51
|
Gajbhiye V, Palanirajan VK, Tekade RK, Jain NK. Dendrimers as therapeutic agents: a systematic review. J Pharm Pharmacol 2010. [PMID: 19703342 DOI: 10.1211/jpp.61.08.0002] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
Objectives
Dendrimers by virtue of their therapeutic value have recently generated enormous interest among biomedical scientists. This review describes the therapeutic prospects of the dendrimer system.
Key findings
Their bioactivity suggests them to be promising therapeutic agents, especially in wound healing, bone mineralisation, cartilage formation and tissue repair, and in topical treatments to prevent HIV transmission. Findings also demonstrate their potential as anti-prion, anti-Alzheimer's, anticoagulant, antidote, anti-inflammatory and anticancer agents. One of the dendrimer-based formulations with activity against herpes simplex virus (VivaGel from Starpharma) has successfully completed phase I clinical trials and is expected to be available on the market soon.
Summary
All reports cited in this review demonstrate the use of dendrimers as medical therapeutics in different ailments. The review focuses on the current state of therapeutic potential of the dendrimer system.
Collapse
Affiliation(s)
- Virendra Gajbhiye
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr Hari Singh Gour University, Sagar, India
| | - Vijayaraj K Palanirajan
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr Hari Singh Gour University, Sagar, India
| | - Rakesh K Tekade
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr Hari Singh Gour University, Sagar, India
| | - Narendra K Jain
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr Hari Singh Gour University, Sagar, India
| |
Collapse
|
52
|
Abstract
Dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN). DC-SIGN is a C-type lectin receptor that recognizes N-linked high-mannose oligosaccharides and branched fucosylated structures. It is now clear that the biological role of DC-SIGN is two-fold. It is primarily expressed by dendritic cells and mediates important functions necessary for the induction of successful immune responses that are essential for the clearance of microbial infections, such as the capture, destruction, and presentation of microbial pathogens to induce successful immune responses. Yet, on the other hand, pathogens may also exploit DC-SIGN to modulate DC functioning thereby skewing the immune response and promoting their own survival. This chapter presents an overview of the structure of DC-SIGN and its expression pattern among immune cells. The current state of knowledge of DC-SIGN-carbohydrate interactions is discussed and how these interactions influence dendritic cell functioning is examined. The molecular aspects that underlie the selectivity of DC-SIGN for mannose-and fucose-containing carbohydrates are detailed. Furthermore, the chapter discusses the role of DC-SIGN in dendritic cell biology and how certain bacterial pathogens exploit DC-SIGN to escape immune surveillance.
Collapse
|
53
|
Ehlers S. DC-SIGN and mannosylated surface structures of Mycobacterium tuberculosis: a deceptive liaison. Eur J Cell Biol 2009; 89:95-101. [PMID: 19892432 DOI: 10.1016/j.ejcb.2009.10.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is recognized by pattern recognition receptors on macrophages and dendritic cells, thereby triggering phagocytosis, antigen presentation to T cells and cytokine secretion. The dendritic cell-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN) is a calcium-dependent carbohydrate-binding protein with specificity for mannose-containing glycoconjugates and fucose-containing Lewis antigens. Mannosylated moieties of the mycobacterial cell wall, such as mannose-capped lipoarabinomannan (manLAM) or higher-order phosphatidylinositol-mannosides (PIMs) of Mtb, were previously shown to bind to DC-SIGN on immature dendritic cells and macrophage subpopulations. This interaction reportedly impaired dendritic cell maturation, modulated cytokine secretion by phagocytes and dendritic cells and was postulated to cause suppression of protective immunity to TB. However, experimental Mtb infections in mice transgenic for human DC-SIGN revealed that, instead of favoring immune evasion of mycobacteria, DC-SIGN may promote host protection by limiting tissue pathology. Furthermore, infection studies with mycobacterial strains genetically engineered to lack manLAM or PIMs demonstrated that the manLAM/PIM-DC-SIGN interaction was not critical for cytokine secretion in vitro and protective immunity in vivo. The dominant Mtb-derived ligands for DC-SIGN are presently unknown, and a major role of DC-SIGN in the immune response to Mtb infection may lie in its capacity to maintain a balanced inflammatory state during chronic TB.
Collapse
Affiliation(s)
- Stefan Ehlers
- Microbial Inflammation Research, Research Center Borstel, Parkallee 1, 23845 Borstel, Germany.
| |
Collapse
|
54
|
Ceballos A, Remes Lenicov F, Sabatté J, Rodríguez Rodrígues C, Cabrini M, Jancic C, Raiden S, Donaldson M, Agustín Pasqualini R, Marin-Briggiler C, Vazquez-Levin M, Capani F, Amigorena S, Geffner J. Spermatozoa capture HIV-1 through heparan sulfate and efficiently transmit the virus to dendritic cells. ACTA ACUST UNITED AC 2009; 206:2717-33. [PMID: 19858326 PMCID: PMC2806607 DOI: 10.1084/jem.20091579] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Semen is the main vector for HIV-1 dissemination worldwide. It contains three major sources of infectious virus: free virions, infected leukocytes, and spermatozoa-associated virions. We focused on the interaction of HIV-1 with human spermatozoa and dendritic cells (DCs). We report that heparan sulfate is expressed in spermatozoa and plays an important role in the capture of HIV-1. Spermatozoa-attached virus is efficiently transmitted to DCs, macrophages, and T cells. Interaction of spermatozoa with DCs not only leads to the transmission of HIV-1 and the internalization of the spermatozoa but also results in the phenotypic maturation of DCs and the production of IL-10 but not IL-12p70. At low values of extracellular pH (∼6.5 pH units), similar to those found in the vaginal mucosa after sexual intercourse, the binding of HIV-1 to the spermatozoa and the consequent transmission of HIV-1 to DCs were strongly enhanced. Our observations support the notion that far from being a passive carrier, spermatozoa acting in concert with DCs might affect the early course of sexual transmission of HIV-1 infection.
Collapse
Affiliation(s)
- Ana Ceballos
- Centro Nacional de Referencia para SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Stax MJ, van Montfort T, Sprenger RR, Melchers M, Sanders RW, van Leeuwen E, Repping S, Pollakis G, Speijer D, Paxton WA. Mucin 6 in seminal plasma binds DC-SIGN and potently blocks dendritic cell mediated transfer of HIV-1 to CD4(+) T-lymphocytes. Virology 2009; 391:203-11. [PMID: 19682628 DOI: 10.1016/j.virol.2009.06.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 04/03/2009] [Accepted: 06/06/2009] [Indexed: 10/20/2022]
Abstract
Many viruses transmitted via the genital or oral mucosa have the potential to interact with dendritic cell-specific intercellular adhesion molecule-3 grabbing non integrin (DC-SIGN) expressed on immature dendritic cells (iDCs) that lie below the mucosal surface. These cells have been postulated to capture and disseminate human immunodeficiency virus type-1 (HIV-1) to CD4(+) lymphocytes, potentially through breaches in the mucosal lining. We have previously described that BSSL (bile salt-stimulated lipase) in human milk can bind DC-SIGN and block transfer. Here we demonstrate that seminal plasma has similar DC-SIGN blocking properties as BSSL in human milk. Using comparative SDS-PAGE and Western blotting combined with mass spectrometry we identified mucin 6 as the DC-SIGN binding component in seminal plasma. Additionally, we demonstrate that purified mucin 6 binds DC-SIGN and successfully inhibits viral transfer. Mucin 6 in seminal plasma may therefore interfere with the sexual transmission of HIV-1 and other DC-SIGN co-opting viruses.
Collapse
Affiliation(s)
- Martijn J Stax
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam, Academic Medical Center of the University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
DC-SIGN mediates cell-free infection and transmission of human T-cell lymphotropic virus type 1 by dendritic cells. J Virol 2009; 83:10908-21. [PMID: 19692463 DOI: 10.1128/jvi.01054-09] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite the susceptibility of dendritic cells (DCs) to human T-cell lymphotropic virus type 1 (HTLV-1) infection and the defined role of these cells in disease pathogenesis, the mechanisms of viral binding to DCs have not been fully delineated. Recently, a glucose transporter, GLUT-1, heparan sulfate proteoglycans (HSPGs), and neuropilin-1 (NRP-1) were demonstrated to facilitate HTLV-1 entry into T cells. DCs express their own array of antigen receptors, the most important being the DC-specific intercellular adhesion molecule-3 (ICAM-3)-grabbing nonintegrin (DC-SIGN) with respect to retrovirus binding. Consequently, the role of DC-SIGN and other HTLV-1 attachment factors was analyzed in viral binding, transmission, and productive infection using monocyte-derived DCs (MDDCs), blood myeloid DCs, and B-cell lines expressing DC-SIGN. The relative expression of DC-SIGN, GLUT-1, HSPGs, and NRP-1 first was examined on both DCs and B-cell lines. Although the inhibition of these molecules reduced viral binding, HTLV-1 transmission from DCs to T cells was mediated primarily by DC-SIGN. DC-SIGN also was shown to play a role in the infection of MDDCs as well as model B-cell lines. The HTLV-1 infection of MDDCs also was achieved in blood myeloid DCs following the enhancement of virus-induced interleukin-4 production and subsequent DC-SIGN expression in this cell population. This study represents the first comprehensive analysis of potential HTLV-1 receptors on DCs and strongly suggests that DC-SIGN plays a critical role in HTLV-1 binding, transmission, and infection, thereby providing an attractive target for the development of antiretroviral therapeutics and microbicides.
Collapse
|
57
|
Role of phosphatidylinositol mannosides in the interaction between mycobacteria and DC-SIGN. Infect Immun 2009; 77:4538-47. [PMID: 19651855 DOI: 10.1128/iai.01256-08] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The C-type lectin dendritic cell (DC)-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) is the major receptor on DCs for mycobacteria of the Mycobacterium tuberculosis complex. Recently, we have shown that although the mannose caps of the mycobacterial surface glycolipid lipoarabinomannan (ManLAM) are essential for the binding to DC-SIGN, genetic removal of these caps did not diminish the interaction of whole mycobacteria with DC-SIGN and DCs. Here we investigated the role of the structurally related glycolipids phosphatidylinositol mannosides (PIMs) as possible ligands for DC-SIGN. In a binding assay with both synthetic and natural PIMs, DC-SIGN exhibited a high affinity for hexamannosylated PIM(6), which contains terminal alpha(1-->2)-linked mannosyl residues identical to the mannose cap on ManLAM, but not for di- and tetramannosylated PIM(2) and PIM(4), respectively. To determine the role of PIM(6) in the binding of whole mycobacteria to DC-SIGN, a mutant strain of M. bovis bacillus Calmette-Guérin deficient in the production of PIM(6) (Delta pimE) was created, as well as a double knockout deficient in the production of both PIM(6) and the mannose caps on LAM (Delta pimE Delta capA). Compared to the wild-type strain, both mutant strains bound similarly well to DC-SIGN and DCs. Furthermore, the wild-type and mutant strains induced comparable levels of interleukin-10 and interleukin-12p40 when used to stimulate DCs. Hence, we conclude that, like ManLAM, PIM(6) represents a bona fide DC-SIGN ligand but that other, as-yet-unknown, ligands dominate in the interaction between mycobacteria and DCs.
Collapse
|
58
|
Abstract
Although most viral infections cause minor, if any, symptoms, a certain number result in serious illness. Viral disease symptoms result both from direct viral replication within host cells and from indirect immunopathological consequences. Dendritic cells (DCs) are key determinants of viral disease outcome; they activate immune responses during viral infection and direct T cells toward distinct T helper type responses. Certain viruses are able to skew cytokine secretion by DCs inducing and/or downregulating the immune system with the aim of facilitating and prolonging release of progeny. Thus, the interaction of DCs with viruses most often results in the absence of disease or complete recovery when natural functions of DCs prevail, but may lead to chronic illness or death when these functions are outmanoeuvred by viruses in the exploitation of DCs.
Collapse
Affiliation(s)
- Giulia Freer
- Retrovirus Center and Virology Section, Department of Experimental Pathology, University of Pisa, Pisa, Italy.
| | | |
Collapse
|
59
|
Ha CT, Waterhouse R, Warren J, Zimmermann W, Dveksler GS. N-glycosylation is required for binding of murine pregnancy-specific glycoproteins 17 and 19 to the receptor CD9. Am J Reprod Immunol 2009; 59:251-8. [PMID: 18275518 DOI: 10.1111/j.1600-0897.2007.00573.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PROBLEM Murine pregnancy-specific glycoproteins (PSGs) are encoded by 17 different genes. Different family members have different expression levels at different stages of embryonic development. It is currently unknown whether all members of this family of placentally secreted proteins have the same function and bind to the same receptor. Furthermore, the requirement of post-translational modifications for the activity of these highly glycosylated proteins remains undetermined. METHOD OF STUDY Recombinant PSG17 and PSG19 were generated and purified by affinity chromatography. An expression library was screened to identify the receptor for mouse PSG19. Binding to the receptor by proteins generated in different expression systems and mapping of the binding domain were analyzed by pull-down assays. Analysis of the carbohydrate composition of the receptor-binding domain was performed with the DIG glycan differentiation kit. RESULTS PSG19 binds to the tetraspanin CD9, specifically to extra cellular loop 2 and can induce secretion of TGFbeta1 by a macrophage cell line. The receptor-binding domain of PSG17 and PSG19 is post-translationally modified by the addition of N-linked carbohydrates and, when expressed in CHO cells, terminal sialic acids are detected. PSGs produced in bacteria do not bind CD9. CONCLUSION PSG19, as previously determined for PSG17, binds to the second extracellular loop 2 of the tetraspanin CD9. The first immunoglobulin variable-like domain of PSG19 is sufficient for receptor binding and function. Analysis of receptor usage by the remaining 15 murine PSGs will most likely require that the proteins be generated in eukaryotic expression systems, as we have demonstrated that the addition of carbohydrates is essential for PSG-receptor interaction.
Collapse
Affiliation(s)
- Cam T Ha
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | | | |
Collapse
|
60
|
Charoensap J, Engering A, Utaisincharoen P, van Kooyk Y, Sirisinha S. Activation of human monocyte-derived dendritic cells by Burkholderia pseudomallei does not require binding to the C-type lectin DC-SIGN. Trans R Soc Trop Med Hyg 2008; 102 Suppl 1:S76-81. [DOI: 10.1016/s0035-9203(08)70020-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
61
|
Sun P, Fernandez S, Marovich MA, Palmer DR, Celluzzi CM, Boonnak K, Liang Z, Subramanian H, Porter KR, Sun W, Burgess TH. Functional characterization of ex vivo blood myeloid and plasmacytoid dendritic cells after infection with dengue virus. Virology 2008; 383:207-15. [PMID: 19013627 DOI: 10.1016/j.virol.2008.10.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 09/17/2008] [Accepted: 10/10/2008] [Indexed: 10/21/2022]
Abstract
Myeloid and plasmacytoid dendritic cells (mDC and pDC) are naturally distinctive subsets. We exposed both subsets to dengue virus (DV) in vitro and investigated their functional characteristics. High levels of DV replication in mDC were found to correlate with DC-SIGN expression. Production of inflammatory cytokines by mDC increased gradually after DV-infection, which was dependent on DV replication. Co-stimulatory markers were upregulated on mDC upon DV-infection. On the contrary, lower levels of DV-replication were observed in pDC, but the cytokine production in pDC was quicker and stronger. This cytokine response was not dependent on viral replication, but dependent on cell endosomal activity and TLR7, and could be also induced by purified DV genome RNA. These results clearly suggested functional differences between mDC and pDC in response to DV infection. Additionally, the TLR7-mediated recognition of DV RNA may be involved in pDC functional activation.
Collapse
Affiliation(s)
- Peifang Sun
- Naval Medical Research Center, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Schneider-Schaulies S, Schneider-Schaulies J. Measles virus-induced immunosuppression. Curr Top Microbiol Immunol 2008; 330:243-69. [PMID: 19203113 DOI: 10.1007/978-3-540-70617-5_12] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immunosuppression is the major cause of infant death associated with acute measles and therefore of substantial clinical importance. Major hallmarks of this generalized modulation of immune functions are (1) lymphopenia, (2) a prolonged cytokine imbalance consistent with suppression of cellular immunity to secondary infections, and (3) silencing of peripheral blood lymphocytes, which cannot expand in response to ex vivo stimulation. Lymphopenia results from depletion, which can occur basically at any stage of lymphocyte development, and evidently, expression of the major MV receptor CD150 plays an important role in targeting these cells. Virus transfer to T cells is thought to be mediated by dendritic cells (DCs), which are considered central to the induction of T cell silencing and functional skewing. As a consequence of MV interaction, viability and functional differentiation of DCs and thereby their expression pattern of co-stimulatory molecules and soluble mediators are modulated. Moreover, MV proteins expressed by these cells actively silence T cells by interfering with signaling pathways essential for T cell activation.
Collapse
Affiliation(s)
- S Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany.
| | | |
Collapse
|
63
|
Op den Brouw ML, De Jong MAWP, Ludwig IS, Van Der Molen RG, Janssen HLA, Geijtenbeek TBH, Woltman AM. Branched oligosaccharide structures on HBV prevent interaction with both DC-SIGN and L-SIGN. J Viral Hepat 2008; 15:675-83. [PMID: 18482282 PMCID: PMC7166686 DOI: 10.1111/j.1365-2893.2008.00993.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatitis B virus (HBV) is a DNA virus that infects the liver as primary target. Currently, a high affinity receptor for HBV is still unknown. The dendritic cell specific C-type lectin DC-SIGN is involved in pathogen recognition through mannose and fucose containing carbohydrates leading to the induction of an anti-viral immune response. Many glycosylated viruses subvert this immune surveillance function and exploit DC-SIGN as a port of entry and for trans-infection of target cells. The glycosylation pattern on HBV surface antigens (HBsAg) together with the tissue distribution of HBV would allow interaction between HBV and DC-SIGN and its liver-expressed homologue L-SIGN. Therefore, a detailed study to investigate the binding of HBV to DC-SIGN and L-SIGN was performed. For HCV, both DC-SIGN and L-SIGN are known to bind envelope glycoproteins E1 and E2. Soluble DC-SIGN and L-SIGN specifically bound HCV virus-like particles, but no interaction with either HBsAg or HepG2.2.15-derived HBV was detected. Also, neither DC-SIGN nor L-SIGN transfected Raji cells bound HBsAg. In contrast, highly mannosylated HBV, obtained by treating HBV producing HepG2.2.15 cells with the alpha-mannosidase I inhibitor kifunensine, is recognized by DC-SIGN. The alpha-mannosidase I trimming of N-linked oligosaccharide structures thus prevents recognition by DC-SIGN. On the basis of these findings, it is tempting to speculate that HBV exploits mannose trimming as a way to escape recognition by DC-SIGN and thereby subvert a possible immune activation response.
Collapse
Affiliation(s)
- M. L. Op den Brouw
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - M. A. W. P. De Jong
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - I. S. Ludwig
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - R. G. Van Der Molen
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - H. L. A. Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - T. B. H. Geijtenbeek
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - A. M. Woltman
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
64
|
Harada T, Miura NN, Adachi Y, Nakajima M, Yadomae T, Ohno N. Highly Expressed Dectin-1 on Bone Marrow-Derived Dendritic Cells Regulates the Sensitivity to β-Glucan in DBA/2 Mice. J Interferon Cytokine Res 2008; 28:477-86. [DOI: 10.1089/jir.2007.0101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Toshie Harada
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo, Japan
| | - Noriko N. Miura
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo, Japan
| | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo, Japan
| | | | - Toshiro Yadomae
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo, Japan
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Horinouchi, Hachioji, Tokyo, Japan
| |
Collapse
|
65
|
Gaudart N, Ekpo P, Pattanapanyasat K, van Kooyk Y, Engering A. Leptospira interrogansis recognized through DC-SIGN and induces maturation and cytokine production by human dendritic cells. ACTA ACUST UNITED AC 2008; 53:359-67. [DOI: 10.1111/j.1574-695x.2008.00437.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
66
|
Harada T, Ohno N. Contribution of dectin-1 and granulocyte macrophage-colony stimulating factor (GM-CSF) to immunomodulating actions of beta-glucan. Int Immunopharmacol 2008; 8:556-66. [PMID: 18328447 DOI: 10.1016/j.intimp.2007.12.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 11/30/2007] [Accepted: 12/18/2007] [Indexed: 11/19/2022]
Abstract
beta-Glucans are major cell wall structural components in fungi. As they are not found in animals, these carbohydrates are considered to be classic pathogen-associated molecular patterns (PAMPs), and are recognized by the innate immune system. Although their immunomodulating activities have been shown to be associated with the recognition of some fungi, and with their medicinal properties in the field of cancer immunotherapy, it is still unclear how beta-glucans mediate their effects. Recent studies have started to shed some light on their cellular receptors, such as dectin-1, and their molecular mechanisms of action. We have extensively investigated the response of leukocytes to beta-glucan, focusing on cytokine induction by SCG, which is a major 6-branched 1,3-beta-d-glucan in Sparassis crispa Fr. There is a strain difference in the reactivity of mice to SCG, and DBA/1 and DBA/2 mice are highly sensitive strains. In the process of research on cytokine induction by SCG in DBA/2 mice, we found that GM-CSF plays a key biological role in this activity. Cytokine induction by SCG was completely abolished in dendritic cells from dectin-1 knockout mice. On the other hand, controlling the level of endogenous GM-CSF production and/or dectin-1 expression could regulate the reactivity to beta-glucan. These results indicate that the key factors in the responsiveness to beta-glucan are GM-CSF production and dectin-1 expression. In this review, we describe how the key molecules related to the expression of the immunomodulating activities of beta-glucan were identified, and how the response to beta-glucan is controlled.
Collapse
Affiliation(s)
- Toshie Harada
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | |
Collapse
|
67
|
Chapter 4 Receptor Interactions, Tropism, and Mechanisms Involved in Morbillivirus‐Induced Immunomodulation. Adv Virus Res 2008; 71:173-205. [DOI: 10.1016/s0065-3527(08)00004-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
68
|
Malcherek G, Mayr L, Roda-Navarro P, Rhodes D, Miller N, Trowsdale J. The B7 homolog butyrophilin BTN2A1 is a novel ligand for DC-SIGN. THE JOURNAL OF IMMUNOLOGY 2007; 179:3804-11. [PMID: 17785817 DOI: 10.4049/jimmunol.179.6.3804] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The MHC-encoded butyrophilin, BTN2A1, is a cell surface glycoprotein related to the extended family of B7 costimulatory molecules. BTN2A1 mRNA was expressed in most human tissues, but protein expression was significantly lower in leukocytes. An Ig-fusion protein of BTN2A1 bound to immature monocyte-derived dendritic cells. Binding diminished upon MoDC maturation and no binding was detected to Langerhans cells. Induction of the counterreceptor was IL-4 dependent and occurred early during dendritic cell differentiation. The interaction required the presence of Ca2+ and was mediated by high-mannose oligosaccharides. These properties matched DC-SIGN, a DC-specific HIV-1 entry receptor. This was confirmed by binding of soluble BTN2A1 to DC-SIGN-transfectants and its inhibition by a specific Ab. DC-SIGN bound to native BTN2A1 expressed on a range of tissues. However, BTN2A1 was not recognized on some normal cells such as HUVECs despite a similar expression level. The BTN2A1 of tumor cells such as HEK293T have more high-mannose moieties in comparison to HUVECs, and those high-mannose moieties are instrumental for binding to DC-SIGN. The data are consistent with tumor- or tissue-specific glycosylation of BTN2A1 governing recognition by DC-SIGN on immature monocyte-derived dendritic cells.
Collapse
Affiliation(s)
- Georg Malcherek
- Department of Pathology, Division of Immunology, University of Cambridge, Cambridge, United Kingdom.
| | | | | | | | | | | |
Collapse
|
69
|
Abstract
Dendritic cells (DCs) act not only as sentinels for detection of, but also as target cells for viruses, and this can be important for viral transport and spread. All subsets of DCs are equipped with a battery of receptors recognizing virus‐associated molecular signatures, and recognition of those launches a maturation programme that results in substantial alterations of morphology, motility and the DCs' interactive properties with the extracellular matrix and scanning T cells. In addition to being sensed, viruses are internalized into DCs and, for the major proportion, processed into peptides that are subsequently presented by major histocompatibility complex (MHC) molecules. Transmission of virus to T cells can occur after completion of their replication cycle if the intracellular milieu of the DC permits that. Alternatively, viruses can remain protected from degradation following entrapment by pattern recognition receptors in intracellular compartments, also referred to as virosomes, which translocate towards the DC/T cell interface. Most likely, transfer of virus to T cells occurs in these junctions, referred to as infectious synapses. In addition to promoting DC maturation, many viruses are able to downmodulate DC development and functions in order to evade immune recognition or to induce a generalized immunosuppression.
Collapse
Affiliation(s)
- Christine Pohl
- University of Wuerzburg, Institute for Virology and Immunobiology, Versbacher Str. 7, D-97078 Wuerzburg, Germany
| | | | | |
Collapse
|
70
|
Abstract
Toll-like receptors (TLRs) are essential mediators of both innate and adaptive immunity by recognizing and eliciting responses upon invasion of pathogens. The response of TLRs must be stringently regulated as exaggerated expression of signalling components as well as pro-inflammatory cytokines can have devastating effects on the host, resulting in chronic inflammatory diseases, autoimmune disorders and aid in the pathogenesis of TLR-associated human diseases. Therefore, it is essential that negative regulators act at multiple levels within TLR signalling cascades, as well as through eliciting negative-feedback mechanisms in order to synchronize the positive activation and negative regulation of signal transduction to avert potentially harmful immunological consequences. This review explores the various mechanisms employed by negative regulators to ensure the appropriate modulation of both immune and inflammatory responses.
Collapse
Affiliation(s)
- Tali Lang
- Centre for Functional Genomics and Human Disease, Monash Institute of Medical Research, Monash University, Victoria, Australia
| | | |
Collapse
|
71
|
Greene W, Kuhne K, Ye F, Chen J, Zhou F, Lei X, Gao SJ. Molecular biology of KSHV in relation to AIDS-associated oncogenesis. Cancer Treat Res 2007; 133:69-127. [PMID: 17672038 PMCID: PMC2798888 DOI: 10.1007/978-0-387-46816-7_3] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
KSHV has been established as the causative agent of KS, PEL, and MCD, malignancies occurring more frequently in AIDS patients. The aggressive nature of KSHV in the context of HIV infection suggests that interactions between the two viruses enhance pathogenesis. KSHV latent infection and lytic reactivation are characterized by distinct gene expression profiles, and both latency and lytic reactivation seem to be required for malignant progression. As a sophisticated oncogenic virus, KSHV has evolved to possess a formidable repertoire of potent mechanisms that enable it to target and manipulate host cell pathways, leading to increased cell proliferation, increased cell survival, dysregulated angiogenesis, evasion of immunity, and malignant progression in the immunocompromised host. Worldwide, approximately 40.3 million people are currently living with HIV infection. Of these, a significant number are coinfected with KSHV. The complex interplay between the two viruses dramatically elevates the risk for development of KSHV-induced malignancies, KS, PEL, and MCD. Although HAART significantly reduces HIV viral load, the entire T-cell repertoire and immune function may not be completely restored. In fact, clinically significant immune deficiency is not necessary for the induction of KSHV-related malignancy. Because of variables such as lack of access to therapy noncompliance with prescribed treatment, failure to respond to treatment and the development of drug-resistant strains of HIV, KSHV-induced malignancies will continue to present as major health concerns.
Collapse
Affiliation(s)
- Whitney Greene
- Tiumor Virology Program, Children's Cancer Research Institute, Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|
72
|
Naarding MA, Dirac AM, Ludwig IS, Speijer D, Lindquist S, Vestman EL, Stax MJ, Geijtenbeek TBH, Pollakis G, Hernell O, Paxton WA. Bile salt-stimulated lipase from human milk binds DC-SIGN and inhibits human immunodeficiency virus type 1 transfer to CD4+ T cells. Antimicrob Agents Chemother 2006; 50:3367-74. [PMID: 17005819 PMCID: PMC1610064 DOI: 10.1128/aac.00593-06] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A wide range of pathogens, including human immunodeficiency virus type 1 (HIV-1), hepatitis C virus, Ebola virus, cytomegalovirus, dengue virus, Mycobacterium, Leishmania, and Helicobacter pylori, can interact with dendritic cell (DC)-specific ICAM3-grabbing nonintegrin (DC-SIGN), expressed on DCs and a subset of B cells. More specifically, the interaction of the gp120 envelope protein of HIV-1 with DC-SIGN can facilitate the transfer of virus to CD4+ T lymphocytes in trans and enhance infection. We have previously demonstrated that a multimeric LeX component in human milk binds to DC-SIGN, preventing HIV-1 from interacting with this receptor. Biochemical analysis reveals that the compound is heat resistant, trypsin sensitive, and larger than 100 kDa, indicating a specific glycoprotein as the inhibitory compound. By testing human milk from three different mothers, we found the levels of DC-SIGN binding and viral inhibition to vary between samples. Using sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blotting, and matrix-assisted laser desorption ionization analysis, we identified bile salt-stimulated lipase (BSSL), a Lewis X (LeX)-containing glycoprotein found in human milk, to be the major variant protein between the samples. BSSL isolated from human milk bound to DC-SIGN and inhibited the transfer of HIV-1 to CD4+ T lymphocytes. Two BSSL isoforms isolated from the same human milk sample showed differences in DC-SIGN binding, illustrating that alterations in the BSSL forms explain the differences observed. These results indicate that variations in BSSL lead to alterations in LeX expression by the protein, which subsequently alters the DC-SIGN binding capacity and the inhibitory effect on HIV-1 transfer. Identifying the specific molecular interaction between the different forms may aid in the future design of antimicrobial agents.
Collapse
Affiliation(s)
- Marloes A Naarding
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Wethmar K, Helmus Y, Lühn K, Jones C, Laskowska A, Varga G, Grabbe S, Lyck R, Engelhardt B, Bixel MG, Butz S, Loser K, Beissert S, Ipe U, Vestweber D, Wild MK. Migration of immature mouse DC across resting endothelium is mediated by ICAM-2 but independent of beta2-integrins and murine DC-SIGN homologues. Eur J Immunol 2006; 36:2781-94. [PMID: 16981228 DOI: 10.1002/eji.200526311] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Immature dendritic cells (DC) reside in tissues where they initiate immune responses by taking up foreign antigens. Since DC have a limited tissue half-life, the DC pool in tissues has to be replenished constantly. This implies that precursor/immature DC must be able to cross non-activated endothelium using as yet unknown mechanisms. Here we show that immature, but not mature bone marrow-derived murine DC migrate across resting endothelial monolayers in vitro. We find that endothelial intercellular adhesion molecule-2 (ICAM-2) is a major player in transendothelial migration (TEM) of immature DC, accounting for at least 41% of TEM. Surprisingly, the ICAM-2-mediated TEM was independent of beta2-integrins, the known ICAM-2 ligands, since neither blocking of beta2-integrins with antibodies nor the use of CD18-deficient DC affected the ICAM-2-specific TEM. In humans, the C-type lectin DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN) was shown to interact with ICAM-2, suggesting a similar role in mice. However, we find that none of the murine DC-SIGN homologues mDC-SIGN, murine DC-SIGN-related molecule-1 (mSIGN-R1) and mSIGN-R3 is expressed on the surface of bone marrow-derived mouse DC. Taken together, this study shows that ICAM-2 strongly supports transmigration of immature DC across resting endothelium by interacting with ligands that are distinct from beta2-integrins and DC-SIGN homologues.
Collapse
Affiliation(s)
- Klaus Wethmar
- Max Planck Institute for Molecular Biomedicine, Münster, Germany, and Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Neyrolles O, Gicquel B, Quintana-Murci L. Towards a crucial role for DC-SIGN in tuberculosis and beyond. Trends Microbiol 2006; 14:383-7. [PMID: 16876999 DOI: 10.1016/j.tim.2006.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 05/19/2006] [Accepted: 07/18/2006] [Indexed: 11/28/2022]
Abstract
The C-type lectin DC-SIGN has recently received considerable attention in the context of tuberculosis and other infectious diseases. Clinical investigations, together with genetic association studies, strongly support the notion that variation in the level of expression of DC-SIGN - but not changes in the structure of the protein - might have a strong impact on the susceptibility to and pathogenesis of several infectious diseases, including tuberculosis. In addition, efforts to decipher the evolutionary history of the gene encoding DC-SIGN clearly demonstrated that this gene is under strong selective constraints that have prevented the accumulation of amino acid changes over time. Altogether, these findings suggest that DC-SIGN might play a crucial part in host immunity to pathogens and possibly beyond, at an early stage of human development.
Collapse
Affiliation(s)
- Olivier Neyrolles
- Unit of Mycobacterial Genetics, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.
| | | | | |
Collapse
|
75
|
Lee SJ, Arora R, Bull LM, Arduino RC, Garza L, Allan J, Kimata JT, Zhou P. A nonneutralizing anti-HIV Type 1 antibody turns into a broad neutralizing antibody when expressed on the surface of HIV type 1-susceptible cells. II. Inhibition of HIV type 1 captured and transferred by DC-SIGN. AIDS Res Hum Retroviruses 2006; 22:874-83. [PMID: 16989613 DOI: 10.1089/aid.2006.22.874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previously, we demonstrated that the expression of a nonneutralizing human anti-HIV-1 gp41 scFv on the surface of HIV-1-susceptible cells markedly inhibits HIV-1 replication and HIV-1 envelope-mediated cell-cell fusion. The inhibition is at the level of viral entry, specific for the HIV-1 envelope, and independent of virus tropism. In the previous studies, cell-free viruses of laboratory-adapted HIV-1 strains from subtype B were used to infect human CD4 T cell lines. To further test the effectiveness of this membrane-bound scFv (m-scFv) on HIV-1 infection, in this study, we carried out experiments to determine whether the m-scFv can neutralize infection of primary isolates from various HIV-1 subtypes and whether the m-scFv can neutralize HIV-1 captured and transferred by DC-SIGN on the surface of monocytic cell lines or DCs. We demonstrated that the m-scFv markedly inhibits primary isolates derived from various subtypes and significantly blocks HIV-1 captured and transferred by DC-SIGN on monocytic cell lines and on human DCs. Therefore, a nonneutralizing antibody acts as a broad neutralizing antibody when expressed on the cell surface, which significantly inhibits infection of both cell-free and DC-SIGN-captured and transferred virus. Our studies further point out the potential use of m-scFv as a inhibitor against HIV-1 transmission as well as a tool to dissect the mechanism of HIV-1 entry via DC-SIGN capture and transfer to CD4 T cells.
Collapse
Affiliation(s)
- Seung-Jae Lee
- Department of Virology and Immunology, Southwest Foundation for Biomedical Research, San Antonio, Texas 78249, USA
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Rappocciolo G, Piazza P, Fuller CL, Reinhart TA, Watkins SC, Rowe DT, Jais M, Gupta P, Rinaldo CR. DC-SIGN on B lymphocytes is required for transmission of HIV-1 to T lymphocytes. PLoS Pathog 2006; 2:e70. [PMID: 16839201 PMCID: PMC1500807 DOI: 10.1371/journal.ppat.0020070] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Accepted: 05/31/2006] [Indexed: 01/10/2023] Open
Abstract
Infection of T cells by HIV-1 can occur through binding of virus to dendritic cell (DC)-specific ICAM-3 grabbing nonintegrin (DC-SIGN) on dendritic cells and transfer of virus to CD4+ T cells. Here we show that a subset of B cells in the blood and tonsils of normal donors expressed DC-SIGN, and that this increased after stimulation in vitro with interleukin 4 and CD40 ligand, with enhanced expression of activation and co-stimulatory molecules CD23, CD58, CD80, and CD86, and CD22. The activated B cells captured and internalized X4 and R5 tropic strains of HIV-1, and mediated trans infection of T cells. Pretreatment of the B cells with anti–DC-SIGN monoclonal antibody blocked trans infection of T cells by both strains of HIV-1. These results indicate that DC-SIGN serves as a portal on B cells for HIV-1 infection of T cells in trans. Transmission of HIV-1 from B cells to T cells through this DC-SIGN pathway could be important in the pathogenesis of HIV-1 infection. A cell surface molecule, DC-SIGN, is known to bind the AIDS virus, human immunodeficiency virus 1 (HIV-1), on dendritic cells. HIV-1 can then be transferred from these dendritic cells to CD4+ T cells, in which the virus replicates and kills the T cells. Here, Rappocciolo and colleagues present their findings that DC-SIGN serves a similar function on a subset of B cells of the peripheral blood and tonsils. Although B cells that express DC-SIGN do not replicate HIV-1, they serve as portals for transfer and enhanced HIV-1 infection of CD4+ T cells, the major site of virus replication in the host. This newly described pathway for HIV-1 infection of T cells via B cells could be important in the pathogenesis of the virus infection.
Collapse
Affiliation(s)
- Giovanna Rappocciolo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Soilleux EJ, Sarno EN, Hernandez MO, Moseley E, Horsley J, Lopes UG, Goddard MJ, Vowler SL, Coleman N, Shattock RJ, Sampaio EP. DC-SIGN association with the Th2 environment of lepromatous lesions: cause or effect? J Pathol 2006; 209:182-9. [PMID: 16583355 DOI: 10.1002/path.1972] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The clinical spectrum of leprosy is related to patients' immune responses. Non-responsiveness towards Mycobacterium leprae (ML) seems to correlate with a Th2 cytokine profile. The reason for such a polarized immune response remains unclear. The C-type lectin, DC-SIGN, expressed by subsets of dendritic cells (DCs) and macrophages, has previously been associated with Th2 responses. Here we show abundant DC-SIGN expression in lepromatous but not borderline tuberculoid leprosy, in both HIV-positive and HIV-negative patients. Moreover, we demonstrate that DC-SIGN can act as an entry receptor for ML, as it does for M. tuberculosis, through the cell wall component lipoarabinomannan. DC-SIGN is expressed on virtually all ML-containing cells, providing further evidence for its role as a receptor. DC-SIGN may therefore be induced on macrophages in lepromatous leprosy and may then contribute to mycobacterial entry into these cells.
Collapse
Affiliation(s)
- E J Soilleux
- Department of Histopathology, Papworth Hospital, Papworth Everard, Cambridge CB3 8RE, and Nuffield Department of Clinical Laboratory Sciences, University of Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Giacomini E, Sotolongo A, Iona E, Severa M, Remoli ME, Gafa V, Lande R, Fattorini L, Smith I, Manganelli R, Coccia EM. Infection of human dendritic cells with a Mycobacterium tuberculosis sigE mutant stimulates production of high levels of interleukin-10 but low levels of CXCL10: impact on the T-cell response. Infect Immun 2006; 74:3296-304. [PMID: 16714557 PMCID: PMC1479299 DOI: 10.1128/iai.01687-05] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Mycobacterium tuberculosis genome encodes 13 sigma factors. We have previously shown that mutations in some of these transcriptional activators render M. tuberculosis sensitive to various environmental stresses and can attenuate the virulence phenotype. In this work, we focused on extracytoplasmic factor sigmaE and studied the effects induced by the deletion of its structural gene (sigE) in the infection of human monocyte-derived dendritic cells (MDDC). We found that the wild-type M. tuberculosis strain (H37Rv), the sigE mutant (ST28), and the complemented strain (ST29) were able to infect dendritic cells (DC) to similar extents, although at 4 days postinfection a reduced ability to grow inside MDDC was observed for the sigE mutant ST28. After mycobacterium capture, the majority of MDDC underwent full maturation and expressed both inflammatory cytokines, such as tumor necrosis factor alpha, and the regulatory cytokines interleukin-12 (IL-12), IL-18, and beta interferon (IFN-beta). Conversely, a higher level of production of IL-10 was observed in ST28-infected MDDC compared to H37Rv- or ST29-infected cell results. However, in spite of the presence of IL-10, supernatants from ST28-infected DC induced IFN-gamma production by T cells similarly to those from H37Rv-infected DC culture. On the other hand, IL-10 impaired CXCL10 production in sigE mutant-infected DC and, indeed, its neutralization restored CXCL10 secretion. In line with these results, supernatants from ST28-infected cells showed a decreased capability to recruit CXCR3+ CD4+ T cells compared to those obtained from H37Rv-infected DC culture. Thus, our findings suggest that the sigE mutant-induced secretion of IL-10 inhibits CXCL10 expression and, in turn, the recruitment of activated-effector cells involved in the formation of granulomas.
Collapse
Affiliation(s)
- Elena Giacomini
- Department of Infectious, Parasitic, and Immuno-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Schneider-Schaulies S, Dittmer U. Silencing T cells or T-cell silencing: concepts in virus-induced immunosuppression. J Gen Virol 2006; 87:1423-1438. [PMID: 16690907 DOI: 10.1099/vir.0.81713-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The ability to evade or suppress the host's immune response is a property of many viruses, indicating that this provides an advantage for the pathogen to spread efficiently or even to establish a persistent infection. The type and complexity of its genome and cell tropism but also its preferred type of host interaction are important parameters which define the strategy of a given virus to modulate the immune system in an optimal manner. Because they take a central position in any antiviral defence, the activation and function of T cells are the predominant target of many viral immunosuppressive regimens. In this review, two different strategies whereby this could be achieved are summarized. Retroviruses can infect professional antigen-presenting cells and impair their maturation and functional properties. This coincides with differentiation and expansion of silencing T cells referred to as regulatory T cells with suppressive activity, mainly to CD8+ effector T cells. The second concept, outlined for measles virus, is a direct, contact-mediated silencing of T cells which acquire a transient paralytic state.
Collapse
Affiliation(s)
| | - Ulf Dittmer
- Institut für Virologie des Universitätsklinikums Essen, D-45122 Essen, Germany
| |
Collapse
|
80
|
Steeghs L, van Vliet SJ, Uronen-Hansson H, van Mourik A, Engering A, Sanchez-Hernandez M, Klein N, Callard R, van Putten JPM, van der Ley P, van Kooyk Y, van de Winkel JGJ. Neisseria meningitidis expressing lgtB lipopolysaccharide targets DC-SIGN and modulates dendritic cell function. Cell Microbiol 2006; 8:316-25. [PMID: 16441441 DOI: 10.1111/j.1462-5822.2005.00623.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neisseria meningitidis lipopolysaccharide (LPS) has been identified as a major determinant of dendritic cell (DC) function. Here we report that one of a series of meningococcal mutants with defined truncations in the lacto-N-neotetraose outer core of the LPS exhibited unique strong adhesion and internalization properties towards DC. These properties were mediated by interaction of the GlcNAc(beta1-3)-Gal(beta1-4)-Glc-R oligosaccharide outer core of lgtB LPS with the dendritic-cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) lectin receptor. Activation of DC-SIGN with this novel oligosaccharide ligand skewed T-cell responses driven by DC towards T helper type 1 activity. Thus, the use of lgtB LPS may provide a powerful instrument to selectively induce the desired arm of the immune response and potentially increase vaccine efficacy.
Collapse
Affiliation(s)
- Liana Steeghs
- Utrecht University, Department of Infectious Diseases and Immunology, PO Box 80.165, 3508 TD Utrecht, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Rappocciolo G, Jenkins FJ, Hensler HR, Piazza P, Jais M, Borowski L, Watkins SC, Rinaldo CR. DC-SIGN is a receptor for human herpesvirus 8 on dendritic cells and macrophages. THE JOURNAL OF IMMUNOLOGY 2006; 176:1741-9. [PMID: 16424204 DOI: 10.4049/jimmunol.176.3.1741] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human herpesvirus 8 (HHV-8) causes Kaposi's sarcoma and pleural effusion lymphoma. In this study, we show that dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN; CD209) is a receptor for HHV-8 infection of myeloid DCs and macrophages. DC-SIGN was required for virus attachment to these cells and DC-SIGN-expressing cell lines. HHV-8 binding and infection were blocked by anti-DC-SIGN mAb and soluble DC-SIGN, and mannan, a natural ligand for DC-SIGN. Infection of DCs and macrophages with HHV-8 led to production of viral proteins, with little production of viral DNA, similar to HHV-8 infection of vascular endothelial cells. Infection of DCs resulted in down-regulation of DC-SIGN, a decrease in endocytic activity, and an inhibition of Ag stimulation of CD8+ T cells. We propose that DC-SIGN serves as a portal for immune dysfunction and oncogenesis caused by HHV-8 infection.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/metabolism
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cell Line
- Cell Line, Transformed
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Herpesviridae Infections/immunology
- Herpesvirus 8, Human/metabolism
- Humans
- Integrin alpha3beta1/physiology
- K562 Cells
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Macrophages/metabolism
- Mannans/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
Collapse
Affiliation(s)
- Giovanna Rappocciolo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Pitarque S, Herrmann JL, Duteyrat JL, Jackson M, Stewart G, Lecointe F, Payre B, Schwartz O, Young D, Marchal G, Lagrange P, Puzo G, Gicquel B, Nigou J, Neyrolles O. Deciphering the molecular bases of Mycobacterium tuberculosis binding to the lectin DC-SIGN reveals an underestimated complexity. Biochem J 2006; 392:615-24. [PMID: 16092920 PMCID: PMC1316302 DOI: 10.1042/bj20050709] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interactions between dendritic cells and Mycobacterium tuberculosis, the aetiological agent of tuberculosis in humans, are thought to be central to anti-mycobacterial immunity. We have previously shown that M. tuberculosis binds to human monocyte-derived dendritic cells mostly through the C-type lectin DC-SIGN (dendritic-cell-specific intercellular molecule-3-grabbing non-integrin)/CD209, and we have suggested that DC-SIGN may discriminate between mycobacterial species through recognition of the mannose-capping residues on the lipoglycan lipoarabinomannan of the bacterial envelope. Here, using a variety of fast- and slow-growing Mycobacterium species, we provide further evidence that mycobacteria recognition by DC-SIGN may be restricted to species of the M. tuberculosis complex. Fine analyses of the lipoarabinomannan molecules purified from these species show that the structure and amount of these molecules alone cannot account for such a preferential recognition. We propose that M. tuberculosis recognition by DC-SIGN relies on both a potential difference of accessibility of lipoarabinomannan in its envelope and, more probably, on the binding of additional ligands, possibly including lipomannan, mannose-capped arabinomannan, as well as the mannosylated 19 kDa and 45 kDa [Apa (alanine/proline-rich antigen)] glycoproteins. Altogether, our results reveal that the molecular basis of M. tuberculosis binding to DC-SIGN is more complicated than previously thought and provides further insight into the mechanisms of M. tuberculosis recognition by the immune system.
Collapse
Affiliation(s)
- Sylvain Pitarque
- *Institut de Pharmacologie et de Biologie Structurale, Département Mécanismes Moléculaires des Infections Mycobactériennes, CNRS UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Jean-Louis Herrmann
- ‡Université Lariboisière-Saint Louis, Equipe d'Accueil EA3510, Hôpital Saint-Louis, Service de Microbiologie, 1 avenue C. Vellefaux, 75010 Paris, France
| | - Jean-Luc Duteyrat
- †Université Paul Sabatier, Centre de Microscopie Electronique Appliquée à la Biologie, route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Mary Jackson
- §Unité de Génétique Mycobactérienne, 28 rue du Dr Roux, 75015 Paris, France
| | - Graham R. Stewart
- ††Centre for Molecular Microbiology and Infection, Department of Biological Sciences, South Kensington Campus, Imperial College London, London SW7 2AZ, U.K
| | - François Lecointe
- ∥Institut Pasteur, Unité de Génétique Mycobactérienne, 28 rue du Dr Roux, 75015 Paris, France
| | - Bruno Payre
- †Université Paul Sabatier, Centre de Microscopie Electronique Appliquée à la Biologie, route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Olivier Schwartz
- ¶Institut Pasteur, Groupe Virus et Immunité, 28 rue du Dr Roux, 75015 Paris, France
| | - Douglas B. Young
- ††Centre for Molecular Microbiology and Infection, Department of Biological Sciences, South Kensington Campus, Imperial College London, London SW7 2AZ, U.K
| | - Gilles Marchal
- ¶Institut Pasteur, Groupe Virus et Immunité, 28 rue du Dr Roux, 75015 Paris, France
| | - Philippe H. Lagrange
- ‡Université Lariboisière-Saint Louis, Equipe d'Accueil EA3510, Hôpital Saint-Louis, Service de Microbiologie, 1 avenue C. Vellefaux, 75010 Paris, France
| | - Germain Puzo
- *Institut de Pharmacologie et de Biologie Structurale, Département Mécanismes Moléculaires des Infections Mycobactériennes, CNRS UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Brigitte Gicquel
- §Unité de Génétique Mycobactérienne, 28 rue du Dr Roux, 75015 Paris, France
| | - Jérôme Nigou
- *Institut de Pharmacologie et de Biologie Structurale, Département Mécanismes Moléculaires des Infections Mycobactériennes, CNRS UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Olivier Neyrolles
- §Unité de Génétique Mycobactérienne, 28 rue du Dr Roux, 75015 Paris, France
- ††Centre for Molecular Microbiology and Infection, Department of Biological Sciences, South Kensington Campus, Imperial College London, London SW7 2AZ, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
83
|
Tailleux L, Pham-Thi N, Bergeron-Lafaurie A, Herrmann JL, Charles P, Schwartz O, Scheinmann P, Lagrange PH, de Blic J, Tazi A, Gicquel B, Neyrolles O. DC-SIGN induction in alveolar macrophages defines privileged target host cells for mycobacteria in patients with tuberculosis. PLoS Med 2005; 2:e381. [PMID: 16279841 PMCID: PMC1283365 DOI: 10.1371/journal.pmed.0020381] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Accepted: 09/15/2005] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Interplays between Mycobacterium tuberculosis, the etiological agent of tuberculosis (TB) in human and host professional phagocytes, namely macrophages (Mphis) and dendritic cells (DCs), are central to immune protection against TB and to TB pathogenesis. We and others have recently shown that the C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN; CD209) mediates important interactions between mycobacteria and human monocyte-derived DCs (MoDCs) in vitro. METHODS AND FINDINGS In order to explore the possible role of DC-SIGN in M. tuberculosis infection in vivo, we have analysed DC-SIGN expression in broncho-alveolar lavage (BAL) cells from patients with TB (n = 40) or with other non-mycobacterial lung pathologies, namely asthma (n = 14) and sarcoidosis (n = 11), as well as from control individuals (n = 9). We show that in patients with TB, up to 70% of alveolar Mphis express DC-SIGN. By contrast, the lectin is barely detected in alveolar Mphis from all other individuals. Flow cytometry, RT-PCR, and enzyme-linked immunosorbent assay analyses of BAL-derived fluids and cells indicated that M. tuberculosis infection induces DC-SIGN expression in alveolar Mphis by a mechanism that is independent of Toll-like receptor-4, interleukin (IL)-4, and IL-13. This mechanism most likely relies on the secretion of soluble host and/or mycobacterial factors that have yet to be identified, as both infected and uninfected bystander Mphis were found to express DC-SIGN in the presence of M. tuberculosis. Immunohistochemical examination of lung biopsy samples from patients with TB showed that the bacilli concentrate in pulmonary regions enriched in DC-SIGN-expressing alveolar Mphis in vivo. Ex vivo binding and inhibition of binding experiments further revealed that DC-SIGN-expressing alveolar Mphis constitute preferential target cells for M. tuberculosis, as compared to their DC-SIGN- counterparts. In contrast with what has been reported previously in MoDCs in vitro, ex vivo DC-SIGN ligation by mycobacterial products failed to induce IL-10 secretion by alveolar Mphis, and IL-10 was not detected in BALs from patients with TB. CONCLUSION Altogether, our results provide further evidence for an important role of DC-SIGN during TB in humans. DC-SIGN induction in alveolar Mphis may have important consequences on lung colonization by the tubercle bacillus, and on pulmonary inflammatory and immune responses in the infected host.
Collapse
Affiliation(s)
- Ludovic Tailleux
- 1Institut Pasteur, Unité de Génétique Mycobactérienne, Paris, France
- *To whom correspondence should be addressed. E-mail: (LT), E-mail: (ON)
| | - Nhan Pham-Thi
- 2Hôpital Necker-Enfants-Malades, AP-HP, Service de Pneumologie et d'Allergologie Pédiatrique, Paris, France
| | | | | | - Patricia Charles
- 1Institut Pasteur, Unité de Génétique Mycobactérienne, Paris, France
| | | | - Pierre Scheinmann
- 2Hôpital Necker-Enfants-Malades, AP-HP, Service de Pneumologie et d'Allergologie Pédiatrique, Paris, France
| | | | - Jacques de Blic
- 2Hôpital Necker-Enfants-Malades, AP-HP, Service de Pneumologie et d'Allergologie Pédiatrique, Paris, France
| | - Abdellatif Tazi
- 3Hôpital Saint-Louis, AP-HP, Service de Pneumologie, Paris, France
| | - Brigitte Gicquel
- 1Institut Pasteur, Unité de Génétique Mycobactérienne, Paris, France
| | - Olivier Neyrolles
- 1Institut Pasteur, Unité de Génétique Mycobactérienne, Paris, France
- 6Centre National de la Recherche Scientifique, URA 2172, Paris, France
- *To whom correspondence should be addressed. E-mail: (LT), E-mail: (ON)
| |
Collapse
|
84
|
Chen CH, Floyd H, Olson NE, Magaletti D, Li C, Draves K, Clark EA. Dendritic-cell-associated C-type lectin 2 (DCAL-2) alters dendritic-cell maturation and cytokine production. Blood 2005; 107:1459-67. [PMID: 16239426 PMCID: PMC1895401 DOI: 10.1182/blood-2005-08-3264] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dendritic-cell (DC)-associated C-type lectin receptors (CLRs) take up antigens to present to T cells and regulate DC functions. DCAL-2 is a CLR with a cytosolic immunoreceptor tyrosine-based inhibitory motif (ITIM), which is restricted to immature DCs (iDCs), monocytes, and CD1a+ DCs. Cross-linking DCAL-2 on iDCs induced protein tyrosine phosphorylation and MAPK activation as well as receptor internalization. To test if DCAL-2 is involved in DC maturation and cytokine expression, we stimulated iDCs with anti-DCAL-2 mAb with or without LPS, zymosan, or CD40L. While anti-DCAL-2 did not induce iDCs to mature, it did up-regulate CCR7 expression and IL-6 and IL-10 production. DCAL-2 signals augmented DC maturation induced by LPS or zymosan, increasing both CCR7 and DC-LAMP expression. Of interest, DCAL-2 ligation had the opposite effects on TLR versus CD40L signaling: anti-DCAL-2 suppressed TLR-induced IL-12 expression, but significantly enhanced CD40L-induced IL-12 production. DCAL-2 ligation also suppressed the ability of TLR-matured DCs to induce IFN-gamma-secreting Th1 cells but augmented the capacity of CD40L-matured DCs to polarize naive T cells into Th1 cells. Thus, DCAL-2 may program DCs differently depending on whether DCs are signaled via TLRs or by T cells. DCAL-2 may be a potential immunotherapeutic target for modulating autoimmune diseases or for developing vaccines.
Collapse
Affiliation(s)
- Chang-Hung Chen
- Department of Immunology, Box 357330, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | |
Collapse
|
85
|
Tian W, Nunez R, Cheng S, Ding Y, Tumang J, Lyddane C, Roman C, Liou HC. C-type lectin OCILRP2/Clr-g and its ligand NKRP1f costimulate T cell proliferation and IL-2 production. Cell Immunol 2005; 234:39-53. [PMID: 15963483 DOI: 10.1016/j.cellimm.2005.04.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 04/14/2005] [Accepted: 04/25/2005] [Indexed: 01/01/2023]
Abstract
We are reporting the identification of a novel C-type lectin receptor-ligand pair that is involved in T cell costimulation. The receptor, OCILRP2/Clr-g, is rapidly induced following T cell activation and maintained at a substantial level of up to 72 h. The ligand, NKRP1f, is predominantly expressed on dendritic cells (DC). The soluble OCILRP2-Ig blocking protein significantly suppresses specific antigen-stimulated T cell proliferation as well as IL-2 secretion both in vitro and in vivo; conversely, NKRP1f-expressing antigen presenting cells (APC) enhance B7.1/CD28-mediated costimulation for T cell proliferation through interaction with OCILRP2/Clr-g. Our studies reveal a unique functional interaction between two C-type lectins, OCILRP2/Clr-g and NKRP1f, during APC-mediated T cell costimulation and suggest a role for C-type lectins in maintaining T cell response or memory in vivo.
Collapse
Affiliation(s)
- Wenzhi Tian
- Division of Immunology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Becker Y. HIV-1 gp120 binding to dendritic cell receptors mobilize the virus to the lymph nodes, but the induced IL-4 synthesis by FcepsilonRI+ hematopoietic cells damages the adaptive immunity--a review, hypothesis, and implications. Virus Genes 2004; 29:147-65. [PMID: 15215692 DOI: 10.1023/b:viru.0000032797.43537.d3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
HIV-1 is equipped with the envelope gp160 glycoprotein for interaction with Langerhans cells (LCs) and dendritic cells (DCs), the members of the innate immune system, which confront the virus at the portal of virus entry in the human body. These cells are equipped with receptors by which they bind and endocytose the virus. The gp120 glycoprotein is used for binding to CD4 receptor and CCR5 co-receptor of T helper 2 (Th2) cells and the virions shed gp120 is able to induce FcepsilonRI+ hematopoietic cells to produce IL-4, which inactivate the host adaptive immune response. The properties of gp120s various functional domains are analyzed together with the regulatory viral proteins, which are involved in the damage to T and B cells during HIV-1 replication. The interaction of HIV-1 virions through their gp120 with LCs and DCs at the portal of virus entry will be discussed. A hypothesis will be presented that the understanding of the role of the different functional domains of gp120 in the life cycle of the virus and during AIDS will help in the design of approaches to prevent and abrogate HIV-1 infection and AIDS.
Collapse
Affiliation(s)
- Yechiel Becker
- Department of Molecular Virology, Faculty of Medicine, the Hebrew University of Jerusalem, P.O. Box 12272, 91120, Israel.
| |
Collapse
|
87
|
Abstract
The filoviruses, marburgvirus and ebolavirus, cause epidemics of haemorrhagic fever with high case-fatality rates. The severe illness results from a complex of pathogenetic mechanisms that enable the virus to suppress innate and adaptive immune responses, infect and kill a broad variety of cell types, and elicit strong inflammatory responses and disseminated intravascular coagulation, producing a syndrome resembling septic shock. Most experimental data have been obtained on Zaire ebolavirus, which causes uniformly lethal disease in experimentally infected non-human primates but produces a broader range of outcomes in naturally infected human beings. 10-30% of patients can survive the illness by mobilising adaptive immune responses, and there is limited evidence that mild or symptomless infections also occur. The other filoviruses that have caused human disease, Sudan ebolavirus, Ivory Coast ebolavirus, and marburgvirus, produce a similar illness but with somewhat lower case-fatality rates. Variations in outcome during an epidemic might be due partly to genetically determined differences in innate immune responses to the viruses. Recent studies in non-human primates have shown that blocking of certain host responses, such as the coagulation cascade, can result in reduced viral replication and improved host survival.
Collapse
Affiliation(s)
- Siddhartha Mahanty
- Malaria Vaccine Development Unit, at the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | |
Collapse
|
88
|
Kanazawa N, Tashiro K, Miyachi Y. Signaling and immune regulatory role of the dendritic cell immunoreceptor (DCIR) family lectins: DCIR, DCAR, dectin-2 and BDCA-2. Immunobiology 2004; 209:179-90. [PMID: 15481152 DOI: 10.1016/j.imbio.2004.03.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This review focuses on a distinct family of dendritic cells (DC) expressing C-type lectins that include DC immunoreceptor (DCIR), DC immunoactivating receptor (DCAR), DC-associated C-type lectin (dectin)-2 and blood DC antigen (BDCA)-2. DCIR is a type II C-type lectin expressed on antigen presenting cells and granulocytes and acts as an inhibitory receptor via an intracellular immunoreceptor tyrosine-based inhibitory motif (ITIM). In contrast, DCAR has been identified as a molecule that forms a putative pair with DCIR. While both molecules share the highly homologous extracellular lectin domain, DCAR lacks the ITIM in its short cytoplasmic tail and acts as an activating receptor through association with the Fc receptor gamma chain. Two other lectins, dectin-2 and BDCA-2, are highly related to DCAR by similarities of their amino acid sequence, molecular structure and chromosomal localization. Although they also lack the ITIM, they are capable of transducing signal to regulate cellular functions positively or negatively. Here we propose to designate these four highly related molecules as the "DCIR family lectins" and discuss their signaling mechanisms, carbohydrate recognition, and other features that contribute to the function of DC to control immunity.
Collapse
Affiliation(s)
- Nobuo Kanazawa
- Department of Dermatology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | |
Collapse
|