the biochemical and cell biological characterization of rafts, and on the emerging concept of the temporary residency of proteins in rafts as a regulatory mechanism of their biological activity.
Collapse
Affiliation(s)
- Héctor A Lucero
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University Medical Center, Boston, MA 02118, USA.
| | | |
Collapse
|
41
|
Hazenbos WLW, Clausen BE, Takeda J, Kinoshita T. GPI-anchor deficiency in myeloid cells causes impaired FcgammaR effector functions. Blood 2004; 104:2825-31. [PMID: 15238423 DOI: 10.1182/blood-2004-02-0671] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Signaling by transmembrane immunoglobulin G (IgG)-Fc receptors (FcgammaRs) in response to ligand involves association with membrane microdomains that contain glycosyl phosphatidylinositol (GPI)-anchored proteins. Recent in vitro studies showed enhancement of FcgammaR signaling by forced monoclonal antibody-mediated cocrosslinking with various GPI-anchored proteins. Here, the possibility that GPI-anchored proteins are involved in normal physiologic FcgammaR effector functions in response to a model ligand was studied using myeloid-specific GPI-anchor-deficient mice, generated by Cre-loxP conditional targeting. GPI-anchor-deficient primary myeloid cells exhibited normal FcgammaR expression and binding or endocytosis of IgG-immune complexes (IgG-ICs). Strikingly, after stimulation with IgG-ICs, tumor necrosis factor-alpha release, dendritic cell maturation, and antigen presentation were strongly reduced by GPI-anchor deficiency. Tyrosine phosphorylation of the FcR gamma-chain in response to IgG-IC was impaired in GPI-anchor-deficient cells. Myeloid GPI-anchor deficiency resulted in attenuated in vivo inflammatory processes during IgG-IC-mediated alveolitis. This study provides the first genetic evidence for an essential role of GPI-anchored proteins in physiologic FcgammaR effector functions in vitro and in vivo.
Collapse
Affiliation(s)
- Wouter L W Hazenbos
- Department of Immunoregulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
42
|
Horejsí V. Transmembrane adaptor proteins in membrane microdomains: important regulators of immunoreceptor signaling. Immunol Lett 2004; 92:43-9. [PMID: 15081526 DOI: 10.1016/j.imlet.2003.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2003] [Accepted: 10/10/2003] [Indexed: 11/26/2022]
Abstract
Membrane microdomains enriched in glycosphingolipids, cholesterol, glycosylphosphatidylinositol-anchored proteins and Src-family kinases (lipid rafts, GEMs) appear to play many important roles, especially in immunoreceptor signaling. Most transmembrane proteins are excluded from these specialized areas of membranes, notable exceptions being several palmitoylated proteins such as the T cell coreceptors CD4 and CD8, and several recently described transmembrane adaptor proteins, LAT, non-T cell activation linker (NTAL)/linker for activation of B cells (LAB), phosphoprotein associated with GEMs (PAG)/Csk-binding protein (Cbp) and LIME. All these molecules possess a very short N-terminal extracellular peptide (4-17 amino acids), transmembrane segment followed by a palmitoylation motif (CxxC) and cytoplasmic domain containing up to 10 tyrosine residues potentially phosphorylated by the Src- or Syk-family kinases. Tyrosine-phosphorylated transmembrane adaptors bind (directly via SH2 domains or indirectly) other signaling molecules such as several cytoplasmic adaptors and enzymes. LAT is indispensable for TCR signaling (and participates also at signal transduction initiated by some other receptors), NTAL/LAB appears to play a LAT-like role in signaling initiated by BCR and some Fc-receptors; PAG/Cbp cooperates with Csk, the cytoplasmic tyrosine kinase negatively regulating Src-family kinases. Additional transmembrane adaptors exist (TRIM, SIT, LAX) that are however not palmitoylated and therefore excluded from the lipid rafts; structurally and functionally, the zeta-chain family proteins tightly associated with immunoreceptors and activating NK-receptors may be also considered as transmembrane adaptors.
Collapse
Affiliation(s)
- Václav Horejsí
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídenská 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
43
|
van Sorge NM, van der Pol WL, van de Winkel JGJ. FcgammaR polymorphisms: Implications for function, disease susceptibility and immunotherapy. TISSUE ANTIGENS 2003; 61:189-202. [PMID: 12694568 DOI: 10.1034/j.1399-0039.2003.00037.x] [Citation(s) in RCA: 209] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Leukocyte Fcgamma receptors (FcgammaR) confer potent cellular effector functions to the specificity of IgG. FcgammaR-induced leukocyte functions, including antibody-dependent cellular cytotoxicity, phagocytosis, superoxide generation, degranulation, cytokine production and regulation of antibody production, are essential for host defense and immune regulation. The efficacy of IgG-induced FcgammaR function displays inter-individual heterogeneity due to genetic polymorphisms of three FcgammaR subclasses, FcgammaRIIa (CD32a), FcgammaRIIIa (CD16a), and FcgammaRIIIb (CD16b). FcgammaR polymorphisms have been associated with infectious and autoimmune disease, or with disease severity. FcgammaR polymorphisms may furthermore serve as markers for therapeutic efficacy and side-effects of treatment with monoclonal antibodies. In this review, FcgammaR function and the relevance of FcgammaR polymorphisms as prognostic markers for inflammatory disease and antibody-based immunotherapy are discussed.
Collapse
Affiliation(s)
- N M van Sorge
- Immunology/Neurology, UMC, Utrecht, the Netherlands.
| | | | | |
Collapse
|
44
|
Abstract
Detergent-resistant membrane microdomains enriched in sphingolipids, cholesterol and glycosylphosphatidylinositol-anchored proteins play essential roles in T cell receptor (TCR) signaling. These 'membrane rafts' accumulate several cytoplasmic lipid-modified molecules, including Src-family kinases, coreceptors CD4 and CD8 and transmembrane adapters LAT and PAG/Cbp, essential for either initiation or amplification of the signaling process, while most other abundant transmembrane proteins are excluded from these structures. TCRs in various T cell subpopulations may differ in their use of membrane rafts. Membrane rafts also seem to be involved in many other aspects of T cell biology, such as functioning of cytokine and chemokine receptors, adhesion molecules, antigen presentation, establishing cell polarity or interaction with important pathogens. Although the concept of membrane rafts explains several diverse biological phenomena, many basic issues, such as composition, size and heterogeneity, under native conditions, as well as the dynamics of their interactions with TCRs and other immunoreceptors, remain unclear, partially because of technical problems.
Collapse
Affiliation(s)
- Václav Horejsí
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Praha, Czech Republic.
| |
Collapse
|