151
|
Lee JH, Park DY, Lee KJ, Kim YK, So YK, Ryu JS, Oh SH, Han YS, Ko K, Choo YK, Park SJ, Brodzik R, Lee KK, Oh DB, Hwang KA, Koprowski H, Lee YS, Ko K. Intracellular reprogramming of expression, glycosylation, and function of a plant-derived antiviral therapeutic monoclonal antibody. PLoS One 2013; 8:e68772. [PMID: 23967055 PMCID: PMC3744537 DOI: 10.1371/journal.pone.0068772] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 06/03/2013] [Indexed: 01/19/2023] Open
Abstract
Plant genetic engineering, which has led to the production of plant-derived monoclonal antibodies (mAb(P)s), provides a safe and economically effective alternative to conventional antibody expression methods. In this study, the expression levels and biological properties of the anti-rabies virus mAb(P) SO57 with or without an endoplasmic reticulum (ER)-retention peptide signal (Lys-Asp-Glu-Leu; KDEL) in transgenic tobacco plants (Nicotiana tabacum) were analyzed. The expression levels of mAb(P) SO57 with KDEL (mAb(P)K) were significantly higher than those of mAb(P) SO57 without KDEL (mAb(P)) regardless of the transcription level. The Fc domains of both purified mAb(P) and mAb(P)K and hybridoma-derived mAb (mAb(H)) had similar levels of binding activity to the FcγRI receptor (CD64). The mAb(P)K had glycan profiles of both oligomannose (OM) type (91.7%) and Golgi type (8.3%), whereas the mAb(P) had mainly Golgi type glycans (96.8%) similar to those seen with mAb(H). Confocal analysis showed that the mAb(P)K was co-localized to ER-tracker signal and cellular areas surrounding the nucleus indicating accumulation of the mAb(P) with KDEL in the ER. Both mAb(P) and mAb(P)K disappeared with similar trends to mAb(H) in BALB/c mice. In addition, mAb(P)K was as effective as mAb(H) at neutralizing the activity of the rabies virus CVS-11. These results suggest that the ER localization of the recombinant mAb(P) by KDEL reprograms OM glycosylation and enhances the production of the functional antivirus therapeutic antibody in the plant.
Collapse
Affiliation(s)
- Jeong-Hwan Lee
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Da-Young Park
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Kyung-Jin Lee
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Young-Kwan Kim
- Department of Herbology, School of Oriental Medicine, Wonkwang University, Iksan, Korea
| | - Yang-Kang So
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jae-Sung Ryu
- Department of Biological Science, Biotechnology Institute, College of Natural Science, Wonkwang University, Iksan, Korea
| | - Seung-Han Oh
- Department of Agricultural Biology, College of Agriculture and Life Science, Chonnam National University, Gwangju, Korea
| | - Yeon-Soo Han
- Department of Agricultural Biology, College of Agriculture and Life Science, Chonnam National University, Gwangju, Korea
| | - Kinarm Ko
- Department of Neuroscience, School of Medicine, Konkuk University, Seoul, Korea
| | - Young-Kug Choo
- Department of Biological Science, Biotechnology Institute, College of Natural Science, Wonkwang University, Iksan, Korea
| | - Sung-Joo Park
- Department of Herbology, School of Oriental Medicine, Wonkwang University, Iksan, Korea
| | - Robert Brodzik
- Biotechnology Foundation Laboratories, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Kyoung-Ki Lee
- National Veterinary Research and Quarantine Service, Anyang, Korea
| | - Doo-Byoung Oh
- Korean Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Kyung-A Hwang
- Department of Agrofood Resources, National Academy of Agricultural Science, RDA, Suwon, Korea
| | - Hilary Koprowski
- Biotechnology Foundation Laboratories, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Yong Seong Lee
- Department of Urology, College of Medicine, Kangnam Sacred Heart Hospital, Hallym University, Seoul, Korea
| | - Kisung Ko
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
152
|
Probert F, Whittaker SBM, Crispin M, Mitchell DA, Dixon AM. Solution NMR analyses of the C-type carbohydrate recognition domain of DC-SIGNR protein reveal different binding modes for HIV-derived oligosaccharides and smaller glycan fragments. J Biol Chem 2013; 288:22745-57. [PMID: 23788638 PMCID: PMC3829359 DOI: 10.1074/jbc.m113.458299] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/13/2013] [Indexed: 01/08/2023] Open
Abstract
The C-type lectin DC-SIGNR (dendritic cell-specific ICAM-3-grabbing non-integrin-related; also known as L-SIGN or CD299) is a promising drug target due to its ability to promote infection and/or within-host survival of several dangerous pathogens (e.g. HIV and severe acute respiratory syndrome coronavirus (SARS)) via interactions with their surface glycans. Crystallography has provided excellent insight into the mechanism by which DC-SIGNR interacts with small glycans, such as (GlcNAc)2Man3; however, direct observation of complexes with larger, physiological oligosaccharides, such as Man9GlcNAc2, remains elusive. We have utilized solution-state nuclear magnetic resonance spectroscopy to investigate DC-SIGNR binding and herein report the first backbone assignment of its active, calcium-bound carbohydrate recognition domain. Direct interactions with the small sugar fragments Man3, Man5, and (GlcNAc)2Man3 were investigated alongside Man9GlcNAc derived from recombinant gp120 (present on the HIV viral envelope), providing the first structural data for DC-SIGNR in complex with a virus-associated ligand, and unique binding modes were observed for each glycan. In particular, our data show that DC-SIGNR has a different binding mode for glycans on the HIV viral envelope compared with the smaller glycans previously observed in the crystalline state. This suggests that using the binding mode of Man9GlcNAc, instead of those of small glycans, may provide a platform for the design of DC-SIGNR inhibitors selective for high mannose glycans (like those on HIV). (15)N relaxation measurements provided the first information on the dynamics of the carbohydrate recognition domain, demonstrating that it is a highly flexible domain that undergoes ligand-induced conformational and dynamic changes that may explain the ability of DC-SIGNR to accommodate a range of glycans on viral surfaces.
Collapse
Affiliation(s)
- Fay Probert
- From the Molecular Organisation and Assembly in Cells Doctoral Training Centre
| | - Sara B.-M. Whittaker
- the Henry Wellcome Building for Biomolecular NMR Spectroscopy, Birmingham Cancer Research UK Centre, School of Cancer Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, United Kingdom, and
| | - Max Crispin
- the Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | | | - Ann M. Dixon
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
153
|
Macrophages in tuberculosis: friend or foe. Semin Immunopathol 2013; 35:563-83. [PMID: 23864058 DOI: 10.1007/s00281-013-0388-2] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/20/2013] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis (Mtb), is acquired by the respiratory route. It is exquisitely human adapted and a prototypic intracellular pathogen of macrophages, with alveolar macrophages (AMs) being the primary conduit of infection and disease. The outcome of primary infection is most often a latently infected healthy human host, in whom the bacteria are held in check by the host immune response. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the host immune response fails to control the growth of bacilli, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols coughed out into the environment and inhaled by new hosts. The molecular details of the Mtb-macrophage interaction continue to be elucidated. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. Macrophages demonstrate tremendous phenotypic heterogeneity and functional plasticity which, depending on the site and stage of infection, facilitate the diverse outcomes. Moreover, host responses vary depending on the specific characteristics of the infecting Mtb strain. In this chapter, we describe a contemporary view of the behavior of AMs and their interaction with various Mtb strains in generating unique immunologic lung-specific responses.
Collapse
|
154
|
Abstract
A number of advances in recent years have significantly furthered our understanding of filovirus attachment and cellular tropism. For example, several cell-surface molecules have been identified as attachment factors with the potential to facilitate the in vivo targeting of particular cell types such as macrophages and hepatic cells. Furthermore, our knowledge of internalization and subsequent events during filovirus entry has also been widened, adding new variations to the paradigms for viral entry established for HIV and influenza. In particular, host cell factors such as endosomal proteases and the intracellular receptor Niemann-Pick C1 are now known to play a vital role in activating the membrane fusion potential of filovirus glycoproteins.
Collapse
Affiliation(s)
- Stefan Pöhlmann
- grid.10423.340000000095299877Institute for Virology, Hannover Medical School, Hannover, Germany ,grid.418215.b0000000085027018German Primate Center, Göttingen, Germany
| | - Graham Simmons
- grid.266102.10000000122976811Blood Systems Research Institute, and Department of Laboratory Medicine, University of California San Francisco, San Francisco, California USA
| |
Collapse
|
155
|
Mannodendrimers prevent acute lung inflammation by inhibiting neutrophil recruitment. Proc Natl Acad Sci U S A 2013; 110:8795-800. [PMID: 23671078 DOI: 10.1073/pnas.1221708110] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mycobacterium tuberculosis mannose-capped lipoarabinomannan inhibits the release of proinflammatory cytokines by LPS-stimulated human dendritic cells (DCs) via targeting the C-type lectin receptor DC-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN). With the aim of mimicking the bioactive supramolecular structure of mannose-capped lipoarabinomannan, we designed and synthesized a set of poly(phosphorhydrazone) dendrimers grafted with mannose units, called mannodendrimers, that differed by size and the number and length of their (α1→2)-oligommanoside caps. A third-generation dendrimer bearing 48 trimannoside caps (3T) and a fourth-generation dendrimer bearing 96 dimannosides (4D) displayed the highest binding avidity for DC-SIGN. Moreover, these dendrimers inhibited proinflammatory cytokines, including TNF-α, production by LPS-stimulated DCs in a DC-SIGN-dependent fashion. Finally, in a model of acute lung inflammation in which mice were exposed to aerosolized LPS, per os administration of 3T mannodendrimer was found to significantly reduce neutrophil influx via targeting the DC-SIGN murine homolog SIGN-related 1. The 3T mannodendrimer therefore represents an innovative fully synthetic compound for the treatment of lung inflammatory diseases.
Collapse
|
156
|
Yu X, Vasiljevic S, Mitchell DA, Crispin M, Scanlan CN. Dissecting the Molecular Mechanism of IVIg Therapy: The Interaction between Serum IgG and DC-SIGN is Independent of Antibody Glycoform or Fc Domain. J Mol Biol 2013; 425:1253-8. [DOI: 10.1016/j.jmb.2013.02.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/04/2013] [Accepted: 02/06/2013] [Indexed: 01/20/2023]
|
157
|
Shah M, Wadood A, Rahman Z, Husnain T. Interaction and inhibition of dengue envelope glycoprotein with mammalian receptor DC-sign, an in-silico approach. PLoS One 2013; 8:e59211. [PMID: 23527139 PMCID: PMC3601059 DOI: 10.1371/journal.pone.0059211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/12/2013] [Indexed: 12/23/2022] Open
Abstract
Membrane fusion is the central molecular event during the entry of enveloped viruses into cells. The critical agents of this process are viral surface proteins, primed to facilitate cell bilayer fusion. The important role of Dendritic-cell-specific ICAM3-grabbing non-integrin (DC-SIGN) in Dengue virus transmission makes it an attractive target to interfere with Dengue virus Propagation. Receptor mediated endocytosis allows the entry of virions due to the presence of endosomal membranes and low pH-induced fusion of the virus. DC-SIGN is the best characterized molecule among the candidate protein receptors and is able to mediate infection with the four serotypes of dengue virus (DENV). Unrestrained pair wise docking was used for the interaction of dengue envelope protein with DC-SIGN and monoclonal antibody 2G12. Pre-processed the PDB coordinates of dengue envelope glycoprotein and other candidate proteins were prepared and energy minimized through AMBER99 force field distributed in MOE software. Protein-protein interaction server, ZDOCK was used to find molecular interaction among the candidate proteins. Based on these interactions it was found that antibody successfully blocks the glycosylation site ASN 67 and other conserved residues present at DC-SIGN-Den-E complex interface. In order to know for certain, the exact location of the antibody in the envelope protein, co-crystallize of the envelope protein with these compounds is needed so that their exact docking locations can be identified with respect to our results.
Collapse
Affiliation(s)
- Masaud Shah
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
- Computational Medicinal Chemistry Laboratory, Department of Biochemistry Abdul Wali Khan University, Mardan, Pakistan
| | - Abdul Wadood
- Computational Medicinal Chemistry Laboratory, Department of Biochemistry Abdul Wali Khan University, Mardan, Pakistan
- * E-mail: (AW); (ZR)
| | - Ziaur Rahman
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
- * E-mail: (AW); (ZR)
| | - Tayyab Husnain
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
158
|
Surfactant Protein D modulates HIV infection of both T-cells and dendritic cells. PLoS One 2013; 8:e59047. [PMID: 23527085 PMCID: PMC3601116 DOI: 10.1371/journal.pone.0059047] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 02/11/2013] [Indexed: 11/19/2022] Open
Abstract
Surfactant Protein D (SP-D) is an oligomerized C-type lectin molecule with immunomodulatory properties and involvement in lung surfactant homeostasis in the respiratory tract. SP-D binds to the enveloped viruses, influenza A virus and respiratory syncytial virus and inhibits their replication in vitro and in vivo. SP-D has been shown to bind to HIV via the HIV envelope protein gp120 and inhibit infectivity in vitro. Here we show that SP-D binds to different strains of HIV (BaL and IIIB) and the binding occurs at both pH 7.4 and 5.0 resembling physiological relevant pH values found in the body and the female urogenital tract, respectively. The binding of SP-D to HIV particles and gp120 was inhibited by the presence of several hexoses with mannose found to be the strongest inhibitor. Competition studies showed that soluble CD4 and CVN did not interfere with the interaction between SP-D and gp120. However, soluble recombinant DC-SIGN was shown to inhibit the binding between SP-D and gp120. SP-D agglutinated HIV and gp120 in a calcium dependent manner. SP-D inhibited the infectivity of HIV strains at both pH values of 7.4 and 5.0 in a concentration dependent manner. The inhibition of the infectivity was abolished by the presence of mannose. SP-D enhanced the binding of HIV to immature monocyte derived dendritic cells (iMDDCs) and was also found to enhance HIV capture and transfer to the T-cell like line PM1. These results suggest that SP-D can bind to and inhibit direct infection of T-cells by HIV but also enhance the transfer of infectious HIV particles from DCs to T-cells in vivo.
Collapse
|
159
|
Zhang Q, Collins J, Anastasaki A, Wallis R, Mitchell DA, Becer CR, Haddleton DM. Sequence-Controlled Multi-Block Glycopolymers to Inhibit DC-SIGN-gp120 Binding. Angew Chem Int Ed Engl 2013; 52:4435-9. [DOI: 10.1002/anie.201300068] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/01/2013] [Indexed: 12/21/2022]
|
160
|
Sequence-Controlled Multi-Block Glycopolymers to Inhibit DC-SIGN-gp120 Binding. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201300068] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
161
|
Zhang Z, Chen K, Yan L, Yang Z, Zhu Z, Chen C, Zeng J, Wei W, Qi X, Ren S, Zuo Y. Low expression of dendritic cell-specific intercellular adhesion molecule-grabbing nonintegrin-related protein in non-Hodgkin lymphoma and significant correlations with lactic acid dehydrogenase and β2-microglobulin. Biochem Cell Biol 2013; 91:214-20. [PMID: 23859015 DOI: 10.1139/bcb-2012-0110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dendritic cell-specific intercellular adhesion molecule-grabbing nonintegrin-related protein (DC-SIGNR), a type II integral membrane protein and a member of the C-type lectins, has been reported to bind various strains of HIV-1, HIV-2, and simian immunodeficiency virus. Serum DC-SIGNR is not currently available for the detection of non-Hodgkin lymphoma (NHL). Using an enzyme-linked immunosorbent assay (ELISA), we assessed the serum levels of DC-SIGNR in 70 cancer patients and 100 healthy controls. Additionally, using immunohistochemistry, we determined the expression of DC-SIGNR in the lymph nodes. Using the ELISA, low serum levels of DC-SIGNR were detected in the patients (median, 4.513 ng·L(-1); range, 1.066-9.232 ng·L(-1); p = 0.0003). Serum concentrations of DC-SIGNR correlated significantly with age (p = 0.0077) and lactic acid dehydrogenase (p = 0.0046) and β2-microglobulin (p = 0.0491) levels. However, we found no statistically significant correlation between serum DC-SIGNR levels and clinical data such as sex, Ann Arbor stage, B symptoms, and histologic subtypes. Moreover, NHL patients with a lower level of serum DC-SIGNR expression in lymphatic endothelial cells also showed negative immunostaining levels. These results suggest that DC-SIGNR is a biological molecule that may be potentially useful in NHL clinical settings.
Collapse
Affiliation(s)
- Zhuqing Zhang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Garcia-Vallejo JJ, Koning N, Ambrosini M, Kalay H, Vuist I, Sarrami-Forooshani R, Geijtenbeek TBH, van Kooyk Y. Glycodendrimers prevent HIV transmission via DC-SIGN on dendritic cells. Int Immunol 2013; 25:221-33. [PMID: 23291968 DOI: 10.1093/intimm/dxs115] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells efficient in capturing pathogens, and processing their antigenic determinants for presentation to antigen-specific T cells to induce robust immune responses. Their location at peripheral tissues and the expression of pattern-recognition receptors, among them DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), facilitates the capture of pathogens before spreading. However, some pathogens have developed strategies to escape the immune system. One of the most successful is HIV-1, which targets DC-SIGN for transport to the lymph node where the virus infects CD4(+) T cells. Contact of HIV-1 with DC-SIGN is thus the first event in the pathogenic cascade and, therefore, it is the primary target point for therapies aimed at HIV infection prevention. DC-SIGN recognizes specific glycans on HIV-1 and this interaction can be blocked by competitive inhibition through glycans. Although the affinity of glycans is relatively low, multivalency may increase avidity and the strength to compete with HIV-1 virions. We have designed multivalent dendrimeric compounds based on Lewis-type antigens that bind DC-SIGN with high selectivity and avidity and that effectively block gp120 binding to DC-SIGN and, consequently, HIV transmission to CD4(+) T cells. Binding to DC-SIGN and gp120 inhibition was higher on glycodendrimers with larger molecular diameter, indicating that the geometry of the compounds is an important factor determining their functionality. Our compounds elicited DC-SIGN internalization, a property of the receptor upon triggering, but did not affect the maturation status of DCs. Thus, Le(X) glycodendrimers could be incorporated into topic prophylactic approaches for the prevention of HIV-1 transmission.
Collapse
Affiliation(s)
- Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Abstract
The key role of carbohydrates in many biological events has attracted the interest of the scientific community. This fact has demanded the access to new tools necessary to understand this role and the interaction of carbohydrates with their corresponding receptors, lectins. Glycodendrimers and glycodendritic structures in general, have demonstrated to be very efficient and interesting tools to intervene in those processes where carbohydrates participate. In this review, we discuss the different glycodendritic structures that have been used to interfere with DC-SIGN, a very attractive lectin involved in infection processes and in the regulation of the immune response.
Collapse
|
164
|
Richichi B, Imberty A, Gillon E, Bosco R, Sutkeviciute I, Fieschi F, Nativi C. Synthesis of a selective inhibitor of a fucose binding bacterial lectin from Burkholderia ambifaria. Org Biomol Chem 2013; 11:4086-94. [DOI: 10.1039/c3ob40520f] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
165
|
Hopkins RA, Connolly JE. The specialized roles of immature and mature dendritic cells in antigen cross-presentation. Immunol Res 2012; 53:91-107. [PMID: 22450675 DOI: 10.1007/s12026-012-8300-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Exogenous antigen cross-presentation is integral to the stimulation of cytotoxic T-lymphocytes against viruses and tumors. Central to this process are dendritic cells (DCs), which specialize in cross-presentation. DCs may be considered to exist in two radically different states of activation, generally referred to as immature and mature. In each of these states, the cell has a series of separate and specialized abilities for the induction of T-cell immunity. In the immature state, the DC is adept in surveying the periphery, acquiring and storing antigen, but has a limited capacity for direct T-cell activation. During a brief and defined window of time following DC stimulation, nearly every aspect of antigen handling changes, as it transitions from an entity focused on protein preservation to one capable of efficient cross-presentation. It is this time period and the underlying molecular mechanisms active here, which form the core of our studies on cross-presentation.
Collapse
Affiliation(s)
- Richard A Hopkins
- Program in Translational Immunology, Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03 Immunos, Biopolis, Singapore
| | | |
Collapse
|
166
|
García-Vallejo JJ, Ambrosini M, Overbeek A, van Riel WE, Bloem K, Unger WWJ, Chiodo F, Bolscher JG, Nazmi K, Kalay H, van Kooyk Y. Multivalent glycopeptide dendrimers for the targeted delivery of antigens to dendritic cells. Mol Immunol 2012; 53:387-97. [PMID: 23103377 DOI: 10.1016/j.molimm.2012.09.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/08/2012] [Accepted: 09/23/2012] [Indexed: 01/21/2023]
Abstract
Dendritic cells are the most powerful type of antigen presenting cells. Current immunotherapies targeting dendritic cells have shown a relative degree of success but still require further improvement. One of the most important issues to solve is the efficiency of antigen delivery to dendritic cells in order to achieve an appropriate uptake, processing, and presentation to Ag-specific T cells. C-type lectins have shown to be ideal receptors for the targeting of antigens to dendritic cells and allow the use of their natural ligands - glycans - instead of antibodies. Amongst them, dendritic cell-specific ICAM-3-grabbing non-integrin (DC-SIGN) is an interesting candidate due to its biological properties and the availability of its natural carbohydrate ligands. Using Le(b)-conjugated poly(amido amine) (PAMAM) dendrimers we aimed to characterize the optimal level of multivalency necessary to achieve the desired internalization, lysosomal delivery, Ag-specific T cell proliferation, and cytokine response. Increasing DC-SIGN ligand multivalency directly translated in an enhanced binding, which might also be interesting for blocking purposes. Internalization, routing to lysosomal compartments, antigen presentation and cytokine response could be optimally achieved with glycopeptide dendrimers carrying 16-32 glycan units. This report provides the basis for the design of efficient targeting of peptide antigens for the immunotherapy of cancer, autoimmunity and infectious diseases.
Collapse
Affiliation(s)
- Juan J García-Vallejo
- Department of Molecular Cell Biology & Immunology, Faculty of Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Manzo C, Torreno-Pina JA, Joosten B, Reinieren-Beeren I, Gualda EJ, Loza-Alvarez P, Figdor CG, Garcia-Parajo MF, Cambi A. The neck region of the C-type lectin DC-SIGN regulates its surface spatiotemporal organization and virus-binding capacity on antigen-presenting cells. J Biol Chem 2012; 287:38946-55. [PMID: 23019323 DOI: 10.1074/jbc.m112.380121] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The C-type lectin DC-SIGN expressed on dendritic cells (DCs) facilitates capture and internalization of a plethora of different pathogens. Although it is known that DC-SIGN organizes in nanoclusters at the surface of DCs, the molecular mechanisms responsible for this well defined nanopatterning and role in viral binding remain enigmatic. By combining biochemical and advanced biophysical techniques, including optical superresolution and single particle tracking, we demonstrate that DC-SIGN intrinsic nanoclustering strictly depends on its molecular structure. DC-SIGN nanoclusters exhibited free, Brownian diffusion on the cell membrane. Truncation of the extracellular neck region, known to abrogate tetramerization, significantly reduced nanoclustering and concomitantly increased lateral diffusion. Importantly, DC-SIGN nanocluster dissolution exclusively compromised binding to nanoscale size pathogens. Monte Carlo simulations revealed that heterogeneity on nanocluster density and spatial distribution confers broader binding capabilities to DC-SIGN. As such, our results underscore a direct relationship between spatial nanopatterning, driven by intermolecular interactions between the neck regions, and receptor diffusion to provide DC-SIGN with the exquisite ability to dock pathogens at the virus length scale. Insight into how virus receptors are organized prior to virus binding and how they assemble into functional platforms for virus docking is helpful to develop novel strategies to prevent virus entry and infection.
Collapse
Affiliation(s)
- Carlo Manzo
- Institut de Ciencies Fotoniques, Mediterranean Technology Park, 08860 Castelldefels (Barcelona), Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Prost LR, Grim JC, Tonelli M, Kiessling LL. Noncarbohydrate glycomimetics and glycoprotein surrogates as DC-SIGN antagonists and agonists. ACS Chem Biol 2012; 7:1603-8. [PMID: 22747463 DOI: 10.1021/cb300260p] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An understanding of the biological roles of lectins will be advanced by ligands that can inhibit or even recruit lectin function. To this end, glycomimetics, noncarbohydrate ligands that function analogously to endogenous carbohydrates, are being sought. The advantage of having such ligands is illustrated by the many roles of the protein DC-SIGN. DC-SIGN is a C-type lectin displayed on dendritic cells, where it binds to mannosides and fucosides to mediate interactions with other host cells or bacterial or viral pathogens. DC-SIGN engagement can modulate host immune responses (e.g., suppress autoimmunity) or benefit pathogens (e.g., promote HIV dissemination). DC-SIGN can bind to glycoconjugates, internalize glycosylated cargo for antigen processing, and transduce signals. DC-SIGN ligands can serve as inhibitors as well as probes of the lectin's function, so they are especially valuable for elucidating and controlling DC-SIGN's roles in immunity. We previously reported a small molecule that embodies key features of the carbohydrates that bind DC-SIGN. Here, we demonstrate that this noncarbohydrate ligand acts as a true glycomimetic. Using NMR HSQC experiments, we found that the compound mimics saccharide ligands: It occupies the same carbohydrate-binding site and interacts with the same amino acid residues on DC-SIGN. The glycomimetic also is functional. It had been shown previously to antagonize DC-SIGN function, but here we use it to generate DC-SIGN agonists. Specifically, appending this glycomimetic to a protein scaffold affords a conjugate that elicits key cellular signaling responses. Thus, the glycomimetic can give rise to functional glycoprotein surrogates that elicit lectin-mediated signaling.
Collapse
Affiliation(s)
- Lynne R. Prost
- Departments of †Biochemistry and ‡Chemistry, University of Wisconsin−Madison, Madison, Wisconsin
53706, United States
| | - Joseph C. Grim
- Departments of †Biochemistry and ‡Chemistry, University of Wisconsin−Madison, Madison, Wisconsin
53706, United States
| | - Marco Tonelli
- Departments of †Biochemistry and ‡Chemistry, University of Wisconsin−Madison, Madison, Wisconsin
53706, United States
| | - Laura L. Kiessling
- Departments of †Biochemistry and ‡Chemistry, University of Wisconsin−Madison, Madison, Wisconsin
53706, United States
| |
Collapse
|
169
|
Dehuyser L, Schaeffer E, Chaloin O, Mueller CG, Baati R, Wagner A. Synthesis of Novel Mannoside Glycolipid Conjugates for Inhibition of HIV-1 Trans-Infection. Bioconjug Chem 2012; 23:1731-9. [DOI: 10.1021/bc200644d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Laure Dehuyser
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| | - Evelyne Schaeffer
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Olivier Chaloin
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Christopher G. Mueller
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Rachid Baati
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| | - Alain Wagner
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| |
Collapse
|
170
|
Kawauchi Y, Kuroda Y, Kojima N. Preferences for uptake of carbohydrate-coated liposomes by C-type lectin receptors as antigen-uptake receptors. Glycoconj J 2012; 29:481-90. [DOI: 10.1007/s10719-012-9406-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/24/2012] [Accepted: 05/25/2012] [Indexed: 12/14/2022]
|
171
|
Interaction of Helicobacter pylori with C-type lectin dendritic cell-specific ICAM grabbing nonintegrin. J Biomed Biotechnol 2012; 2012:206463. [PMID: 22550396 PMCID: PMC3328334 DOI: 10.1155/2012/206463] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 01/23/2012] [Accepted: 01/26/2012] [Indexed: 02/07/2023] Open
Abstract
In this study we asked whether Helicobacter pylori whole cells and lipopolysaccharide (LPS) utilize sugar moieties of Lewis (Le) antigenic determinants to interact with DC-SIGN (dendritic cell specific ICAM grabbing nonintegrin) receptor on dendritic cells (DCs). For this purpose the soluble DC-SIGN/Fc adhesion assay and the THP-1 leukemia cells with induced expression of DC-SIGN were used. We showed that the binding specificity of DC-SIGN with H. pylori Le(X/Y) positive whole cells and H. pylori LPS of Le(X/Y) type was fucose dependent, whereas in Le(XY) negative H. pylori strains and LPS preparations without Lewis determinants, this binding was galactose dependent. The binding of soluble synthetic Le(X) and Le(Y) to the DC-SIGN-like receptor on THP-1 cells was also observed. In conclusion, the Le(XY) dependent as well as independent binding of H. pylori whole cells and H. pylori LPS to DC-SIGN was described. Moreover, we demonstrated that THP-1 cells may serve as an in vitro model for the assessment of H. pylori-DC-SIGN interactions mediated by Le(X) and Le(Y) determinants.
Collapse
|
172
|
Itano MS, Steinhauer C, Schmied JJ, Forthmann C, Liu P, Neumann AK, Thompson NL, Tinnefeld P, Jacobson K. Super-resolution imaging of C-type lectin and influenza hemagglutinin nanodomains on plasma membranes using blink microscopy. Biophys J 2012; 102:1534-42. [PMID: 22500753 DOI: 10.1016/j.bpj.2012.02.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 01/30/2012] [Accepted: 02/13/2012] [Indexed: 11/28/2022] Open
Abstract
Dendritic cells express DC-SIGN, a C-type lectin (CTL) that binds a variety of pathogens and facilitates their uptake for subsequent antigen presentation. DC-SIGN forms remarkably stable microdomains on the plasma membrane. However, inner leaflet lipid markers are able to diffuse through these microdomains suggesting that, rather than being densely packed with DC-SIGN proteins, an elemental substructure exists. Therefore, a super-resolution imaging technique, Blink Microscopy (Blink), was applied to further investigate the lateral distribution of DC-SIGN. Blink indicates that DC-SIGN, another CTL (CD206), and influenza hemagglutinin (HA) are all localized in small (∼80 nm in diameter) nanodomains. DC-SIGN and CD206 nanodomains are randomly distributed on the plasma membrane, whereas HA nanodomains cluster on length scales up to several microns. We estimate, as a lower limit, that DC-SIGN and HA nanodomains contain on average two tetramers or two trimers, respectively, whereas CD206 is often nonoligomerized. Two-color Blink determined that different CTLs rarely occupy the same nanodomain, although they appear colocalized using wide-field microscopy. What to our knowledge is a novel domain structure emerges in which elemental nanodomains, potentially capable of binding viruses, are organized in a random fashion; evidently, these nanodomains can be clustered into larger microdomains that act as receptor platforms for larger pathogens like yeasts.
Collapse
Affiliation(s)
- Michelle S Itano
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Dengue virus entry as target for antiviral therapy. J Trop Med 2012; 2012:628475. [PMID: 22529868 PMCID: PMC3317058 DOI: 10.1155/2012/628475] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 11/10/2011] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV) infections are expanding worldwide and, because of the lack of a vaccine, the search for antiviral products is imperative. Four serotypes of DENV are described and they all cause a similar disease outcome. It would be interesting to develop an antiviral product that can interact with all four serotypes, prevent host cell infection and subsequent immune activation. DENV entry is thus an interesting target for antiviral therapy. DENV enters the host cell through receptor-mediated endocytosis. Several cellular receptors have been proposed, and DC-SIGN, present on dendritic cells, is considered as the most important DENV receptor until now. Because DENV entry is a target for antiviral therapy, various classes of compounds have been investigated to inhibit this process. In this paper, an overview is given of all the putative DENV receptors, and the most promising DENV entry inhibitors are discussed.
Collapse
|
174
|
Specific glycan elements determine differential binding of individual egg glycoproteins of the human parasite Schistosoma mansoni by host C-type lectin receptors. Int J Parasitol 2012; 42:269-77. [DOI: 10.1016/j.ijpara.2012.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/19/2012] [Accepted: 01/19/2012] [Indexed: 11/23/2022]
|
175
|
Measles virus glycoprotein-pseudotyped lentiviral vectors are highly superior to vesicular stomatitis virus G pseudotypes for genetic modification of monocyte-derived dendritic cells. J Virol 2012; 86:5192-203. [PMID: 22345444 DOI: 10.1128/jvi.06283-11] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells capable of promoting or regulating innate and adaptive immune responses against non-self antigens. To better understand the DC biology or to use them for immune intervention, a tremendous effort has been made to improve gene transfer in these cells. Lentiviral vectors (LVs) have conferred a huge advantage in that they can transduce nondividing cells such as human monocyte-derived DCs (MDDCs) but required high amounts of viral particles and/or accessory proteins such as Vpx or Vpr to achieve sufficient transduction rates. As a consequence, these LVs have been shown to cause dramatic functional modifications, such as the activation or maturation of transduced MDDCs. Taking advantage of new pseudotyped LVs, i.e., with envelope glycoproteins from the measles virus (MV), we demonstrate that MDDCs are transduced very efficiently with these new LVs compared to the classically used vesicular stomatitis virus G-pseudotyped LVs and thus allowed to achieve high transduction rates at relatively low multiplicities of infection. Moreover, in this experimental setting, no activation or maturation markers were upregulated, while MV-LV-transduced cells remained able to mature after an appropriate Toll-like receptor stimulation. We then demonstrate that our MV-pseudotyped LVs use DC-SIGN, CD46, and CD150/SLAM as receptors to transduce MDDCs. Altogether, our results show that MV-pseudotyped LVs provide the most accurate and simple viral method for efficiently transferring genes into MDDCs without affecting their activation and/or maturation status.
Collapse
|
176
|
Unger WWJ, van Beelen AJ, Bruijns SC, Joshi M, Fehres CM, van Bloois L, Verstege MI, Ambrosini M, Kalay H, Nazmi K, Bolscher JG, Hooijberg E, de Gruijl TD, Storm G, van Kooyk Y. Glycan-modified liposomes boost CD4+ and CD8+ T-cell responses by targeting DC-SIGN on dendritic cells. J Control Release 2012; 160:88-95. [PMID: 22366522 DOI: 10.1016/j.jconrel.2012.02.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/07/2012] [Accepted: 02/09/2012] [Indexed: 01/23/2023]
Abstract
Cancer immunotherapy requires potent tumor-specific CD8(+) and CD4(+) T-cell responses, initiated by dendritic cells (DCs). Tumor antigens can be specifically targeted to DCs in vivo by exploiting their expression of C-type lectin receptors (CLR), which bind carbohydrate structures on antigens, resulting in internalization and antigen presentation to T-cells. We explored the potential of glycan-modified liposomes to target antigens to DCs to boost murine and human T-cell responses. Since DC-SIGN is a CLR expressed on DCs, liposomes were modified with DC-SIGN-binding glycans Lewis (Le)(B) or Le(X). Glycan modification of liposomes resulted in increased binding and internalization by BMDCs expressing human DC-SIGN. In the presence of LPS, this led to 100-fold more efficient presentation of the encapsulated antigens to CD4(+) and CD8(+) T-cells compared to unmodified liposomes or soluble antigen. Similarly, incubation of human moDC with melanoma antigen MART-1-encapsulated liposomes coated with Le(X) in the presence of LPS led to enhanced antigen-presentation to MART-1-specific CD8(+) T-cell clones. Moreover, this formulation drove primary CD8(+) T-cells to differentiate into high numbers of tetramer-specific, IFN-γ-producing effector T-cells. Together, our data demonstrate the potency of a glycoliposome-based vaccine targeting DC-SIGN for CD4(+) and CD8(+) effector T-cell activation. This approach may offer improved options for treatment of cancer patients and opens the way to in situ DC-targeted vaccination.
Collapse
Affiliation(s)
- Wendy W J Unger
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Difference in fine specificity to polysaccharides of Candida albicans mannoprotein between mouse SIGNR1 and human DC-SIGN. Infect Immun 2012; 80:1699-706. [PMID: 22331432 DOI: 10.1128/iai.06308-11] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
C-type lectin SIGNR1 directly recognizes Candida albicans and zymosan and has been considered to share properties of polysaccharide recognition with human DC-SIGN (hDC-SIGN). However, the precise specificity of SIGNR1 and the difference from that of hDC-SIGN remain to be elucidated. We prepared soluble forms of SIGNR1 and hDC-SIGN and conducted experiments to examine their respective specificities. Soluble SIGNR1 (sSIGNR1) bound several types of live C. albicans clinical isolate strains in an EDTA-sensitive manner. Inhibition analyses of sSIGNR1 binding by glycans from various yeast strains demonstrated that SIGNR1 preferentially recognizes N-glycan α-mannose side chains in Candida mannoproteins, as reported in hDC-SIGN. Unlike shDC-SIGN, however, sSIGNR1 recognized not only Saccharomyces cerevisiae, but also C. albicans J-1012 glycan, even after α-mannosidase treatment that leaves only β1,2-mannose-capped α-mannose side chains. In addition, glycomicroarray analyses showed that sSIGNR1 binds mannans from C. albicans and S. cerevisiae but does not recognize Lewis(a/b/x/y) antigen polysaccharides as in shDC-SIGN. Consistent with these results, RAW264.7 cells expressing hDC-SIGN in which the carbohydrate recognition domain (CRD) was replaced with that of SIGNR1 (RAW-chimera) produced comparable amounts of interleukin 10 (IL-10) in response to glycans from C. albicans and S. cerevisiae, but those expressing hDC-SIGN produced less IL-10 in response to S. cerevisiae than C. albicans. Furthermore, RAW-hDC-SIGN cells remarkably reduced IL-10 production after α-mannosidase treatment compared with RAW-chimera cells. These results indicate that SIGNR1 recognizes C. albicans/yeast through a specificity partly distinct from that of its homologue hDC-SIGN.
Collapse
|
178
|
Soybean agglutinin coated PLA particles entrapping candidate vaccines induces enhanced primary and sustained secondary antibody response from single point immunization. Eur J Pharm Sci 2012; 45:282-95. [DOI: 10.1016/j.ejps.2011.11.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/31/2011] [Accepted: 11/28/2011] [Indexed: 11/22/2022]
|
179
|
Ahmed Z, Czubala M, Blanchet F, Piguet V. HIV impairment of immune responses in dendritic cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 762:201-38. [PMID: 22975877 DOI: 10.1007/978-1-4614-4433-6_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Dendritic cells and their subsets are diverse populations of immune cells in the skin and mucous membranes that possess the ability to sense the presence of microbes and orchestrate an efficient and adapted immune response. Dendritic cells (DC) have the unique ability to act as a bridge between the innate and adaptive immune responses. These cells are composed of a number of subsets behaving with preferential and specific features depending on their location and surrounding environment. Langerhans cells (LC) or dermal DC (dDC) are readily present in mucosal areas. Other DC subsets such as plasmacytoid DC (pDC), myeloid DC (myDC), or monocyte-derived DC (MDDC) are thought to be recruited or differentiated in sites of pathogenic challenge. Upon HIV infection, DC and their subsets are likely among the very first immune cells to encounter incoming pathogens and initiate innate and adaptive immune responses. However, as evidenced during HIV infection, some pathogens have evolved subtle strategies to hijack key cellular machineries essential to generate efficient antiviral responses and subvert immune responses for spread and survival.In this chapter, we review recent research aimed at investigating the involvement of DC subtypes in HIV transmission at mucosal sites, concentrating on HIV impact on cellular signalling and trafficking pathways in DC leading to DC-mediated immune response alterations and viral immune evasion. We also address some aspects of DC functions during the chronic immune pathogenesis and conclude with an overview of the current and novel therapeutic and prophylactic strategies aimed at improving DC-mediated immune responses, thus to potentially tackle the early events of mucosal HIV infection and spread.
Collapse
Affiliation(s)
- Zahra Ahmed
- Department of Dermatology and Wound Healing, Cardiff University School of Medicine, Cardiff, Wales, UK
| | | | | | | |
Collapse
|
180
|
Abstract
In the immune system, C-type lectins and CTLDs have been shown to act both as adhesion and as pathogen recognition receptors. The Dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) and its homologs in human and mouse represent an important C-type lectin family. DC-SIGN contains a lectin domain that recognizes in a Ca2+-dependent manner carbohydrates such as mannose-containing structures present on glycoproteins such as ICAM-2 and ICAM-3. DC-SIGN is a prototype C-type lectin organized in microdomains, which have their role as pathogen recognition receptors in sensing microbes. Although the integrin LFA-1 is a counter-receptor for both ICAM-2 and ICAM-3 on DC, DC-SIGN is the high affinity adhesion receptor for ICAM-2/-3. While cell–cell contact is a primary function of selectins, collectins are specialized in recognition of pathogens. Interestingly, DC-SIGN is a cell adhesion receptor as well as a pathogen recognition receptor. As adhesion receptor, DC-SIGN mediates the contact between dendritic cells (DCs) and T lymphocytes, by binding to ICAM-3, and mediates rolling of DCs on endothelium, by interacting with ICAM-2. As pathogen receptor, DC-SIGN recognizes a variety of microorganisms, including viruses, bacteria, fungi and several parasites (Cambi et al. 2005). The natural ligands of DC-SIGN consist of mannose oligosaccharides or fucose-containing Lewis-type determinants. In this chapter, we shall focus on the structure and functions of DC-SIGN and related CTLDs in the recognition of pathogens, the molecular and structural determinants that regulate the interaction with pathogen-associated molecular patterns. The heterogeneity of carbohydrate residues exposed on cellular proteins and pathogens regulates specific binding of DC-expressed C-type lectins that contribute to the diversity of immune responses created by DCs (van Kooyk et al. 2003a; Cambi et al. 2005).
Collapse
|
181
|
Clark GF, Grassi P, Pang PC, Panico M, Lafrenz D, Drobnis EZ, Baldwin MR, Morris HR, Haslam SM, Schedin-Weiss S, Sun W, Dell A. Tumor biomarker glycoproteins in the seminal plasma of healthy human males are endogenous ligands for DC-SIGN. Mol Cell Proteomics 2012; 11:M111.008730. [PMID: 21986992 PMCID: PMC3270097 DOI: 10.1074/mcp.m111.008730] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 09/06/2011] [Indexed: 01/15/2023] Open
Abstract
DC-SIGN is an immune C-type lectin that is expressed on both immature and mature dendritic cells associated with peripheral and lymphoid tissues in humans. It is a pattern recognition receptor that binds to several pathogens including HIV-1, Ebola virus, Mycobacterium tuberculosis, Candida albicans, Helicobacter pylori, and Schistosoma mansoni. Evidence is now mounting that DC-SIGN also recognizes endogenous glycoproteins, and that such interactions play a major role in maintaining immune homeostasis in humans and mice. Autoantigens (neoantigens) are produced for the first time in the human testes and other organs of the male urogenital tract under androgenic stimulus during puberty. Such antigens trigger autoimmune orchitis if the immune response is not tightly regulated within this system. Endogenous ligands for DC-SIGN could play a role in modulating such responses. Human seminal plasma glycoproteins express a high level of terminal Lewis(x) and Lewis(y) carbohydrate antigens. These epitopes react specifically with the lectin domains of DC-SIGN. However, because the expression of these sequences is necessary but not sufficient for interaction with DC-SIGN, this study was undertaken to determine if any seminal plasma glycoproteins are also endogenous ligands for DC-SIGN. Glycoproteins bearing terminal Lewis(x) and Lewis(y) sequences were initially isolated by lectin affinity chromatography. Protein sequencing established that three tumor biomarker glycoproteins (clusterin, galectin-3 binding glycoprotein, prostatic acid phosphatase) and protein C inhibitor were purified by using this affinity method. The binding of DC-SIGN to these seminal plasma glycoproteins was demonstrated in both Western blot and immunoprecipitation studies. These findings have confirmed that human seminal plasma contains endogenous glycoprotein ligands for DC-SIGN that could play a role in maintaining immune homeostasis both in the male urogenital tract and the vagina after coitus.
Collapse
Affiliation(s)
- Gary F. Clark
- From the ‡Division of Reproductive and Perinatal Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211
| | - Paola Grassi
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| | - Poh-Choo Pang
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| | - Maria Panico
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| | - David Lafrenz
- From the ‡Division of Reproductive and Perinatal Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211
| | - Erma Z. Drobnis
- ¶Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211
| | - Michael R. Baldwin
- ‖Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri 65211
| | - Howard R. Morris
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| | - Stuart M. Haslam
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| | - Sophia Schedin-Weiss
- **Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Wei Sun
- **Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anne Dell
- §Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London, SW7 2AZ, United Kingdom
| |
Collapse
|
182
|
Uriel C, Gómez AM, López JC, Fraser-Reid B. Ready access to a branched Man5 oligosaccharide based on regioselective glycosylations of a mannose-tetraol with n-pentenyl orthoesters. Org Biomol Chem 2012; 10:8361-70. [DOI: 10.1039/c2ob26432c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
183
|
Hijazi K, Wang Y, Scala C, Jeffs S, Longstaff C, Stieh D, Haggarty B, Vanham G, Schols D, Balzarini J, Jones IM, Hoxie J, Shattock R, Kelly CG. DC-SIGN increases the affinity of HIV-1 envelope glycoprotein interaction with CD4. PLoS One 2011; 6:e28307. [PMID: 22163292 PMCID: PMC3233575 DOI: 10.1371/journal.pone.0028307] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 11/05/2011] [Indexed: 11/30/2022] Open
Abstract
Mannose-binding C-type lectin receptors, expressed on Langerhans cells and subepithelial dendritic cells (DCs) of cervico-vaginal tissues, play an important role in HIV-1 capture and subsequent dissemination to lymph nodes. DC-SIGN has been implicated in both productive infection of DCs and the DC-mediated trans infection of CD4+ T cells that occurs in the absence of replication. However, the molecular events that underlie this efficient transmission have not been fully defined. In this study, we have examined the effect of the extracellular domains of DC-SIGN and Langerin on the stability of the interaction of the HIV-1 envelope glycoprotein with CD4 and also on replication in permissive cells. Surface plasmon resonance analysis showed that DC-SIGN increases the binding affinity of trimeric gp140 envelope glycoproteins to CD4. In contrast, Langerin had no effect on the stability of the gp140:CD4 complex. In vitro infection experiments to compare DC-SIGN enhancement of CD4-dependent and CD4-independent strains demonstrated significantly lower enhancement of the CD4-independent strain. In addition DC-SIGN increased the relative rate of infection of the CD4-dependent strain but had no effect on the CD4-independent strain. DC-SIGN binding to the HIV envelope protein effectively increases exposure of the CD4 binding site, which in turn contributes to enhancement of infection.
Collapse
Affiliation(s)
- Karolin Hijazi
- King's College London, Dental Institute, Oral Immunology, Tower Wing, Guy's Hospital, London, United Kingdom
| | - Yufei Wang
- King's College London, Dental Institute, Oral Immunology, Tower Wing, Guy's Hospital, London, United Kingdom
| | - Carlo Scala
- King's College London, Dental Institute, Oral Immunology, Tower Wing, Guy's Hospital, London, United Kingdom
| | - Simon Jeffs
- Jefferiss Trust Research Laboratories, Wright-Fleming Institute, Division of Medicine, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Colin Longstaff
- Biotherapeutics Section, National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom
| | - Daniel Stieh
- Centre for Infection, Department of Cellular and Molecular Medicine, St George's, University of London, London, United Kingdom
| | - Beth Haggarty
- Penn Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Guido Vanham
- Virology Unit, Division of Microbiology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Faculty of Medicine and Pharmacy Free University of Brussels, Brussels, Belgium
| | - Dominique Schols
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jan Balzarini
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - James Hoxie
- Penn Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robin Shattock
- Centre for Infection, Department of Cellular and Molecular Medicine, St George's, University of London, London, United Kingdom
| | - Charles G. Kelly
- King's College London, Dental Institute, Oral Immunology, Tower Wing, Guy's Hospital, London, United Kingdom
- * E-mail:
| |
Collapse
|
184
|
Londrigan SL, Tate MD, Brooks AG, Reading PC. Cell-surface receptors on macrophages and dendritic cells for attachment and entry of influenza virus. J Leukoc Biol 2011; 92:97-106. [PMID: 22124137 PMCID: PMC7166464 DOI: 10.1189/jlb.1011492] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Review of interactions between influenza A virus and C‐type lectin receptors on macrophages and dendritic cells that may result in virus entry and infection. Airway MΦ and DCs are important components of innate host defense and can play a critical role in limiting the severity of influenza virus infection. Although it has been well established that cell‐surface SA acts as a primary attachment receptor for IAV, the particular receptor(s) or coreceptor(s) that mediate IAV entry into any cell, including MΦ and DC, have not been clearly defined. Identifying which receptors are involved in attachment and entry of IAV into immune cells may have important implications in regard to understanding IAV tropism and pathogenesis. Recent evidence suggests that specialized receptors on MΦ and DCs, namely CLRs, can act as capture and/or entry receptors for many viral pathogens, including IAV. Herein, we review the early stages of infection of MΦ and DC by IAV. Specifically, we examine the potential role of CLRs expressed on MΦ and DC to act as attachment and/or entry receptors for IAV.
Collapse
Affiliation(s)
- Sarah L Londrigan
- The Department of Microbiology and Immunology, The University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
185
|
Thomas RJ. Receptor mimicry as novel therapeutic treatment for biothreat agents. Bioeng Bugs 2011; 1:17-30. [PMID: 21327124 DOI: 10.4161/bbug.1.1.10049] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 09/10/2009] [Accepted: 09/11/2009] [Indexed: 12/20/2022] Open
Abstract
The specter of intentional release of pathogenic microbes and their toxins is a real threat. This article reviews the literature on adhesins of biothreat agents, their interactions with oligosaccharides and the potential for anti-adhesion compounds as an alternative to conventional therapeutics. The minimal binding structure of ricin has been well characterised and offers the best candidate for successful anti-adhesion therapy based on the Galβ1-4GlcNAc structure. The botulinum toxin serotypes A-F bind to a low number of gangliosides (GT1b, GQ1b, GD1a and GD1b) hence it should be possible to determine the minimal structure for binding. The minimal disaccharide sequence of GalNAcβ1-4Gal found in the gangliosides asialo-GM1 and asialo-GM2 is required for adhesion for many respiratory pathogens. Although a number of adhesins have been identified in bacterial biothreat agents such as Yersinia pestis, Bacillus anthracis, Francisella tularensis, Brucella species and Burkholderia pseudomallei, specific information regarding their in vivo expression during pneumonic infection is lacking. Limited oligosaccharide inhibition studies indicate the potential of GalNAcβ1-4Gal, GalNAcβ-3Gal and the hydrophobic compound, para-nitrophenol as starting points for the rational design of generic anti-adhesion compounds. A cocktail of multivalent oligosaccharides based on the minimal binding structures of identified adhesins would offer the best candidates for anti-adhesion therapy.
Collapse
|
186
|
Joshi MD, Unger WW, van Beelen AJ, Bruijns SC, Litjens M, van Bloois L, Kalay H, van Kooyk Y, Storm G. DC-SIGN mediated antigen-targeting using glycan-modified liposomes: Formulation considerations. Int J Pharm 2011; 416:426-32. [DOI: 10.1016/j.ijpharm.2011.02.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/19/2011] [Accepted: 02/21/2011] [Indexed: 10/18/2022]
|
187
|
Ciobanu M, Huang KT, Daguer JP, Barluenga S, Chaloin O, Schaeffer E, Mueller CG, Mitchell DA, Winssinger N. Selection of a synthetic glycan oligomer from a library of DNA-templated fragments against DC-SIGN and inhibition of HIV gp120 binding to dendritic cells. Chem Commun (Camb) 2011; 47:9321-3. [PMID: 21785785 DOI: 10.1039/c1cc13213j] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We report the synthesis of a nucleic acid-encoded carbohydrate library, its combinatorial self-assembly into 37,485 pairs and a screen against DC-SIGN leading to the identification of consensus ligand motifs. A prototypical example from the selected pairs was shown to have enhanced binding. A dendrimer incorporating the selected motifs inhibited gp120's binding to dendritic cells with higher efficiency than mannan.
Collapse
Affiliation(s)
- Mihai Ciobanu
- Institut de Science et Ingenierie Supramoleculaires (ISIS), Université de Strasbourg-CNRS, 8 allee Gaspard Monge, 67000 Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
188
|
HIV microbicides: state-of-the-art and new perspectives on the development of entry inhibitors. Future Med Chem 2011; 2:1141-59. [PMID: 21426161 DOI: 10.4155/fmc.10.203] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Since the discovery of HIV at the beginning of the 1980s, numerous efforts have been devoted to the search of an efficient vaccine. There are at least 25 drugs available for HIV treatment, but no cure is available. The observation that therapy for HIV disease is life long and that these drugs are associated with a number of side effects underlines the need for approaches aimed at preventing rather than treating infection. Additionally, the economic burden of treatment for the HIV infection occupies an increasing share of healthcare expenditure, making current practices likely to become difficult to sustain in the long run. Unfortunately, no effective vaccine for this disease is foreseeable in the near future. Microbicides could be an alternate way to build preventative approaches to HIV infection. Strategies based on preventing the virus from reaching its target cells seem to have some room for development and application. In this review we explore the state-of-the-art of available microbicides, focusing on HIV entry inhibitors. In addition, we discuss new compounds that show anti-HIV activity, which could be effective candidates.
Collapse
|
189
|
Virus-receptor mediated transduction of dendritic cells by lentiviruses enveloped with glycoproteins derived from Semliki Forest virus. PLoS One 2011; 6:e21491. [PMID: 21738680 PMCID: PMC3124512 DOI: 10.1371/journal.pone.0021491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 05/30/2011] [Indexed: 11/14/2022] Open
Abstract
Lentiviruses have recently attracted considerable interest for their potential as a genetic modification tool for dendritic cells (DCs). In this study, we explore the ability of lentiviruses enveloped with alphaviral envelope glycoproteins derived from Semliki Forest virus (SFV) to mediate transduction of DCs. We found that SFV glycoprotein (SFV-G)-pseudotyped lentiviruses use C-type lectins (DC-SIGN and L-SIGN) as attachment factors for transduction of DCs. Importantly, SFV-G pseudotypes appear to have enhanced transduction towards C-type lectin-expressing cells when produced under conditions limiting glycosylation to simple high-mannose, N-linked glycans. These results, in addition to the natural DC tropism of SFV-G, offer evidence to support the use of SFV-G-bearing lentiviruses to genetically modify DCs for the study of DC biology and DC-based immunotherapy.
Collapse
|
190
|
Luczkowiak J, Sattin S, Sutkevičiu̅tė I, Reina JJ, Sánchez-Navarro M, Thépaut M, Martínez-Prats L, Daghetti A, Fieschi F, Delgado R, Bernardi A, Rojo J. Pseudosaccharide Functionalized Dendrimers as Potent Inhibitors of DC-SIGN Dependent Ebola Pseudotyped Viral Infection. Bioconjug Chem 2011; 22:1354-65. [DOI: 10.1021/bc2000403] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Joanna Luczkowiak
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Sara Sattin
- Università degli Studi di Milano, Dipartimento di Chimica Organica e Industriale, and CISI, Milano, Italy
| | - Ieva Sutkevičiu̅tė
- Institut de Biologie Structurale, CNRS, UMR 5075, 41 rue Jules Horowitz, 38027 Grenoble France
- Université Joseph Fourier, Institut Universitaire de France, 38000 Grenoble, France
| | - José Juan Reina
- Università degli Studi di Milano, Dipartimento di Chimica Organica e Industriale, and CISI, Milano, Italy
| | - Macarena Sánchez-Navarro
- Glycosystems Laboratory, Instituto de Investigaciones Químicas, CSIC − Universidad de Sevilla, Américo Vespucio 49, 41092 Seville, Spain
| | - Michel Thépaut
- Institut de Biologie Structurale, CNRS, UMR 5075, 41 rue Jules Horowitz, 38027 Grenoble France
- CEA, DSV, 38027 Grenoble France
| | - Lorena Martínez-Prats
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Anna Daghetti
- Università degli Studi di Milano, Dipartimento di Chimica Organica e Industriale, and CISI, Milano, Italy
| | - Franck Fieschi
- Institut de Biologie Structurale, CNRS, UMR 5075, 41 rue Jules Horowitz, 38027 Grenoble France
- Université Joseph Fourier, Institut Universitaire de France, 38000 Grenoble, France
| | - Rafael Delgado
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Anna Bernardi
- Università degli Studi di Milano, Dipartimento di Chimica Organica e Industriale, and CISI, Milano, Italy
- CNR-ISTM, Institute of Molecular Sciences and Technologies, Milano, Italy
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas, CSIC − Universidad de Sevilla, Américo Vespucio 49, 41092 Seville, Spain
| |
Collapse
|
191
|
Leckband DE, Menon S, Rosenberg K, Graham SA, Taylor ME, Drickamer K. Geometry and adhesion of extracellular domains of DC-SIGNR neck length variants analyzed by force-distance measurements. Biochemistry 2011; 50:6125-32. [PMID: 21650186 PMCID: PMC3140775 DOI: 10.1021/bi2003444] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
![]()
Force–distance measurements have been used to examine differences in the interaction of the dendritic cell glycan-binding receptor DC-SIGN and the closely related endothelial cell receptor DC-SIGNR (L-SIGN) with membranes bearing glycan ligands. The results demonstrate that upon binding to membrane-anchored ligand, DC-SIGNR undergoes a conformational change similar to that previously observed for DC-SIGN. The results also validate a model for the extracellular domain of DC-SIGNR derived from crystallographic studies. Force measurements were performed with DC-SIGNR variants that differ in the length of the neck that result from genetic polymorphisms, which encode different numbers of the 23-amino acid repeat sequences that constitute the neck. The findings are consistent with an elongated, relatively rigid structure of the neck repeat observed in crystals. In addition, differences in the lengths of DC-SIGN and DC-SIGNR extracellular domains with equivalent numbers of neck repeats support a model in which the different dispositions of the carbohydrate-recognition domains in DC-SIGN and DC-SIGNR result from variations in the sequences of the necks.
Collapse
Affiliation(s)
- Deborah E Leckband
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | | | | | | | | | | |
Collapse
|
192
|
Froelich S, Tai A, Kennedy K, Zubair A, Wang P. Pseudotyping lentiviral vectors with aura virus envelope glycoproteins for DC-SIGN-mediated transduction of dendritic cells. Hum Gene Ther 2011; 22:1281-91. [PMID: 21452926 DOI: 10.1089/hum.2010.196] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Lentiviral vectors (LVs) pseudotyped with envelope proteins of alphaviruses have recently attracted considerable interest for their potential as gene delivery tools. We report the production of human immunodeficiency virus type 1 (HIV-1)-derived LVs pseudotyped with envelope glycoproteins derived from the Aura virus (AURA). We found that the AURA-glycoprotein-pseudotyped LVs use C-type lectins (DC-SIGN and L-SIGN) as attachment factors. These interactions with DC-SIGN are specific as determined by inhibition assays and appear to facilitate transduction through a pH-dependent pathway. AURA-pseudotyped LVs were used to transduce monocyte-derived dendritic cells (DCs) and the transduction was shown to be DC-SIGN mediated, as illustrated by competitive inhibition with DC-SIGN and L-SIGN antibodies and yeast mannan. Comparisons with LVs enveloped with glycoproteins derived from vesicular stomatitis virus and Sindbis virus suggest that AURA-glycoprotein-bearing LVs might be useful to genetically modify DCs for the study of DC biology and DC-based immunotherapy.
Collapse
Affiliation(s)
- Steven Froelich
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | |
Collapse
|
193
|
Meevissen MHJ, Yazdanbakhsh M, Hokke CH. Schistosoma mansoni egg glycoproteins and C-type lectins of host immune cells: molecular partners that shape immune responses. Exp Parasitol 2011; 132:14-21. [PMID: 21616068 DOI: 10.1016/j.exppara.2011.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 04/22/2011] [Accepted: 05/10/2011] [Indexed: 01/13/2023]
Abstract
Schistosome eggs and egg-derived molecules are potent immunomodulatory agents. There is increasing evidence that the interplay between egg glycoproteins and host C-type lectins plays an important role in shaping immune responses during schistosomiasis. As most experiments in this field so far have been performed using complex protein/glycoprotein mixtures or synthetic model glycoconjugates, it is still largely unclear which individual moieties of schistosome eggs are immunologically active. In this review we will discuss molecular aspects of Schistosoma mansoni egg glycoproteins, their interactions with C-type lectins, and the relevance to schistosome egg immunobiology.
Collapse
Affiliation(s)
- Moniek H J Meevissen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
194
|
Holla A, Skerra A. Comparative analysis reveals selective recognition of glycans by the dendritic cell receptors DC-SIGN and Langerin. Protein Eng Des Sel 2011; 24:659-69. [PMID: 21540232 DOI: 10.1093/protein/gzr016] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
DC-SIGN (dendritic cell-specific ICAM-3 grabbing non-integrin) and Langerin are homologous C-type lectins expressed as cell-surface receptors on different populations of dendritic cells (DCs). DC-SIGN interacts with glycan structures on HIV-1, facilitating virus survival, transmission and infection, whereas Langerin, which is characteristic of Langerhans cells (LCs), promotes HIV-1 uptake and degradation. Here we describe a comprehensive comparison of the glycan specificities of both proteins by probing a synthetic carbohydrate microarray comprising 275 sugar compounds using the bacterially produced and fluorescence-labeled, monomeric carbohydrate-recognition domains (CRDs) of DC-SIGN and Langerin. In this side-by-side study DC-SIGN was found to preferentially bind internal mannose residues of high-mannose-type saccharides and the fucose-containing blood-type antigens H, A, B, Le(a), Le(b) Le(x), Le(y), sialyl-Le(a) as well as sulfatated derivatives of Le(a) and Le(x). In contrast, Langerin appeared to recognize a different spectrum of compounds, especially those containing terminal mannose, terminal N-acetylglucosamine and 6-sulfogalactose residues, but also the blood-type antigens H, A and B. Of the Lewis antigens, only Le(b), Le(y), sialyl-Le(a) and the sialyl-Le(x) derivative with 6'-sulfatation at the galactose (sialyl-6SGal Le(x)) were weakly bound by Langerin. Notably, Ca(2+)-independent glycan-binding activity of Langerin could not be detected either by probing the glycan array or by isothermal titration calorimetry of the CRD with mannose and mannobiose. The precise knowledge of carbohydrate specificity of DC-SIGN and Langerin receptors resulting from our study may aid the future design of microbicides that specifically affect the DC-SIGN/HIV-1 interaction while not compromising the protective function of Langerin.
Collapse
Affiliation(s)
- Andrea Holla
- Munich Center for Integrated Protein Science, CIPS-M, Technische Universität München, Freising-Weihenstephan, Germany
| | | |
Collapse
|
195
|
Cui L, Cohen JA, Broaders KE, Beaudette TT, Fréchet JMJ. Mannosylated dextran nanoparticles: a pH-sensitive system engineered for immunomodulation through mannose targeting. Bioconjug Chem 2011; 22:949-57. [PMID: 21476603 DOI: 10.1021/bc100596w] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Biotherapeutic delivery is a rapidly growing field in need of new materials that are easy to modify, are biocompatible, and provide for triggered release of their encapsulated cargo. Herein, we report on a particulate system made of a polysaccharide-based pH-sensitive material that can be efficiently modified to display mannose-based ligands of cell-surface receptors. These ligands are beneficial for antigen delivery, as they enhance internalization and activation of APCs, and are thus capable of modulating immune responses. When compared to unmodified particles or particles modified with a nonspecific sugar residue used in the delivery of antigens to dendritic cells (DCs), the mannosylated particles exhibited enhanced antigen presentation in the context of major histocompatibility complex (MHC) class I molecules. This represents the first demonstration of a mannosylated particulate system that enables enhanced MHC I antigen presentation by DCs in vitro. Our readily functionalized pH-sensitive material may also open new avenues in the development of optimally modulated vaccine delivery systems.
Collapse
Affiliation(s)
- Lina Cui
- College of Chemistry, University of California-Berkeley, CA 94720-1460, United States
| | | | | | | | | |
Collapse
|
196
|
Gustafsson A, Sjöblom M, Strindelius L, Johansson T, Fleckenstein T, Chatzissavidou N, Lindberg L, Angström J, Rova U, Holgersson J. Pichia pastoris-produced mucin-type fusion proteins with multivalent O-glycan substitution as targeting molecules for mannose-specific receptors of the immune system. Glycobiology 2011; 21:1071-86. [PMID: 21474492 DOI: 10.1093/glycob/cwr046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mannose-binding proteins like the macrophage mannose receptor (MR), the dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) and mannose-binding lectin (MBL) play crucial roles in both innate and adaptive immune responses. Immunoglobulin fusion proteins of the P-selectin glycoprotein ligand-1 (PSGL-1/mIgG(2b)) carrying mostly O-glycans and, as a control, the α1-acid glycoprotein (AGP/mIgG(2b)) carrying mainly N-linked glycans were stably expressed in the yeast Pichia pastoris. Pichia pastoris-produced PSGL-1/mIgG(2b) was shown to carry O-glycans that mediated strong binding to mannose-specific lectins in a lectin array and were susceptible to cleavage by α-mannosidases including an α1,2- but not an α1,6-mannosidase. Electrospray ionization ion-trap mass spectrometry confirmed the presence of O-glycans containing up to nine hexoses with the penta- and hexasaccharides being the predominant ones. α1,2- and α1,3-linked, but not α1,6-linked, mannose residues were detected by (1)H-nuclear magnetic resonance spectroscopy confirming the results of the mannosidase cleavage. The apparent equilibrium dissociation constants for binding of PNGase F-treated mannosylated PSGL-1/mIgG(2b) to MR, DC-SIGN and MBL were shown by surface plasmon resonance to be 126, 56 and 16 nM, respectively. In conclusion, PSGL-1/mIgG(2b) expressed in P. pastoris carried O-glycans mainly comprised of α-linked mannoses and with up to nine residues. It bound mannose-specific receptors with high apparent affinity and may become a potent targeting molecule for these receptors in vivo.
Collapse
|
197
|
Dendritic cell-directed lentivector vaccine induces antigen-specific immune responses against murine melanoma. Cancer Gene Ther 2011; 18:370-80. [PMID: 21372855 DOI: 10.1038/cgt.2011.13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lentivectors are potential vaccine delivery vehicles because they can efficiently transduce a variety of non-dividing cells, including antigen-presenting cells, and do not cause expression of extra viral proteins. To improve safety while retaining efficiency, a dendritic cell (DC)-specific lentivector was constructed by pseudotyping the vector with an engineered viral glycoprotein derived from Sindbis virus. We assessed the level of anti-tumor immunity conferred by this engineered lentivector encoding the melanoma antigen gp100 in a mouse model. Footpad injection of the engineered lentivectors results in the best antigen-specific immune response as compared with subcutaneous and intraperitoneal injections. A single prime vaccination of the engineered lentivectors can elicit a high frequency (up to 10%) of gp100-specific CD8(+) T cells in peripheral blood 3 weeks after the vaccination and this response will be maintained at around 5% for up to 8 weeks. We found that these engineered lentivectors elicited relatively low levels of anti-vector neutralizing antibody responses. Importantly, direct injection of this engineered lentivector inhibited the growth of aggressive B16 murine melanoma. These data suggest that DC-specific lentivectors can be a novel and alternative vaccine carrier with the potential to deliver effective anti-tumor immunity for cancer immunotherapy.
Collapse
|
198
|
Tai A, Froelich S, Joo KI, Wang P. Production of lentiviral vectors with enhanced efficiency to target dendritic cells by attenuating mannosidase activity of mammalian cells. J Biol Eng 2011; 5:1. [PMID: 21276219 PMCID: PMC3039557 DOI: 10.1186/1754-1611-5-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 01/28/2011] [Indexed: 01/07/2023] Open
Abstract
Background Dendritic cells (DCs) are antigen-presenting immune cells that interact with T cells and have been widely studied for vaccine applications. To achieve this, DCs can be manipulated by lentiviral vectors (LVs) to express antigens to stimulate the desired antigen-specific T cell response, which gives this approach great potential to fight diseases such as cancers, HIV, and autoimmune diseases. Previously we showed that LVs enveloped with an engineered Sindbis virus glycoprotein (SVGmu) could target DCs through a specific interaction with DC-SIGN, a surface molecule predominantly expressed by DCs. We hypothesized that SVGmu interacts with DC-SIGN in a mannose-dependent manner, and that an increase in high-mannose structures on the glycoprotein surface could result in higher targeting efficiencies of LVs towards DCs. It is known that 1-deoxymannojirimycin (DMJ) can inhibit mannosidase, which is an enzyme that removes high-mannose structures during the glycosylation process. Thus, we investigated the possibility of generating LVs with enhanced capability to modify DCs by supplying DMJ during vector production. Results Through western blot analysis and binding tests, we were able to infer that binding of SVGmu to DC-SIGN is directly related to amount of high-mannose structures on SVGmu. We also found that the titer for the LV (FUGW/SVGmu) produced with DMJ against 293T.DCSIGN, a human cell line expressing the human DC-SIGN atnibody, was over four times higher than that of vector produced without DMJ. In addition, transduction of a human DC cell line, MUTZ-3, yielded a higher transduction efficiency for the LV produced with DMJ. Conclusion We conclude that LVs produced under conditions with inhibited mannosidase activity can effectively modify cells displaying the DC-specific marker DC-SIGN. This study offers evidence to support the utilization of DMJ in producing LVs that are enhanced carriers for the development of DC-directed vaccines.
Collapse
Affiliation(s)
- April Tai
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
199
|
Pipirou Z, Powlesland AS, Steffen I, Pöhlmann S, Taylor ME, Drickamer K. Mouse LSECtin as a model for a human Ebola virus receptor. Glycobiology 2011; 21:806-12. [PMID: 21257728 PMCID: PMC3091528 DOI: 10.1093/glycob/cwr008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The biochemical properties of mouse LSECtin, a glycan-binding receptor that is a member of the C-type lectin family found on sinusoidal endothelial cells, have been investigated. The C-type carbohydrate-recognition domain of mouse LSECtin, expressed in bacteria, has been used in solid-phase binding assays, and a tetramerized form has been used to probe a glycan array. In spite of sequence differences near the glycan-binding sites, the mouse receptor closely mimics the properties of the human receptor, showing high affinity binding to glycans bearing terminal GlcNAcβ1-2Man motifs. Site-directed mutagenesis has been used to confirm that residues near the binding site that differ between the human and the mouse proteins do not affect this binding specificity. Mouse and human LSECtin have been shown to bind Ebola virus glycoprotein with equivalent affinities, and the GlcNAcβ1-2Man disaccharide has been demonstrated to be an effective inhibitor of this interaction. These studies provide a basis for using mouse LSECtin, and knockout mice lacking this receptor, to model the biological properties of the human receptor.
Collapse
Affiliation(s)
- Zoi Pipirou
- Division of Molecular Biosciences, Department of Life Sciences, Imperial College, London, UK
| | | | | | | | | | | |
Collapse
|
200
|
Guzzi C, Angulo J, Doro F, Reina JJ, Thépaut M, Fieschi F, Bernardi A, Rojo J, Nieto PM. Insights into molecular recognition of LewisX mimics by DC-SIGN using NMR and molecular modelling. Org Biomol Chem 2011; 9:7705-12. [DOI: 10.1039/c1ob05938f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|