151
|
Lalor PF, Lai WK, Curbishley SM, Shetty S, Adams DH. Human hepatic sinusoidal endothelial cells can be distinguished by expression of phenotypic markers related to their specialised functions in vivo. World J Gastroenterol 2006; 12:5429-39. [PMID: 17006978 PMCID: PMC4088223 DOI: 10.3748/wjg.v12.i34.5429] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Revised: 05/28/2006] [Accepted: 06/14/2006] [Indexed: 02/06/2023] Open
Abstract
The hepatic sinusoids are lined by a unique population of hepatic sinusoidal endothelial cells (HSEC), which is one of the first hepatic cell populations to come into contact with blood components. However, HSEC are not simply barrier cells that restrict the access of blood-borne compounds to the parenchyma. They are functionally specialised endothelial cells that have complex roles, including not only receptor-mediated clearance of endotoxin, bacteria and other compounds, but also the regulation of inflammation, leukocyte recruitment and host immune responses to pathogens. Thus understanding the differentiation and function of HSEC is critical for the elucidation of liver biology and pathophysiology. This article reviews methods for isolating and studying human hepatic endothelial cell populations using in vitro models. We also discuss the expression and functions of phenotypic markers, such as the presence of fenestrations and expression of VAP-1, Stabilin-1, L-SIGN, which can be used to identify sinusoidal endothelium and to permit discrimination from vascular and lymphatic endothelial cells.
Collapse
MESH Headings
- Amine Oxidase (Copper-Containing)/genetics
- Amine Oxidase (Copper-Containing)/metabolism
- Biomarkers/metabolism
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelium, Lymphatic/cytology
- Endothelium, Vascular/cytology
- Gene Expression Regulation/genetics
- Humans
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Liver/blood supply
- Liver/cytology
- Liver/metabolism
- Liver Circulation
- Phenotype
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Lymphocyte Homing/genetics
- Receptors, Lymphocyte Homing/metabolism
Collapse
Affiliation(s)
- P F Lalor
- Liver Research Group, Institute of Biomedical Research, Division of Medical Science, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | | | | | | | | |
Collapse
|
152
|
Marzi A, Akhavan A, Simmons G, Gramberg T, Hofmann H, Bates P, Lingappa VR, Pöhlmann S. The signal peptide of the ebolavirus glycoprotein influences interaction with the cellular lectins DC-SIGN and DC-SIGNR. J Virol 2006; 80:6305-17. [PMID: 16775318 PMCID: PMC1488929 DOI: 10.1128/jvi.02545-05] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The C-type lectins DC-SIGN and DC-SIGNR (collectively referred to as DC-SIGN/R) bind to the ebolavirus glycoprotein (EBOV-GP) and augment viral infectivity. DC-SIGN/R strongly enhance infection driven by the GP of EBOV subspecies. Zaire (ZEBOV) but have a much less pronounced effect on infection mediated by the GP of EBOV subspecies. Sudan (SEBOV). For this study, we analyzed the determinants of the differential DC-SIGN/R interactions with ZEBOV- and SEBOV-GP. The efficiency of DC-SIGN engagement by ZEBOV-GP was dependent on the rate of GP incorporation into lentiviral particles, while appreciable virion incorporation of SEBOV-GP did not allow robust DC-SIGN/R usage. Forced incorporation of high-mannose carbohydrates into SEBOV-GP augmented the engagement of DC-SIGN/R to the levels observed with ZEBOV-GP, indicating that appropriate glycosylation of SEBOV-GP is sufficient for efficient DC-SIGN/R usage. However, neither signals for N-linked glycosylation unique to SEBOV- or ZEBOV-GP nor the highly variable and heavily glycosylated mucin-like domain modulated the interaction with DC-SIGN/R. In contrast, analysis of chimeric GPs identified the signal peptide as a determinant of DC-SIGN/R engagement. Thus, ZEBOV- but not SEBOV-GP was shown to harbor high-mannose carbohydrates, and GP modification with these glycans was controlled by the signal peptide. These results suggest that the signal peptide governs EBOV-GP interactions with DC-SIGN/R by modulating the incorporation of high-mannose carbohydrates into EBOV-GP. In summary, we identified the level of GP incorporation into virions and signal peptide-controlled glycosylation of GP as determinants of attachment factor engagement.
Collapse
Affiliation(s)
- Andrea Marzi
- Institute for Clinical and Molecular Virology and Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, Glückstrasse 6, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Seeds RE, Gordon S, Miller JL. Receptors and ligands involved in viral induction of type I interferon production by plasmacytoid dendritic cells. Immunobiology 2006; 211:525-35. [PMID: 16920491 PMCID: PMC7132488 DOI: 10.1016/j.imbio.2006.05.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Virus infection is sensed by the innate immune system which then rapidly initiates biosynthesis of type I interferon (IFN). The IFN signaling systems produce a broadly effective innate antiviral response by creating an antiviral state in both an autocrine and paracrine manner in cells and by activating innate and adaptive immunity. Plasmacytoid dendritic cells (pDCs) have the unique ability to produce very high levels of type I IFN following viral infection in vivo. Most recent research has focused on oligonucleotide-mediated induction of type I IFN production, implicating viral genome and replication intermediates as the stimulus for this response. However there are additional viral ligands which can potentially induce type I IFN production in pDCs, such as envelope glycoproteins, viral glycolipids, tegument, capsid or nuclear proteins. This area of viral immunology, which has been neglected in the literature, will be discussed here.
Collapse
Affiliation(s)
- Rosalind E Seeds
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | | | | |
Collapse
|
154
|
Lai WK, Sun PJ, Zhang J, Jennings A, Lalor PF, Hubscher S, McKeating JA, Adams DH. Expression of DC-SIGN and DC-SIGNR on human sinusoidal endothelium: a role for capturing hepatitis C virus particles. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:200-8. [PMID: 16816373 PMCID: PMC1698775 DOI: 10.2353/ajpath.2006.051191] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 04/04/2006] [Indexed: 12/11/2022]
Abstract
Hepatic sinusoidal endothelial cells are unique among endothelial cells in their ability to internalize and process a diverse range of antigens. DC-SIGNR, a type 2 C-type lectin expressed on liver sinusoids, has been shown to bind with high affinity to hepatitis C virus (HCV) E2 glycoprotein. DC-SIGN is a closely related homologue reported to be expressed only on dendritic cells and a subset of macrophages and has similar binding affinity to HCV E2 glycoprotein. These receptors function as adhesion and antigen presentation molecules. We report distinct patterns of DC-SIGNR and DC-SIGN expression in human liver tissue and show for the first time that both C-type lectins are expressed on sinusoidal endothelial cells. We confirmed that these receptors are functional by demonstrating their ability to bind HCV E2 glycoproteins. Although these lectins on primary sinusoidal cells support HCV E2 binding, they are unable to support HCV entry. These data support a model where DC-SIGN and DC-SIGNR on sinusoidal endothelium provide a mechanism for high affinity binding of circulating HCV within the liver sinusoids allowing subsequent transfer of the virus to underlying hepatocytes, in a manner analogous to DC-SIGN presentation of human immunodeficiency virus on dendritic cells.
Collapse
Affiliation(s)
- Wai K Lai
- Liver Research Laboratories, Institute of Biomedical Research, Birmingham University, Edgbaston, B15 2TH, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Methamphetamine modulates DC-SIGN expression by mature dendritic cells. J Neuroimmune Pharmacol 2006; 1:296-304. [PMID: 18040806 DOI: 10.1007/s11481-006-9027-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Accepted: 05/26/2006] [Indexed: 01/01/2023]
Abstract
We report that methamphetamine (meth) may act as cofactor in human immunodeficiency virus (HIV)-1 pathogenesis by increasing dendritic cell (DC)-specific intercellular adhesion molecule-3 (ICAM-3) grabbing non-integrin (DC-SIGN) expression on DCs. Mature DCs (MDCs), obtained from normal subjects, cultured with meth show an up-regulation of DC-SIGN gene and protein expression as analyzed by real-time quantitative polymerase chain reaction and fluorescence-activated cell-sorting analyses, respectively. Furthermore, these meth-induced effects were reversed by a dopamine D1 receptor antagonist (SCH 23390) and small interfering RNA specific to the D1 receptor (D1R) demonstrating that meth-induced effects are mediated through these receptors. Furthermore, meth in synergy with the HIV-1 peptide gp120 up-regulates DC-SIGN gene expression by MDCs. These data are the first evidence that meth up-regulates the expression of DC-SIGN on MDCs. A better understanding of the role of DC-SIGN in HIV-1 infection may help to design novel therapeutic strategies against the progression of HIV-1 disease in the drug-using population.
Collapse
|
156
|
Gramberg T, Zhu T, Chaipan C, Marzi A, Liu H, Wegele A, Andrus T, Hofmann H, Pöhlmann S. Impact of polymorphisms in the DC-SIGNR neck domain on the interaction with pathogens. Virology 2006; 347:354-63. [PMID: 16413044 PMCID: PMC7111803 DOI: 10.1016/j.virol.2005.11.033] [Citation(s) in RCA: 25] [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: 08/23/2005] [Revised: 09/19/2005] [Accepted: 11/23/2005] [Indexed: 11/28/2022]
Abstract
The lectins DC-SIGN and DC-SIGNR augment infection by human immunodeficiency virus (HIV), Ebolavirus (EBOV) and other pathogens. The neck domain of these proteins drives multimerization, which is believed to be required for efficient recognition of multivalent ligands. The neck domain of DC-SIGN consists of seven sequence repeats with rare variations. In contrast, the DC-SIGNR neck domain is polymorphic and, in addition to the wild type (wt) allele with seven repeat units, allelic forms with five and six sequence repeats are frequently found. A potential association of the DC-SIGNR genotype and risk of HIV-1 infection is currently under debate. Therefore, we investigated if DC-SIGNR alleles with five and six repeat units exhibit defects in pathogen capture. Here, we show that wt DC-SIGNR and patient derived alleles with five and six repeats bind viral glycoproteins, augment viral infection and tetramerize with comparable efficiency. Moreover, coexpression of wt DC-SIGNR and alleles with five repeats did not decrease the interaction with pathogens compared to expression of each allele alone, suggesting that potential formation of hetero-oligomers does not appreciably reduce pathogen binding, at least under conditions of high expression. Thus, our results do not provide evidence for diminished pathogen capture by DC-SIGNR alleles with five and six repeat units. Albeit, we cannot exclude that subtle, but in vivo relevant differences remained undetected, our analysis suggests that indirect mechanisms could account for the association of polymorphisms in the DC-SIGNR neck region with reduced risk of HIV-1 infection.
Collapse
MESH Headings
- Cell Adhesion Molecules/chemistry
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules/physiology
- Cell Culture Techniques
- HIV Infections/metabolism
- HIV-1/metabolism
- Lectins, C-Type/chemistry
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/physiology
- Polymorphism, Genetic
- Protein Structure, Tertiary/genetics
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/physiology
- Repetitive Sequences, Nucleic Acid/genetics
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
- Thomas Gramberg
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tuofu Zhu
- Department of Laboratory Medicine, University of Washington, School of Medicine, Seattle, WA 98195, USA
- Microbiology, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Chawaree Chaipan
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andrea Marzi
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Huanliang Liu
- Department of Laboratory Medicine, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Anja Wegele
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas Andrus
- Department of Laboratory Medicine, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Heike Hofmann
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Department of Medical Microbiology and Virology, University of Kiel, 24105 Kiel, Germany
| | - Stefan Pöhlmann
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
157
|
Davis CW, Nguyen HY, Hanna SL, Sánchez MD, Doms RW, Pierson TC. West Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and infection. J Virol 2006; 80:1290-301. [PMID: 16415006 PMCID: PMC1346927 DOI: 10.1128/jvi.80.3.1290-1301.2006] [Citation(s) in RCA: 266] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The C-type lectins DC-SIGN and DC-SIGNR bind mannose-rich glycans with high affinity. In vitro, cells expressing these attachment factors efficiently capture, and are infected by, a diverse array of appropriately glycosylated pathogens, including dengue virus. In this study, we investigated whether these lectins could enhance cellular infection by West Nile virus (WNV), a mosquito-borne flavivirus related to dengue virus. We discovered that DC-SIGNR promoted WNV infection much more efficiently than did DC-SIGN, particularly when the virus was grown in human cell types. The presence of a single N-linked glycosylation site on either the prM or E glycoprotein of WNV was sufficient to allow DC-SIGNR-mediated infection, demonstrating that uncleaved prM protein present on a flavivirus virion can influence viral tropism under certain circumstances. Preferential utilization of DC-SIGNR was a specific property conferred by the WNV envelope glycoproteins. Chimeras between DC-SIGN and DC-SIGNR demonstrated that the ability of DC-SIGNR to promote WNV infection maps to its carbohydrate recognition domain. WNV virions and subviral particles bound to DC-SIGNR with much greater affinity than DC-SIGN. We believe this is the first report of a pathogen interacting more efficiently with DC-SIGNR than with DC-SIGN. Our results should lead to the discovery of new mechanisms by which these well-studied lectins discriminate among ligands.
Collapse
Affiliation(s)
- Carl W Davis
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
158
|
Gramberg T, Caminschi I, Wegele A, Hofmann H, Pöhlmann S. Evidence that multiple defects in murine DC-SIGN inhibit a functional interaction with pathogens. Virology 2006; 345:482-91. [PMID: 16297949 PMCID: PMC7111805 DOI: 10.1016/j.virol.2005.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2005] [Revised: 08/09/2005] [Accepted: 10/12/2005] [Indexed: 12/14/2022]
Abstract
Certain viruses, bacteria, fungi and parasites target dendritic cells through the interaction with the cellular attachment factor DC-SIGN, making this C-type lectin an attractive target for therapeutic intervention. Studies on DC-SIGN function would be greatly aided by the establishment of a mouse model, however, it is unclear if the murine (m) homologue of human (h) DC-SIGN also binds to pathogens. Here, we investigated the interaction of mDC-SIGN, also termed CIRE, with the Ebolavirus glycoprotein (EBOV-GP), a ligand of hDC-SIGN. We found that mDC-SIGN neither binds EBOV-GP nor enhances infection by reporterviruses pseudotyped with EBOV-GP. Analysis of chimeras between mDC-SIGN and hDC-SIGN provided evidence that determinants in the carbohydrate recognition domain and in the neck domain of mDC-SIGN inhibit a functional interaction with EBOV-GP. Moreover, mDC-SIGN was found be monomeric, suggesting that lack of multimerization, which is believed to be required for efficient pathogen recognition by hDC-SIGN, might be one factor that prevents binding of mDC-SIGN to EBOV-GP. Our results suggest that mDC-SIGN on murine dendritic cells is not an adequate model for pathogen interactions with hDC-SIGN.
Collapse
Affiliation(s)
- Thomas Gramberg
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | | |
Collapse
|
159
|
Dakappagari N, Maruyama T, Renshaw M, Tacken P, Figdor C, Torensma R, Wild MA, Wu D, Bowdish K, Kretz-Rommel A. Internalizing antibodies to the C-type lectins, L-SIGN and DC-SIGN, inhibit viral glycoprotein binding and deliver antigen to human dendritic cells for the induction of T cell responses. THE JOURNAL OF IMMUNOLOGY 2006; 176:426-40. [PMID: 16365436 DOI: 10.4049/jimmunol.176.1.426] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The C-type lectin L-SIGN is expressed on liver and lymph node endothelial cells, where it serves as a receptor for a variety of carbohydrate ligands, including ICAM-3, Ebola, and HIV. To consider targeting liver/lymph node-specific ICAM-3-grabbing nonintegrin (L-SIGN) for therapeutic purposes in autoimmunity and infectious disease, we isolated and characterized Fabs that bind strongly to L-SIGN, but to a lesser degree or not at all to dendritic cell-specific ICAM-grabbing nonintegrin (DC-SIGN). Six Fabs with distinct relative affinities and epitope specificities were characterized. The Fabs and those selected for conversion to IgG were tested for their ability to block ligand (HIV gp120, Ebola gp, and ICAM-3) binding. Receptor internalization upon Fab binding was evaluated on primary human liver sinusoidal endothelial cells by flow cytometry and confirmed by confocal microscopy. Although all six Fabs internalized, three Fabs that showed the most complete blocking of HIVgp120 and ICAM-3 binding to L-SIGN also internalized most efficiently. Differences among the Fab panel in the ability to efficiently block Ebola gp compared with HIVgp120 suggested distinct binding sites. As a first step to consider the potential of these Abs for Ab-mediated Ag delivery, we evaluated specific peptide delivery to human dendritic cells. A durable human T cell response was induced when a tetanus toxide epitope embedded into a L-SIGN/DC-SIGN-cross-reactive Ab was targeted to dendritic cells. We believe that the isolated Abs may be useful for selective delivery of Ags to DC-SIGN- or L-SIGN-bearing APCs for the modulation of immune responses and for blocking viral infections.
Collapse
|
160
|
Chan VSF, Chan KYK, Chen Y, Poon LLM, Cheung ANY, Zheng B, Chan KH, Mak W, Ngan HYS, Xu X, Screaton G, Tam PKH, Austyn JM, Chan LC, Yip SP, Peiris M, Khoo US, Lin CLS. Homozygous L-SIGN (CLEC4M) plays a protective role in SARS coronavirus infection. Nat Genet 2006; 38:38-46. [PMID: 16369534 PMCID: PMC7097088 DOI: 10.1038/ng1698] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Accepted: 10/03/2005] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome (SARS) is caused by infection of a previously undescribed coronavirus (CoV). L-SIGN, encoded by CLEC4M (also known as CD209L), is a SARS-CoV binding receptor that has polymorphism in its extracellular neck region encoded by the tandem repeat domain in exon 4. Our genetic risk association study shows that individuals homozygous for CLEC4M tandem repeats are less susceptible to SARS infection. L-SIGN is expressed in both non-SARS and SARS-CoV-infected lung. Compared with cells heterozygous for L-SIGN, cells homozygous for L-SIGN show higher binding capacity for SARS-CoV, higher proteasome-dependent viral degradation and a lower capacity for trans infection. Thus, homozygosity for L-SIGN plays a protective role during SARS infection.
Collapse
Affiliation(s)
- Vera S F Chan
- Department of Surgery, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Kelvin Y K Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
- Department of Obsterics and Gynecology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Yongxiong Chen
- Department of Surgery, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Leo L M Poon
- Department of Microbiology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Annie N Y Cheung
- Department of Pathology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Bojian Zheng
- Department of Microbiology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Kwok-Hung Chan
- Department of Microbiology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - William Mak
- Department of Genome Research Centre, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Hextan Y S Ngan
- Department of Obsterics and Gynecology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Xiaoning Xu
- Human Immunology Unit, The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - Paul K H Tam
- Department of Surgery, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
- Department of Genome Research Centre, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | | | - Li-Chong Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Shea-Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, SAR China
| | - Malik Peiris
- Department of Microbiology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Ui-Soon Khoo
- Department of Pathology, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| | - Chen-Lung S Lin
- Department of Surgery, Hong Kong Jockey Club Clinical Research Center, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, SAR China
| |
Collapse
|
161
|
Perlman S, Holmes KV. Attachment factor and receptor engagement of SARS coronavirus and human coronavirus NL63. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 581:219-27. [PMID: 17037533 PMCID: PMC7124012 DOI: 10.1007/978-0-387-33012-9_37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Stanley Perlman
- Department of Pediatrics, University of Iowa, 52242 Iowa City, IA USA
| | - Kathryn V. Holmes
- Department of Microbiology, University of Colorado Health Sciences Center at Fitzsimons, 80045-8333 Aurora, CO USA
| |
Collapse
|
162
|
Balasubramanian A, Munshi N, Koziel MJ, Hu Z, Liang TJ, Groopman JE, Ganju RK. Structural proteins of Hepatitis C virus induce interleukin 8 production and apoptosis in human endothelial cells. J Gen Virol 2005; 86:3291-3301. [PMID: 16298974 DOI: 10.1099/vir.0.81056-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) infection is associated with inflammation of liver endothelium, which contributes to the pathogenesis of chronic hepatitis. The mechanism of this endothelitis is not understood, since the virus does not appear to infect endothelial cells productively. Here, an 'innocent bystander' mechanism related to HCV proteins was hypothesized and it was investigated whether the binding of HCV particles to human endothelium induced functional changes in the cells. Exposure of human umbilical vein endothelial cells (HUVECs) to HCV-like particles (HCV-LPs) resulted in increased interleukin 8 (IL8) production and induction of apoptosis. The IL8 supernatants collected after stimulation of HUVECs with HCV-LPs, BV-GUS (control baculovirus containing beta-glucuronidase) and appropriate controls were used to assay the transendothelial migration of neutrophils. This assay confirmed that HCV-LP-induced IL8 was functionally active. Using specific NF-kappaB inhibitors, it was also shown that HCV-LP-induced NF-kappaB activity mediated IL8 production in HUVECs. Apoptosis appeared to be mediated by the Fas/Fas-L pathway, as neutralizing antibodies for Fas and Fas-L significantly protected HUVECs against HCV-LP-induced apoptosis. Treatment of HUVECs with HCV-LPs also enhanced cellular Fas-L expression and augmented caspase-3 activation. This was confirmed by using a specific caspase-3 inhibitor, Z-Asp-Glu-Val-Asp-fluoromethyl ketone. As shown by blocking of specific chemokine receptors for IL8 on HUVECs, the induction of IL8 did not appear to contribute to HCV-LP-induced apoptosis. These results suggest that HCV proteins can trigger the release of inflammatory chemokines such as IL8 and cause endothelial apoptosis, thereby facilitating endothelitis.
Collapse
Affiliation(s)
- Anuradha Balasubramanian
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 4 Blackfan Circle, 3rd Floor, Boston, MA 02115, USA
| | - Neru Munshi
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 4 Blackfan Circle, 3rd Floor, Boston, MA 02115, USA
| | - Margaret J Koziel
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, 4 Blackfan Circle, 3rd Floor, Boston, MA 02115, USA
| | - Zongyi Hu
- Liver Diseases Section, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - T Jake Liang
- Liver Diseases Section, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jerome E Groopman
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 4 Blackfan Circle, 3rd Floor, Boston, MA 02115, USA
| | - Ramesh K Ganju
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 4 Blackfan Circle, 3rd Floor, Boston, MA 02115, USA
| |
Collapse
|
163
|
Gramberg T, Hofmann H, Möller P, Lalor PF, Marzi A, Geier M, Krumbiegel M, Winkler T, Kirchhoff F, Adams DH, Becker S, Münch J, Pöhlmann S. LSECtin interacts with filovirus glycoproteins and the spike protein of SARS coronavirus. Virology 2005; 340:224-36. [PMID: 16051304 PMCID: PMC7111772 DOI: 10.1016/j.virol.2005.06.026] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2005] [Revised: 05/10/2005] [Accepted: 06/13/2005] [Indexed: 11/23/2022]
Abstract
Cellular attachment factors like the C-type lectins DC-SIGN and DC-SIGNR (collectively referred to as DC-SIGN/R) can augment viral infection and might promote viral dissemination in and between hosts. The lectin LSECtin is encoded in the same chromosomal locus as DC-SIGN/R and is coexpressed with DC-SIGNR on sinusoidal endothelial cells in liver and lymphnodes. Here, we show that LSECtin enhances infection driven by filovirus glycoproteins (GP) and the S protein of SARS coronavirus, but does not interact with human immunodeficiency virus type-1 and hepatitis C virus envelope proteins. Ligand binding to LSECtin was inhibited by EGTA but not by mannan, suggesting that LSECtin unlike DC-SIGN/R does not recognize high-mannose glycans on viral GPs. Finally, we demonstrate that LSECtin is N-linked glycosylated and that glycosylation is required for cell surface expression. In summary, we identified LSECtin as an attachment factor that in conjunction with DC-SIGNR might concentrate viral pathogens in liver and lymph nodes.
Collapse
Affiliation(s)
- Thomas Gramberg
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Heike Hofmann
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Peggy Möller
- Institute for Virology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Patricia F. Lalor
- Liver Research Group, Institute for Biomedical Science, The University of Birmingham Medical School, Edgbaston, Birmingham, UK
- MRC Centre for Immune Regulation, The University of Birmingham Medical School, Edgbaston, Birmingham, UK
| | - Andrea Marzi
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Martina Geier
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mandy Krumbiegel
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas Winkler
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Chair of Genetics, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Frank Kirchhoff
- Department of Virology, Universitätsklinikum Ulm, 89081 Ulm, Germany
| | - David H. Adams
- Liver Research Group, Institute for Biomedical Science, The University of Birmingham Medical School, Edgbaston, Birmingham, UK
- MRC Centre for Immune Regulation, The University of Birmingham Medical School, Edgbaston, Birmingham, UK
| | - Stephan Becker
- Institute for Virology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Jan Münch
- Department of Virology, Universitätsklinikum Ulm, 89081 Ulm, Germany
| | - Stefan Pöhlmann
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Corresponding author. Nikolaus-Fiebiger-Center, University Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany. Fax: +49 9131 8529111.
| |
Collapse
|
164
|
Abstract
Genetic polymorphisms in human genes can influence the risk for HIV-1 infection and disease progression, although the reported effects of these alleles have been inconsistent. This review highlights the recent discoveries on global and Chinese genetic polymorphisms and their association with HIV-1 transmission and disease progression.
Collapse
Affiliation(s)
- Tuo Fu Zhu
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98195-8070, USA.
| | | | | | | | | |
Collapse
|
165
|
Barreiro LB, Patin E, Neyrolles O, Cann HM, Gicquel B, Quintana-Murci L. The heritage of pathogen pressures and ancient demography in the human innate-immunity CD209/CD209L region. Am J Hum Genet 2005; 77:869-86. [PMID: 16252244 PMCID: PMC1271393 DOI: 10.1086/497613] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 08/26/2005] [Indexed: 10/26/2022] Open
Abstract
The innate immunity system constitutes the first line of host defense against pathogens. Two closely related innate immunity genes, CD209 and CD209L, are particularly interesting because they directly recognize a plethora of pathogens, including bacteria, viruses, and parasites. Both genes, which result from an ancient duplication, possess a neck region, made up of seven repeats of 23 amino acids each, known to play a major role in the pathogen-binding properties of these proteins. To explore the extent to which pathogens have exerted selective pressures on these innate immunity genes, we resequenced them in a group of samples from sub-Saharan Africa, Europe, and East Asia. Moreover, variation in the number of repeats of the neck region was defined in the entire Human Genome Diversity Panel for both genes. Our results, which are based on diversity levels, neutrality tests, population genetic distances, and neck-region length variation, provide genetic evidence that CD209 has been under a strong selective constraint that prevents accumulation of any amino acid changes, whereas CD209L variability has most likely been shaped by the action of balancing selection in non-African populations. In addition, our data point to the neck region as the functional target of such selective pressures: CD209 presents a constant size in the neck region populationwide, whereas CD209L presents an excess of length variation, particularly in non-African populations. An additional interesting observation came from the coalescent-based CD209 gene tree, whose binary topology and time depth (approximately 2.8 million years ago) are compatible with an ancestral population structure in Africa. Altogether, our study has revealed that even a short segment of the human genome can uncover an extraordinarily complex evolutionary history, including different pathogen pressures on host genes as well as traces of admixture among archaic hominid populations.
Collapse
Affiliation(s)
- Luis B Barreiro
- Centre National de la Recherche Scientifique FRE 2849, Unit of Molecular Prevention and Therapy of Human Diseases, Institut Pasteur, 25, 75724 Paris Cedex 15, France
| | | | | | | | | | | |
Collapse
|
166
|
Caparrós E, Serrano D, Puig-Kröger A, Riol L, Lasala F, Martinez I, Vidal-Vanaclocha F, Delgado R, Rodríguez-Fernández JL, Rivas L, Corbí AL, Colmenares M. Role of the C-type lectins DC-SIGN and L-SIGN in Leishmania interaction with host phagocytes. Immunobiology 2005; 210:185-93. [PMID: 16164025 PMCID: PMC7114652 DOI: 10.1016/j.imbio.2005.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Leishmaniasis is a parasitic disease that courses with cutaneous or visceral clinical manifestations. The amastigote stage of the parasite infects phagocytes and modulates the effector function of the host cells. Our group has described that the interaction between Leishmania and immature monocyte-derived dendritic cells (DCs) takes place through dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN), a C-type lectin that specifically recognizes fungal, viral and bacterial pathogens. The DC-SIGN-mediated recognition of Leishmania amastigotes does not induce DC maturation, and the DC-SIGN ligand/s on Leishmania parasites is/are still unknown. We have also found that the DC-SIGN-related molecule L-SIGN, specifically expressed in lymph node and liver sinusoidal endothelial cells, acts as a receptor for L. infantum, the parasite responsible for visceral leishmaniasis, but does not recognize L. pifanoi, which causes the cutaneous form of the disease. Therefore, DC-SIGN and L-SIGN differ in their ability to interact with Leishmania species responsible for either visceral or cutaneous leishmaniasis. A deeper knowledge of the parasite-C-type lectin interaction may be helpful for the design of new DC-based therapeutic vaccines against Leishmania infections.
Collapse
Affiliation(s)
- Esther Caparrós
- Centro de Investigaciones Biológicas (CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Goffard A, Callens N, Bartosch B, Wychowski C, Cosset FL, Montpellier C, Dubuisson J. Role of N-linked glycans in the functions of hepatitis C virus envelope glycoproteins. J Virol 2005; 79:8400-9. [PMID: 15956584 PMCID: PMC1143753 DOI: 10.1128/jvi.79.13.8400-8409.2005] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) encodes two viral envelope glycoproteins. E1 contains 4 or 5 N-linked glycosylation sites and E2 contains up to 11, with most of the sites being well conserved, suggesting that they play an essential role in some functions of these proteins. For this study, we used retroviral pseudotyped particles harboring mutated HCV envelope glycoproteins to study these glycans. The mutants were named with an N followed by a number related to the relative position of the potential glycosylation site in each glycoprotein (E1N1 to E1N4 for E1 mutants and E2N1 to E2N11 for E2 mutants). The characterization of these mutants allowed us to define three phenotypes. For the first group (E1N3, E2N3, E2N5, E2N6, E2N7, and E2N9), the infectivities of the mutants were close to that of the wild type. The second group (E1N1, E1N2, E1N4, E2N1, and E2N11) contained mutants that were still infectious but whose infectivities were reduced to <50% that of the wild type. The third group (E2N2, E2N4, E2N8, and E2N10) contained mutants that had almost totally lost infectivity. The absence of infectivity of the E2N8 and E2N10 mutants was due to the lack of incorporation of the E1E2 heterodimer into HCVpp, which was due to misfolding of the heterodimer, as shown by immunoprecipitation with conformation-sensitive antibodies and by a CD81 pull-down assay. The absence of infectivity of the E2N2 and E2N4 mutants indicated that these two glycans are involved in controlling HCV entry. Altogether, the data indicate that some glycans of HCV envelope glycoproteins play a major role in protein folding and others play a role in HCV entry.
Collapse
Affiliation(s)
- Anne Goffard
- CNRS-UPR2511, Institut de Biologie de Lille, France
| | | | | | | | | | | | | |
Collapse
|
168
|
Liu H, Hladik F, Andrus T, Sakchalathorn P, Lentz GM, Fialkow MF, Corey L, McElrath MJ, Zhu T. Most DC-SIGNR transcripts at mucosal HIV transmission sites are alternatively spliced isoforms. Eur J Hum Genet 2005; 13:707-15. [PMID: 15812562 DOI: 10.1038/sj.ejhg.5201409] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The repeat region of DC-SIGNR (CD209L) is polymorphic on the genomic level, and, in a separate study, we observed a correlation between the DC-SIGNR genotype and HIV-1 susceptibility during sexual contact. However, previous investigations using immunohistochemistry failed to detect membrane-bound DC-SIGNR on cells in the genital and rectal mucosa. We therefore explored the presence of DC-SIGNR in these compartments with a more sensitive limiting dilution RT-PCR, which also allowed for quantification of alternatively spliced mRNA isoforms. DC-SIGN (CD209) and DC-SIGNR mRNA transcript isoforms were found in all 12 vaginal and two rectal biopsies obtained from 14 healthy individuals. For DC-SIGNR, we detected significantly more isoform than full-length transcripts (mean copy numbers/mug RNA: 602 vs 26; P=0.0009). Four mucosal samples lacked full-length DC-SIGNR transcripts entirely. Cloning and sequencing of DC-SIGNR mRNA in three additional individuals revealed a diverse repertoire of DC-SIGNR isoforms, many of which encoded for proteins predicted to be soluble and secreted. Indeed, in one vaginal sample, we detected only soluble isoforms. In conjunction with our prior observation that the DC-SIGNR genotype has an effect on HIV-1 transmission in vivo, these findings emphasize that DC-SIGNR, in addition to DC-SIGN, should be considered as a cofactor in sexual HIV-1 transmission. Soluble isoforms, in particular, may modulate the efficiency of viral transmission and dissemination.
Collapse
Affiliation(s)
- Huanliang Liu
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Nair MPN, Mahajan SD, Schwartz SA, Reynolds J, Whitney R, Bernstein Z, Chawda RP, Sykes D, Hewitt R, Hsiao CB. Cocaine modulates dendritic cell-specific C type intercellular adhesion molecule-3-grabbing nonintegrin expression by dendritic cells in HIV-1 patients. THE JOURNAL OF IMMUNOLOGY 2005; 174:6617-26. [PMID: 15905500 DOI: 10.4049/jimmunol.174.11.6617] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We report that cocaine may act as cofactor in HIV pathogenesis by increasing dendritic cell-specific C type ICAM-3-grabbing nonintegrin (DC-SIGN) expression on dendritic cells (DC). Our results show that cocaine-using, long-term nonprogressors and normal progressors of HIV infection manifest significantly higher levels of DC-SIGN compared with cocaine-nonusing long-term nonprogressors and normal progressors, respectively. Furthermore, in vitro HIV infection of MDC from normal subjects cultured with cocaine and/or HIV peptides up-regulated DC-SIGN, confirming our in vivo finding. Cocaine, in synergy with HIV peptides, also up-regulates DC-SIGN gene expression by MDC. Furthermore, the cocaine-induced effects were reversed by a D1 receptor antagonist demonstrating the specificity of the reaction. Our results indicate that cocaine exacerbates HIV infection by up-regulating DC-SIGN on DC and these effects are mediated via dysregulation of MAPKs. These data are the first evidence that cocaine up-regulates the expression of DC-SIGN on DC. A better understanding of the role of DC-SIGN in HIV infection may help to design novel therapeutic strategies against the progression of HIV disease in the drug-using population.
Collapse
Affiliation(s)
- Madhavan P N Nair
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York and Buffalo General Hospital, 14203, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Sansonno D, Lauletta G, Montrone M, Grandaliano G, Schena FP, Dammacco F. Hepatitis C virus RNA and core protein in kidney glomerular and tubular structures isolated with laser capture microdissection. Clin Exp Immunol 2005; 140:498-506. [PMID: 15932511 PMCID: PMC1809381 DOI: 10.1111/j.1365-2249.2005.02778.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The role of hepatitis C virus (HCV) in the production of renal injury has been extensively investigated, though with conflicting results. Laser capture microdissection (LCM) was performed to isolate and collect glomeruli and tubules from 20 consecutive chronically HCV-infected patients, namely 6 with membranoproliferative glomerulonephritis, 4 with membranous glomerulonephritis, 7 with focal segmental glomerulosclerosis and 3 with IgA-nephropathy. RNA for amplification of specific viral sequences was provided by terminal continuation methodology and compared with the expression profile of HCV core protein. For each case two glomeruli and two tubular structures were microdissected and processed. HCV RNA sequences were demonstrated in 26 (65%) of 40 glomeruli, but in only 4 (10%) of the tubules (P < 0.05). HCV core protein was concomitant with viral sequences in the glomeruli and present in 31 of the 40 tubules. HCV RNA and/or HCV core protein was found in all four disease types. The immunohistochemical picture of HCV core protein was compared with the LCM-based immunoassays of the adjacent tissue sections. Immune deposits were detected in 7 (44%) of 16 biopsy samples shown to be positive by extraction methods. The present study indicates that LCM is a reliable method for measuring both HCV RNA genomic sequences and HCV core protein in kidney functional structures from chronically HCV-infected patients with different glomerulopathies and provides a useful baseline estimate to define the role of HCV in the production of renal injury. The different distribution of HCV RNA and HCV-related proteins may reflect a peculiar 'affinity' of kidney microenvironments for HCV and point to distinct pathways of HCV-related damage in glomeruli and tubules.
Collapse
Affiliation(s)
- D Sansonno
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy.
| | | | | | | | | | | |
Collapse
|
171
|
Tang BSF, Chan KH, Cheng VCC, Woo PCY, Lau SKP, Lam CCK, Chan TL, Wu AKL, Hung IFN, Leung SY, Yuen KY. Comparative host gene transcription by microarray analysis early after infection of the Huh7 cell line by severe acute respiratory syndrome coronavirus and human coronavirus 229E. J Virol 2005; 79:6180-93. [PMID: 15858003 PMCID: PMC1091719 DOI: 10.1128/jvi.79.10.6180-6193.2005] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The pathogenesis of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) at the cellular level is unclear. No human cell line was previously known to be susceptible to both SARS-CoV and other human coronaviruses. Huh7 cells were found to be susceptible to both SARS-CoV, associated with SARS, and human coronavirus 229E (HCoV-229E), usually associated with the common cold. Highly lytic and productive rates of infections within 48 h of inoculation were reproducible with both viruses. The early transcriptional profiles of host cell response to both types of infection at 2 and 4 h postinoculation were determined by using the Affymetrix HG-U133A microarray (about 22,000 genes). Much more perturbation of cellular gene transcription was observed after infection by SARS-CoV than after infection by HCoV-229E. Besides the upregulation of genes associated with apoptosis, which was exactly opposite to the previously reported effect of SARS-CoV in a colonic carcinoma cell line, genes related to inflammation, stress response, and procoagulation were also upregulated. These findings were confirmed by semiquantitative reverse transcription-PCR, reverse transcription-quantitative PCR for mRNA of genes, and immunoassays for some encoded proteins. These transcriptomal changes are compatible with the histological changes of pulmonary vasculitis and microvascular thrombosis in addition to the diffuse alveolar damage involving the pneumocytes.
Collapse
Affiliation(s)
- Bone S F Tang
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Center of Infection and Immunology, Queen Mary Hospital, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Cheynet V, Ruggieri A, Oriol G, Blond JL, Boson B, Vachot L, Verrier B, Cosset FL, Mallet F. Synthesis, assembly, and processing of the Env ERVWE1/syncytin human endogenous retroviral envelope. J Virol 2005; 79:5585-93. [PMID: 15827173 PMCID: PMC1082723 DOI: 10.1128/jvi.79.9.5585-5593.2005] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Syncytin is a fusogenic protein involved in the formation of the placental syncytiotrophoblast layer. This protein is encoded by the envelope gene of the ERVWE1 proviral locus belonging to the human endogenous retrovirus W (HERV-W) family. The HERV-W infectious ancestor entered the primate lineage 25 to 40 million years ago. Although the syncytin fusion property has been clearly demonstrated, little is known about this cellular protein maturation process with respect to classical infectious retrovirus envelope proteins. Here we show that the cellular syncytin protein is synthesized as a glycosylated gPr73 precursor cleaved into two mature proteins, a gp50 surface subunit (SU) and a gp24 transmembrane subunit (TM). These SU and TM subunits are found associated as homotrimers. The intracytoplasmic tail is critical to the fusogenic phenotype, although its cleavage requirements seem to have diverged from those of classical retroviral maturation.
Collapse
Affiliation(s)
- V Cheynet
- Laboratoire de Vectorologie Rétrovirale et Thérapie Génique, INSERM U412, Ecole Normale Supérieure de Lyon, 69364 Lyon, France
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Sharova N, Swingler C, Sharkey M, Stevenson M. Macrophages archive HIV-1 virions for dissemination in trans. EMBO J 2005; 24:2481-9. [PMID: 15920469 PMCID: PMC1173148 DOI: 10.1038/sj.emboj.7600707] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Accepted: 05/12/2005] [Indexed: 01/25/2023] Open
Abstract
Viruses have evolved various strategies in order to persist within the host. To date, most information on mechanisms of HIV-1 persistence has been derived from studies with lymphocytes, but there is little information regarding mechanisms that govern HIV-1 persistence in macrophages. It has previously been demonstrated that virus assembly in macrophages occurs in cytoplasmic vesicles, which exhibit the characteristics of multivesicular bodies or late endosomes. The infectious stability of virions that assemble intracellularly in macrophages has not been evaluated. We demonstrate that virions assembling intracellularly in primary macrophages retain infectivity for extended intervals. Infectious virus was recovered directly from cytoplasmic lysates of macrophages and could be transmitted from macrophages to peripheral blood lymphocytes in trans 6 weeks after ongoing viral replication was blocked. Cell-associated virus decayed significantly from 1 to 2 weeks post infection, but decreased minimally thereafter. The persistence of intracellular virions did not require the viral accessory proteins Vpu or Nef. The stable sequestration of infectious virions within cytoplasmic compartments of macrophages may represent an additional mechanism for viral persistence in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Natalia Sharova
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Catherine Swingler
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mark Sharkey
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mario Stevenson
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Medical School, Two Biotech, Suite 319, 373 Plantation Street, Worcester, MA 01605, USA. Tel.: +1 508 856 3645; Fax: +1 508 856 4075; E-mail:
| |
Collapse
|
174
|
Linehan SA, Weber R, McKercher S, Ripley RM, Gordon S, Martin P. Enhanced expression of the mannose receptor by endothelial cells of the liver and spleen microvascular beds in the macrophage-deficient PU.1 null mouse. Histochem Cell Biol 2005; 123:365-76. [PMID: 15912359 DOI: 10.1007/s00418-005-0767-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2005] [Indexed: 11/26/2022]
Abstract
Mice null for the haematopoietic lineage-specific transcription factor PU.1 lack mature Mphi and are compromised in their ability to clear cellular debris from the blood circulation. We investigated the possibility that non-professional phagocytes may partially compensate for the lack of Mphi in clearance functions. In the absence of Kupffer cells (resident liver Mphi) in the PU.1 null mice, electron microscopy revealed ingested debris in sinusoidal endothelial cells and hepatocytes although debris was also seen free in blood vessels. To investigate whether an increased clearance function of non-professional phagocytes might be linked to expression of Mphi-associated phagocytic and pinocytic receptors by other cells in PU.1 null mouse, we examined expression of several candidate proteins by immunocytochemistry and Western blotting. We found mannose receptor (MR) comparably expressed in PU.1 null and PU.1+ mice liver and spleen whereas class A scavenger receptor was substantially reduced and complement receptor 3 was absent in PU.1 null animals. By morphometric analysis, liver and spleen sinusoidal endothelial cells were seen to express significantly more MR in the PU.1 null mouse. This study provides the first evidence of apparently compensatory alterations in the microvasculature of the Mphi-deficient PU.1 null mouse.
Collapse
MESH Headings
- Animals
- Blotting, Western
- CD11b Antigen/analysis
- Endothelial Cells/metabolism
- Endothelial Cells/ultrastructure
- Female
- Genotype
- Immunohistochemistry
- Kupffer Cells/metabolism
- Kupffer Cells/ultrastructure
- Lectins, C-Type/biosynthesis
- Liver/blood supply
- Liver/cytology
- Macrophages/cytology
- Macrophages/metabolism
- Male
- Mannose Receptor
- Mannose-Binding Lectins/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Microscopy, Electron, Transmission
- Phagocytes/metabolism
- Phagocytes/ultrastructure
- Proto-Oncogene Proteins/genetics
- Receptors, Cell Surface/biosynthesis
- Receptors, Immunologic/analysis
- Receptors, Scavenger
- Scavenger Receptors, Class A
- Spleen/blood supply
- Spleen/cytology
- Trans-Activators/genetics
Collapse
Affiliation(s)
- Sheena A Linehan
- Sir William Dunn School of Pathology, South Parks Road, Oxford, OX1 3RE, UK
| | | | | | | | | | | |
Collapse
|
175
|
Snyder GA, Ford J, Torabi-Parizi P, Arthos JA, Schuck P, Colonna M, Sun PD. Characterization of DC-SIGN/R interaction with human immunodeficiency virus type 1 gp120 and ICAM molecules favors the receptor's role as an antigen-capturing rather than an adhesion receptor. J Virol 2005; 79:4589-98. [PMID: 15795245 PMCID: PMC1069580 DOI: 10.1128/jvi.79.8.4589-4598.2005] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The dendritic cell (DC)-specific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin binding receptor (DC-SIGN) was shown to bind human immunodeficiency virus type 1 (HIV-1) viral envelope protein gp120 and proposed to function as a Trojan horse to enhance trans-virus infection to host T cells. To better understand the mechanism by which DC-SIGN and DC-SIGNR selectively bind HIV-1 gp120, we constructed a series of deletion mutations in the repeat regions of both receptors. Different truncated receptors exist in different oligomeric forms. The carbohydrate binding domain without any repeats was monomeric, whereas the full extracellular receptors existed as tetramers. All reconstituted receptors retained their ability to bind gp120. The dissociation constant, however, differed drastically from micromolar values for the monomeric receptors to nanomolar values for the tetrameric receptors, suggesting that the repeat region of these receptors contributes to the avidity of gp120 binding. Such oligomerization may provide a mechanism for the receptor to selectively recognize pathogens containing multiple high-mannose-concentration carbohydrates. In contrast, the receptors bound to ICAMs with submicromolar affinities that are similar to those of two nonspecific cell surface glycoproteins, FcgammaRIIb and FcgammaRIII, and the oligomerization of DC-SIGNR resulted in no increase in binding affinity to ICAM-3. These findings suggest that DC-SIGN may not discriminate other cell surface glycoproteins from ICAM-3 binding. The pH dependence in DC-SIGN binding to gp120 showed that the receptor retained high-affinity gp120 binding at neutral pH but lost gp120 binding at pH 5, suggesting a release mechanism of HIV in the acidic endosomal compartment by DC-SIGN. Our work contradicts the function of DC-SIGN as a Trojan horse to facilitate HIV-1 infection; rather, it supports the function of DC-SIGN/R (a designation referring to both DC-SIGN and DC-SIGNR) as an antigen-capturing receptor.
Collapse
Affiliation(s)
- Greg A Snyder
- Laboratory of Immunogenetics, Structural Immunology Section, NIAID, NIH, Twinbrook II, 12441 Parklawn Dr., Rockville, MD 20852, USA
| | | | | | | | | | | | | |
Collapse
|
176
|
Koppel EA, Ludwig IS, Hernandez MS, Lowary TL, Gadikota RR, Tuzikov AB, Vandenbroucke-Grauls CMJE, van Kooyk Y, Appelmelk BJ, Geijtenbeek TBH. Identification of the mycobacterial carbohydrate structure that binds the C-type lectins DC-SIGN, L-SIGN and SIGNR1. Immunobiology 2005; 209:117-27. [PMID: 15481146 DOI: 10.1016/j.imbio.2004.03.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Mycobacterium tuberculosis represents a worldwide health risk and although macrophages are primarily infected, dendritic cells (DC) are important in inducing cellular immune responses against M. tuberculosis. Recent studies have demonstrated that M. tuberculosis targets the DC-specific C-type lectin DC-SIGN to inhibit the immuno-stimulatory function of DC through the interaction of the mycobacterial mannosylated lipoarabinomannan (ManLAM) to DC-SIGN, which prevents DC maturation and induces the immuno-suppressive cytokine IL-10. This may contribute to survival and persistence of M. tuberculosis. Here, we have identified the specific pathogen-derived carbohydrate structure on ManLAM that is recognized by DC-SIGN. We have synthesized the mannose-cap oligosaccharides man-ara, (man)2-ara and (man)3-ara, and demonstrate that these neoglycoconjugates are specifically bound by DC-SIGN. Moreover, we demonstrate that the human and murine DC-SIGN homologue L-SIGN and SIGNR1, respectively, also interact with mycobacteria through ManLAM. Both homologues have the highest affinity for the (man)3-ara structure, similar to DC-SIGN. This study provides information about the specific carbohydrate structures on pathogens that are recognized by DC-SIGN, and may provide strategies to develop vaccines against these pathogens. Moreover, the identification of SIGNR1 as a receptor for ManLAM will enable in vivo studies to investigate the role of DC-SIGN in M. tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Estella A Koppel
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Snyder GA, Colonna M, Sun PD. The structure of DC-SIGNR with a portion of its repeat domain lends insights to modeling of the receptor tetramer. J Mol Biol 2005; 347:979-89. [PMID: 15784257 PMCID: PMC7094344 DOI: 10.1016/j.jmb.2005.01.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Revised: 01/17/2005] [Accepted: 01/26/2005] [Indexed: 11/29/2022]
Abstract
The dendritic cell-specific ICAM-3 non-integrin (DC-SIGN) and its close relative DC-SIGNR recognize various glycoproteins, both pathogenic and cellular, through the receptor lectin domain-mediated carbohydrate recognition. While the carbohydrate-recognition domains (CRD) exist as monomers and bind individual carbohydrates with low affinity and are permissive in nature, the full-length receptors form tetramers through their repeat domain and recognize specific ligands with high affinity. To understand the tetramer-based ligand binding avidity, we determined the crystal structure of DC-SIGNR with its last repeat region. Compared to the carbohydrate-bound CRD structure, the structure revealed conformational changes in the calcium and carbohydrate coordination loops of CRD, an additional disulfide bond between the N and the C termini of the CRD, and a helical conformation for the last repeat. On the basis of the current crystal structure and other published structures with sequence homology to the repeat domain, we generated a tetramer model for DC-SIGN/R using homology modeling and propose a ligand-recognition index to identify potential receptor ligands.
Collapse
MESH Headings
- Amino Acid Sequence
- Binding Sites
- Calcium/metabolism
- Carbohydrate Metabolism
- Cell Adhesion Molecules/chemistry
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Crystallography, X-Ray
- Humans
- Lectins, C-Type/chemistry
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Ligands
- Models, Molecular
- Molecular Sequence Data
- Protein Binding
- Protein Structure, Quaternary
- Protein Structure, Tertiary
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Sequence Alignment
Collapse
Affiliation(s)
- Greg A. Snyder
- Laboratory of Immunogenetics, Structural Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, 12441 Parklawn Drive, Rockville, MD 20852, USA
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Peter D. Sun
- Laboratory of Immunogenetics, Structural Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, 12441 Parklawn Drive, Rockville, MD 20852, USA
- Corresponding author.
| |
Collapse
|
178
|
Botos I, Wlodawer A. Proteins that bind high-mannose sugars of the HIV envelope. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2005; 88:233-82. [PMID: 15572157 DOI: 10.1016/j.pbiomolbio.2004.05.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A broad range of proteins bind high-mannose carbohydrates found on the surface of the envelope protein gp120 of the human immunodeficiency virus and thus interfere with the viral life cycle, providing a potential new way of controlling HIV infection. These proteins interact with the carbohydrate moieties in different ways. A group of them interacts as typical C-type lectins via a Ca2+ ion. Another group interacts with specific single, terminal sugars, without the help of a metal cation. A third group is involved in more intimate interactions, with multiple carbohydrate rings and no metal ion. Finally, there is a group of lectins for which the interaction mode has not yet been elucidated. This review summarizes, principally from a structural point of view, the current state of knowledge about these high-mannose binding proteins and their mode of sugar binding.
Collapse
Affiliation(s)
- Istvan Botos
- Macromolecular Crystallography Laboratory, National Cancer Institute, NCI-Frederick, Building 536, Room 5, Frederick, MD 21702-1201, USA
| | | |
Collapse
|
179
|
Arrighi JF, Pion M, Garcia E, Escola JM, van Kooyk Y, Geijtenbeek TB, Piguet V. DC-SIGN-mediated infectious synapse formation enhances X4 HIV-1 transmission from dendritic cells to T cells. ACTA ACUST UNITED AC 2005; 200:1279-88. [PMID: 15545354 PMCID: PMC2211914 DOI: 10.1084/jem.20041356] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dendritic cells (DCs) are essential for the early events of human immunodeficiency virus (HIV) infection. Model systems of HIV sexual transmission have shown that DCs expressing the DC-specific C-type lectin DC-SIGN capture and internalize HIV at mucosal surfaces and efficiently transfer HIV to CD4+ T cells in lymph nodes, where viral replication occurs. Upon DC–T cell clustering, internalized HIV accumulates on the DC side at the contact zone (infectious synapse), between DCs and T cells, whereas HIV receptors and coreceptors are enriched on the T cell side. Viral concentration at the infectious synapse may explain, at least in part, why DC transmission of HIV to T cells is so efficient. Here, we have investigated the role of DC-SIGN on primary DCs in X4 HIV-1 capture and transmission using small interfering RNA–expressing lentiviral vectors to specifically knockdown DC-SIGN. We demonstrate that DC-SIGN− DCs internalize X4 HIV-1 as well as DC-SIGN+ DCs, although binding of virions is reduced. Strikingly, DC-SIGN knockdown in DCs selectively impairs infectious synapse formation between DCs and resting CD4+ T cells, but does not prevent the formation of DC–T cells conjugates. Our results demonstrate that DC-SIGN is required downstream from viral capture for the formation of the infectious synapse between DCs and T cells. These findings provide a novel explanation for the role of DC-SIGN in the transfer and enhancement of HIV infection from DCs to T cells, a crucial step for HIV transmission and pathogenesis.
Collapse
Affiliation(s)
- Jean-François Arrighi
- Dept. of Dermatology and Venereology, University Hospital of Geneva, 4-752, 24 Rue Micheli-du-Crest, 1211 Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
180
|
Lanoue A, Clatworthy MR, Smith P, Green S, Townsend MJ, Jolin HE, Smith KGC, Fallon PG, McKenzie ANJ. SIGN-R1 contributes to protection against lethal pneumococcal infection in mice. ACTA ACUST UNITED AC 2005; 200:1383-93. [PMID: 15583012 PMCID: PMC2211941 DOI: 10.1084/jem.20040795] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Rapid clearance of pathogens is essential for successful control of pyogenic bacterial infection. Previous experiments have shown that antibody to specific intracellular adhesion molecule-grabbing nonintegrin (SIGN)-R1 inhibits uptake of capsular polysaccharide by marginal zone macrophages, suggesting a role for SIGN-R1 in this process. We now demonstrate that mice lacking SIGN-R1 (a mouse homologue of human dendritic cell–SIGN receptor) are significantly more susceptible to Streptococcus pneumoniae infection and fail to clear S. pneumoniae from the circulation. Marginal zone and peritoneal macrophages show impaired bacterial recognition associated with an inability to bind T-independent type 2 antigens such as dextran. Our work represents the first evidence for a protective in vivo role for a SIGN family molecule.
Collapse
Affiliation(s)
- Astrid Lanoue
- Laboratory of Molecular Biology, Medical Research Council, Hills Rd., Cambridge CB2 2QH, England, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Bartenschlager R, Frese M, Pietschmann T. Novel insights into hepatitis C virus replication and persistence. Adv Virus Res 2005; 63:71-180. [PMID: 15530561 DOI: 10.1016/s0065-3527(04)63002-8] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatitis C virus (HCV) is a small enveloped RNA virus that belongs to the family Flaviviridae. A hallmark of HCV is its high propensity to establish a persistent infection that in many cases leads to chronic liver disease. Molecular studies of the virus became possible with the first successful cloning of its genome in 1989. Since then, the genomic organization has been delineated, and viral proteins have been studied in some detail. In 1999, an efficient cell culture system became available that recapitulates the intracellular part of the HCV life cycle, thereby allowing detailed molecular studies of various aspects of viral RNA replication and persistence. This chapter attempts to summarize the current state of knowledge in these most actively worked on fields of HCV research.
Collapse
Affiliation(s)
- Ralf Bartenschlager
- Department of Molecular Virology, University of Heidelberg, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
182
|
Peyrefitte CN, Pastorino B, Grau GE, Lou J, Tolou H, Couissinier-Paris P. Dengue virus infection of human microvascular endothelial cells from different vascular beds promotes both common and specific functional changes. J Med Virol 2005; 78:229-42. [PMID: 16372301 DOI: 10.1002/jmv.20532] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dengue shock syndrome (DSS), the major life threatening outcome of severe dengue disease, which occurs in some patients in the course of dengue infection, is the consequence of plasma leakage in the microvascular territories. Data from clinical and in vitro studies suggest that an inadequate immunological response is partly responsible for the pathophysiology of DSS, but few is known concerning the consequences of direct infection of endothelial cells by dengue virus per se. In this study, an attempt was made to study the response of two microvascular human cell lines originating, respectively, from liver and dermis to infection by a dengue type 2 virus, by analyzing the virus-induced modulation of functional markers. It is shown that the two microvascular cell lines exhibit both common and specific behaviors upon infection. In particular, LSEC and HMEC-1 replicate efficiently the low-passage virus and respond to infection by over-producing inflammatory mediators involved in the cross talk with circulating immune cells. However, direct infection modulates differently the cell surface expression of molecules critically involved in the interactions between endothelial and inflammatory cells. ICAM-1 and HLA-I are up regulated as a consequence of infection in LSEC whereas direct infection results in downregulation of ICAM-1 in HMEC-1. The present results show that infection of human microvascular cells by unadapted dengue virus results in both common and specific activation patterns depending likely on the tissue origin of the cells, thus suggesting that endothelia from different territories may contribute differently to the pathophysiological events in the course of dengue infection.
Collapse
Affiliation(s)
- Christophe N Peyrefitte
- Unité de virologie tropicale, Institut de Médecine Tropicale du Service de Santé des Armées, Marseille, France
| | | | | | | | | | | |
Collapse
|
183
|
Nair MPN, Schwartz SA, Mahajan SD, Tsiao C, Chawda RP, Whitney R, Don Sykes BB, Hewitt R. Drug abuse and neuropathogenesis of HIV infection: role of DC-SIGN and IDO. J Neuroimmunol 2004; 157:56-60. [PMID: 15579280 DOI: 10.1016/j.jneuroim.2004.08.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 10/26/2022]
Abstract
Dendritic cells are the critical mediators of various immune responses and are the first line of defense against any infection including HIV. They play a major role in harboring HIV and the subsequent infection of T cells and passage of virus through the blood-brain barrier (BBB). The recently discovered DC-specific, CD4-independent HIV attachment receptor, DC-SIGN, and T-cell suppressing factor, indolamine 2,3-dioxygenase (IDO), are known to play a critical role in the immuno-neuropathogenesis of HIV infection. Since brain microvascular cells (BMVEC) express dendritic cell (DC)-specific C type ICAM-3 grabbing nonintegrin (DC-SIGN), it is possible that DC-SIGN may play a critical role in human immunodeficiency virus-type 1 (HIV-1) infection and migration of infected DC across BBB. Matrix metalloproteinases (MMPs) are proteolytic enzymes known to be responsible for maintenance, turnover and integrity of extracellular matrix. Our results show that cocaine upregulates IDO and DC-SIGN expression by DC. Further, cocaine upregulates DC-SIGN and MMPs in BMVEC supporting the hypothesis that cocaine causes membrane permeability facilitating endothelial transmigration of infected DC in to the CNS. Targeting DC-SIGN and IDO with specific monoclonal antibodies, inexpensive synthetic antagonists, antisense oligonucleotides and siRNA may lead to develop novel treatment strategies particularly in high-risk populations such as cocaine users.
Collapse
MESH Headings
- Anesthetics, Local/pharmacology
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cells, Cultured
- Cocaine/pharmacology
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Dioxygenases/genetics
- Dioxygenases/metabolism
- Dose-Response Relationship, Drug
- HIV Infections/complications
- HIV Infections/enzymology
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Matrix Metalloproteinases/genetics
- Matrix Metalloproteinases/metabolism
- Matrix Metalloproteinases, Membrane-Associated
- Metalloendopeptidases/genetics
- Metalloendopeptidases/metabolism
- RNA, Messenger/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Substance-Related Disorders/complications
Collapse
Affiliation(s)
- Madhavan P N Nair
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology and State University of New York and Buffalo General Hospital, Kaleida Health System 100 High Street, Buffalo, NY 14203, USA.
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Marzi A, Gramberg T, Simmons G, Möller P, Rennekamp AJ, Krumbiegel M, Geier M, Eisemann J, Turza N, Saunier B, Steinkasserer A, Becker S, Bates P, Hofmann H, Pöhlmann S. DC-SIGN and DC-SIGNR interact with the glycoprotein of Marburg virus and the S protein of severe acute respiratory syndrome coronavirus. J Virol 2004; 78:12090-5. [PMID: 15479853 PMCID: PMC523257 DOI: 10.1128/jvi.78.21.12090-12095.2004] [Citation(s) in RCA: 304] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lectins DC-SIGN and DC-SIGNR can augment viral infection; however, the range of pathogens interacting with these attachment factors is incompletely defined. Here we show that DC-SIGN and DC-SIGNR enhance infection mediated by the glycoprotein (GP) of Marburg virus (MARV) and the S protein of severe acute respiratory syndrome coronavirus and might promote viral dissemination. SIGNR1, a murine DC-SIGN homologue, also enhanced infection driven by MARV and Ebola virus GP and could be targeted to assess the role of attachment factors in filovirus infection in vivo.
Collapse
Affiliation(s)
- Andrea Marzi
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, Nikolaus-Fiebiger-Center, Glückstrasse 6, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Jeffers SA, Tusell SM, Gillim-Ross L, Hemmila EM, Achenbach JE, Babcock GJ, Thomas WD, Thackray LB, Young MD, Mason RJ, Ambrosino DM, Wentworth DE, Demartini JC, Holmes KV. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus. Proc Natl Acad Sci U S A 2004; 101:15748-53. [PMID: 15496474 PMCID: PMC524836 DOI: 10.1073/pnas.0403812101] [Citation(s) in RCA: 466] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Accepted: 09/23/2004] [Indexed: 01/03/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a receptor for SARS-CoV, the novel coronavirus that causes severe acute respiratory syndrome [Li, W. Moore, M. J., Vasilieva, N., Sui, J., Wong, S. K., Berne, M. A., Somasundaran, M., Sullivan, J. L., Luzuriaga, K., Greenough, T. C., et al. (2003) Nature 426, 450-454]. We have identified a different human cellular glycoprotein that can serve as an alternative receptor for SARS-CoV. A human lung cDNA library in vesicular stomatitis virus G pseudotyped retrovirus was transduced into Chinese hamster ovary cells, and the cells were sorted for binding of soluble SARS-CoV spike (S) glycoproteins, S(590) and S(1180). Clones of transduced cells that bound SARS-CoV S glycoprotein were inoculated with SARS-CoV, and increases in subgenomic viral RNA from 1-16 h or more were detected by multiplex RT-PCR in four cloned cell lines. Sequencing of the human lung cDNA inserts showed that each of the cloned cell lines contained cDNA that encoded human CD209L, a C-type lectin (also called L-SIGN). When the cDNA encoding CD209L from clone 2.27 was cloned and transfected into Chinese hamster ovary cells, the cells expressed human CD209L glycoprotein and became susceptible to infection with SARS-CoV. Immunohistochemistry showed that CD209L is expressed in human lung in type II alveolar cells and endothelial cells, both potential targets for SARS-CoV. Several other enveloped viruses including Ebola and Sindbis also use CD209L as a portal of entry, and HIV and hepatitis C virus can bind to CD209L on cell membranes but do not use it to mediate virus entry. Our data suggest that the large S glycoprotein of SARS-CoV may use both ACE2 and CD209L in virus infection and pathogenesis.
Collapse
Affiliation(s)
- Scott A Jeffers
- Department of Microbiology and Molecular Biology Program, University Colorado Health Sciences Center, 4200 East 9th Avenue, Denver, CO 80262, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Cormier EG, Durso RJ, Tsamis F, Boussemart L, Manix C, Olson WC, Gardner JP, Dragic T. L-SIGN (CD209L) and DC-SIGN (CD209) mediate transinfection of liver cells by hepatitis C virus. Proc Natl Acad Sci U S A 2004; 101:14067-72. [PMID: 15371595 PMCID: PMC521122 DOI: 10.1073/pnas.0405695101] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Target cell tropism of enveloped viruses is regulated by interactions between viral and cellular factors during transmission, dissemination, and replication within the host. Binding of viral envelope glycoproteins to specific cell-surface receptors determines susceptibility to viral entry. However, a number of cell-surface molecules bind viral envelope glycoproteins without mediating entry. Instead, they serve as capture receptors that disseminate viral particles to target organs or susceptible cells. We and others recently demonstrated that the C type lectins L-SIGN and DC-SIGN capture hepatitis C virus (HCV) by specific binding to envelope glycoprotein E2. In this study, we use an entry assay to demonstrate that HCV pseudoviruses captured by L-SIGN+ or DC-SIGN+ cells efficiently transinfect adjacent human liver cells. Virus capture and transinfection require internalization of the SIGN-HCV pseudovirus complex. In vivo, L-SIGN is largely expressed on endothelial cells in liver sinusoids, whereas DC-SIGN is expressed on dendritic cells. Capture of circulating HCV particles by these SIGN+ cells may facilitate virus infection of proximal hepatocytes and lymphocyte subpopulations and may be essential for the establishment of persistent infection.
Collapse
Affiliation(s)
- Emmanuel G Cormier
- Microbiology and Immunology Department, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Ludwig IS, Lekkerkerker AN, Depla E, Bosman F, Musters RJP, Depraetere S, van Kooyk Y, Geijtenbeek TBH. Hepatitis C virus targets DC-SIGN and L-SIGN to escape lysosomal degradation. J Virol 2004; 78:8322-32. [PMID: 15254204 PMCID: PMC446128 DOI: 10.1128/jvi.78.15.8322-8332.2004] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) is a major health problem. However, the mechanism of hepatocyte infection is largely unknown. We demonstrate that the dendritic cell (DC)-specific C-type lectin DC-SIGN and its liver-expressed homologue L-SIGN/DC-SIGNR are important receptors for HCV envelope glycoproteins E1 and E2. Mutagenesis analyses demonstrated that both HCV E1 and E2 bind the same binding site on DC-SIGN as the pathogens human immunodeficiency virus type 1 (HIV-1) and mycobacteria, which is distinct from the cellular ligand ICAM-3. HCV virus-like particles are efficiently captured and internalized by DCs through binding of DC-SIGN. Antibodies against DC-SIGN specifically block HCV capture by both immature and mature DCs, demonstrating that DC-SIGN is the major receptor on DCs. Interestingly, internalized HCV virus-like particles were targeted to nonlysosomal compartments within immature DCs, where they are protected from lysosomal degradation in a manner similar to that demonstrated for HIV-1. Lewis X antigen, another ligand of DC-SIGN, was internalized to lysosomes, demonstrating that the internalization pathway of DC-SIGN-captured ligands may depend on the structure of the ligand. Our results suggest that HCV may target DC-SIGN to "hide" within DCs and facilitate viral dissemination. L-SIGN, expressed by THP-1 cells, internalized HCV particles into similar nonlysosomal compartments, suggesting that L-SIGN on liver sinusoidal endothelial cells may capture HCV from blood and transmit it to hepatocytes, the primary target for HCV. We therefore conclude that both DCs and liver sinusoidal endothelial cells may act as reservoirs for HCV and that the C-type lectins DC-SIGN and L-SIGN, as important HCV receptors, may represent a molecular target for clinical intervention in HCV infection.
Collapse
Affiliation(s)
- Irene S Ludwig
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Biggins JE, Yu Kimata MT, Kimata JT. Domains of macaque DC-SIGN essential for capture and transfer of simian immunodeficiency virus. Virology 2004; 324:194-203. [PMID: 15183066 DOI: 10.1016/j.virol.2004.03.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Revised: 11/25/2003] [Accepted: 03/22/2004] [Indexed: 11/19/2022]
Abstract
The C-type lectin DC-SIGN mediates the capture and transfer of simian immunodeficiency virus (SIV) from macaque dendritic cells (DCs) to permissive T-cells. To further identify the determinants in macaque DC-SIGN required for capture and transfer of virus, we created mutants containing deletions or point mutations in the extracellular domains, and tested their ability to capture and transmit SIV. We found that SIV bound to the carbohydrate recognition domain (CRD) of macaque DC-SIGN via the envelope protein. In addition, deleting the C-terminal half of the CRD, or mutating amino acids within this region that contact Ca(2+) or mannose, disrupted virion capture activity. However, an N-terminal CRD deletion mutant was capable of binding SIV, indicating that this region was not necessary for binding. Finally, deletion of the neck domain also reduced the capacity for macaque DC-SIGN to capture SIV. Interestingly, ICAM-3, the cellular ligand for DC-SIGN, did not bind to any of the DC-SIGN mutants, including mutants with amino acid changes in the N-terminal region of the CRD. These data suggest that the binding sites for SIV and ICAM-3 may be distinct but overlapping. Together, the data demonstrate the importance of both the neck and the CRD of macaque DC-SIGN for efficient capture of SIV and binding to ICAM-3.
Collapse
Affiliation(s)
- Julia E Biggins
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
189
|
Lozach PY, Amara A, Bartosch B, Virelizier JL, Arenzana-Seisdedos F, Cosset FL, Altmeyer R. C-type Lectins L-SIGN and DC-SIGN Capture and Transmit Infectious Hepatitis C Virus Pseudotype Particles. J Biol Chem 2004; 279:32035-45. [PMID: 15166245 DOI: 10.1074/jbc.m402296200] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The molecular mechanisms involved in the hepatic tropism of hepatitis C virus (HCV) have not been identified. We have shown previously that liver-expressed C-type lectins L-SIGN and DC-SIGN bind the HCV E2 glycoprotein with high affinity (Lozach, P. Y., Lortat-Jacob, H., de Lacroix de Lavalette, A., Staropoli, I., Foung, S., Amara, A., Houles, C., Fieschi, F., Schwartz, O., Virelizier, J. L., Arenzana-Seisdedos, F., and Altmeyer, R. (2003) J. Biol. Chem. 278, 20358-20366). To analyze the functional relevance of this interaction, we generated pseudotyped lentivirus particles presenting HCV glycoproteins E1 and E2 at the virion surface (HCV-pp). High mannose N-glycans are present on E1 and, to a lesser extent, on E2 proteins of mature infectious HCV-pp. Such particles bind to both L-SIGN and DC-SIGN, but they cannot use these receptors for entry into cells. However, infectious virus is transmitted efficiently when permissive Huh-7 cells are cocultured with HCV-pp bound to L-SIGN or to DC-SIGN-positive cell lines. HCV-pp transmission via L-SIGN or DC-SIGN is inhibited by characteristic inhibitors such as the calcium chelator EGTA and monoclonal antibodies directed against lectin carbohydrate recognition domains of both lectins. In support of the biological relevance of this phenomenon, dendritic cells expressing endogenous DC-SIGN transmitted HCV-pp with high efficiency in a DC-SIGN-dependent manner. Our results support the hypothesis that C-type lectins such as the liver sinusoidal endothelial cell-expressed L-SIGN could act as a capture receptor for HCV in the liver and transmit infectious virions to neighboring hepatocytes.
Collapse
Affiliation(s)
- Pierre-Yves Lozach
- Unité d'Immunologie Virale, Institut Pasteur, 28, rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
190
|
Bobardt MD, Salmon P, Wang L, Esko JD, Gabuzda D, Fiala M, Trono D, Van der Schueren B, David G, Gallay PA. Contribution of proteoglycans to human immunodeficiency virus type 1 brain invasion. J Virol 2004; 78:6567-84. [PMID: 15163749 PMCID: PMC416544 DOI: 10.1128/jvi.78.12.6567-6584.2004] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
As a neurotropic virus, human immunodeficiency virus type 1 (HIV-1) invades the brain and causes severe neuronal, astrocyte, and myelin damage in AIDS patients. To gain access to the brain, HIV-1 must migrate through brain microvascular endothelial cells (BMECs), which compose the blood-brain barrier (BBB). Given that BMECs lack the entry receptor CD4, HIV-1 must use receptors distinct from CD4 to enter these cells. We previously reported that cell surface proteoglycans serve as major HIV-1 receptors on primary human endothelial cells. In this study, we examined whether proteoglycans also impact cell-free HIV-1 invasion of the brain. Using an artificial BBB transmigration assay, we found that both heparan and chondroitin sulfate proteoglycans (HSPGs and CSPGs, respectively) are abundantly expressed on primary BMECs and promote HIV-1 attachment and entry. In contrast, the classical entry receptors, CXCR4 and CCR5, only moderately enhanced these processes. HSPGs and CSPGs captured HIV-1 in a gp120-dependent manner. However, no correlation between coreceptor usage and transmigration was identified. Furthermore, brain-derived viruses did not transmigrate more efficiently than lymphoid-derived viruses, suggesting that the ability of HIV-1 to replicate in the brain does not correlate with its capacity to migrate through the BBB as cell-free virus. Given that HIV-1-proteoglycan interactions are based on electrostatic contacts between basic residues in gp120 and sulfate groups in proteoglycans, HIV-1 may exploit these interactions to rapidly enter and migrate through the BBB to invade the brain.
Collapse
Affiliation(s)
- Michael D Bobardt
- Department of Immunology, IMM-9, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Barat C, Gervais P, Tremblay MJ. Engagement of ICAM-3 provides a costimulatory signal for human immunodeficiency virus type 1 replication in both activated and quiescent CD4+ T lymphocytes: implications for virus pathogenesis. J Virol 2004; 78:6692-7. [PMID: 15163761 PMCID: PMC416497 DOI: 10.1128/jvi.78.12.6692-6697.2004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) replication is regulated by several extracellular signals. We demonstrate that intercellular adhesion molecule 3 (ICAM-3) acts as a costimulating molecule to increase HIV-1 transcription and viral production, a process allowing productive infection of quiescent CD4+ T lymphocytes. The present work suggests an important role for ICAM-3 in HIV-1 replication.
Collapse
Affiliation(s)
- Corinne Barat
- Laboratory of Human Immuno-Retrovirology, Research Center in Infectious Diseases, RC709, CHUL Research Center, 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2
| | | | | |
Collapse
|
192
|
Wu L, Martin TD, Han YC, Breun SKJ, KewalRamani VN. Trans-dominant cellular inhibition of DC-SIGN-mediated HIV-1 transmission. Retrovirology 2004; 1:14. [PMID: 15222882 PMCID: PMC446230 DOI: 10.1186/1742-4690-1-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2004] [Accepted: 06/28/2004] [Indexed: 11/20/2022] Open
Abstract
Background Dendritic cell (DC) transmission of human immunodeficiency virus (HIV) to CD4+ T cells occurs across a point of cell-cell contact referred to as the infectious synapse. The relationship between the infectious synapse and the classically defined immunological synapse is not currently understood. We have recently demonstrated that human B cells expressing exogenous DC-SIGN, DC-specific intercellular adhesion molecule-3 (ICAM-3)-grabbing nonintegrin, efficiently transmit captured HIV type 1 (HIV-1) to CD4+ T cells. K562, another human cell line of hematopoietic origin that has been extensively used in functional analyses of DC-SIGN and related molecules, lacks the principal molecules involved in the formation of immunological synaptic junctions, namely major histocompatibility complex (MHC) class II molecules and leukocyte function-associated antigen-1 (LFA-1). We thus examined whether K562 erythroleukemic cells could recapitulate efficient DC-SIGN-mediated HIV-1 transmission (DMHT). Results Here we demonstrate that DMHT requires cell-cell contact. Despite similar expression of functional DC-SIGN, K562/DC-SIGN cells were inefficient in the transmission of HIV-1 to CD4+ T cells when compared with Raji/DC-SIGN cells. Expression of MHC class II molecules or LFA-1 on K562/DC-SIGN cells was insufficient to rescue HIV-1 transmission efficiency. Strikingly, we observed that co-culture of K562 cells with Raji/DC-SIGN cells impaired DMHT to CD4+ T cells. The K562 cell inhibition of transmission was not directly exerted on the CD4+ T cell targets and required contact between K562 and Raji/DC-SIGN cells. Conclusions DMHT is cell type dependent and requires cell-cell contact. We also find that the cellular milieu can negatively regulate DC-SIGN transmission of HIV-1 in trans.
Collapse
Affiliation(s)
- Li Wu
- Model Development Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Thomas D Martin
- Model Development Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Yoon-Chi Han
- Model Development Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Sabine KJ Breun
- Model Development Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Vineet N KewalRamani
- Model Development Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| |
Collapse
|
193
|
Bounou S, Giguere JF, Cantin R, Gilbert C, Imbeault M, Martin G, Tremblay MJ. The importance of virus-associated host ICAM-1 in human immunodeficiency virus type 1 dissemination depends on the cellular context. FASEB J 2004; 18:1294-6. [PMID: 15208262 DOI: 10.1096/fj.04-1755fje] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The primary objective of this study was to define whether the nature of virion-bound host cell membrane proteins influenced the process of human immunodeficiency virus 1 (HIV-1) capture and transmission. We pulsed cells of monocytoid lineage (established and primary) and CD4-negative epithelial cells transiently expressing DC-SIGN or LFA-1 with isogenic HIV-1 particles either devoid or bearing host-derived ICAM-1 or ICAM-3 before incubation with an indicator cell line. To our surprise, the ICAM-1/LFA-1 association was a more efficient transmission factor than the combined gp120/DC-SIGN and ICAM-3/DC-SIGN interactions. The involvement of the association between virus-bound ICAM-1 and its natural ligand LFA-1 in virus binding and carriage was confirmed when using more physiological cellular targets, i.e., human lymphoid tissues cultured ex vivo. However, the contribution of virus-anchored host ICAM-1 to the process of retention and transmission of HIV-1 could not be confirmed when using primary human cells of macrophage/dendritic lineage as transmitter cells and autologous CD4+ T lymphocytes as targets. Altogether these data underscore the complexity of factors participating in virus-cell contact and efficient dissemination of HIV-1 to target cells.
Collapse
Affiliation(s)
- Salim Bounou
- Research Center in Infectious Diseases, CHUL Research Center, and Faculty of Medicine, Laval University, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
194
|
Van Liempt E, Imberty A, Bank CMC, Van Vliet SJ, Van Kooyk Y, Geijtenbeek TBH, Van Die I. Molecular basis of the differences in binding properties of the highly related C-type lectins DC-SIGN and L-SIGN to Lewis X trisaccharide and Schistosoma mansoni egg antigens. J Biol Chem 2004; 279:33161-7. [PMID: 15184372 DOI: 10.1074/jbc.m404988200] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The dendritic cell-specific C-type lectin DC-SIGN functions as a pathogen receptor that recognizes Schistosoma mansoni egg antigens through its major glycan epitope Galbeta1,4(Fucalpha1,3)GlcNAc (Lex). Here we report that L-SIGN, a highly related homologue of DC-SIGN found on liver sinusoidal endothelial cells, binds to S. mansoni egg antigens but not to the Lex epitope. L-SIGN does bind the Lewis antigens Lea, Leb, and Ley, similar as DC-SIGN. A specific mutation in the carbohydrate recognition domain of DC-SIGN (V351G) abrogates binding to all Lewis antigens. In L-SIGN Ser363 is present at the corresponding position of Val351 in DC-SIGN. Replacement of this Ser into Val resulted in a "gain of function" L-SIGN mutant that binds to Lex, and shows increased binding to the other Lewis antigens. These data indicate that Val351 is important for the fucose specificity of DC-SIGN. Molecular modeling and docking of the different Lewis antigens in the carbohydrate recognition domains of L-SIGN, DC-SIGN, and their mutant forms, demonstrate that Val351 in DC-SIGN creates a hydrophobic pocket that strongly interacts with the Fucalpha1,3/4-GlcNAc moiety of the Lewis antigens. The equivalent amino acid residue Ser363 in L-SIGN creates a hydrophilic pocket that prevents interaction with Fucalpha1,3-GlcNAc in Lex but supports interactions with the Fucalpha1,4-GlcNAc moiety in Lea and Leb antigens. These data demonstrate for the first time that DC-SIGN and L-SIGN differ in their carbohydrate binding profiles and will contribute to our understanding of the functional roles of these C-type lectin receptors, both in recognition of pathogen and self-glycan antigens.
Collapse
Affiliation(s)
- Ellis Van Liempt
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
195
|
Engering A, van Vliet SJ, Hebeda K, Jackson DG, Prevo R, Singh SK, Geijtenbeek TBH, van Krieken H, van Kooyk Y. Dynamic populations of dendritic cell-specific ICAM-3 grabbing nonintegrin-positive immature dendritic cells and liver/lymph node-specific ICAM-3 grabbing nonintegrin-positive endothelial cells in the outer zones of the paracortex of human lymph nodes. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1587-95. [PMID: 15111305 PMCID: PMC1615649 DOI: 10.1016/s0002-9440(10)63717-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the paracortex of lymph nodes, cellular immune responses are generated against antigens captured in peripheral tissues by dendritic cells (DCs). DC-SIGN (dendritic cell-specific ICAM-3 grabbing nonintegrin), a C-type lectin exclusively expressed by DCs, functions as an antigen receptor as well as an adhesion receptor. A functional homologue of DC-SIGN, L-SIGN (liver/lymph node-SIGN, also called DC-SIGN-related), is expressed by liver sinus endothelial cells. In lymph nodes, both DC-SIGN and L-SIGN are expressed. In this study, we analyzed the distribution of these two SIGN molecules in detail in both normal and immunoreactive lymph nodes. DC-SIGN is expressed by mature DCs in paracortical areas and in addition by DCs with an immature phenotype in the outer zones of the paracortex. L-SIGN expression was also detected in the outer zones on sinus endothelial cells characterized by their expression of the lymphatic endothelial markers LYVE-1 and CLEVER-1. During both cellular and humoral immune responses changes in the amount of DC-SIGN+ immature and mature DCs and L-SIGN+ endothelial cells were observed, indicating that the influx or proliferation of these cells is dynamically regulated.
Collapse
Affiliation(s)
- Anneke Engering
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Nisole S, Saïb A. Early steps of retrovirus replicative cycle. Retrovirology 2004; 1:9. [PMID: 15169567 PMCID: PMC421752 DOI: 10.1186/1742-4690-1-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2004] [Accepted: 05/14/2004] [Indexed: 12/28/2022] Open
Abstract
During the last two decades, the profusion of HIV research due to the urge to identify new therapeutic targets has led to a wealth of information on the retroviral replication cycle. However, while the late stages of the retrovirus life cycle, consisting of virus replication and egress, have been partly unraveled, the early steps remain largely enigmatic. These early steps consist of a long and perilous journey from the cell surface to the nucleus where the proviral DNA integrates into the host genome. Retroviral particles must bind specifically to their target cells, cross the plasma membrane, reverse-transcribe their RNA genome, while uncoating the cores, find their way to the nuclear membrane and penetrate into the nucleus to finally dock and integrate into the cellular genome. Along this journey, retroviruses hijack the cellular machinery, while at the same time counteracting cellular defenses. Elucidating these mechanisms and identifying which cellular factors are exploited by the retroviruses and which hinder their life cycle, will certainly lead to the discovery of new ways to inhibit viral replication and to improve retroviral vectors for gene transfer. Finally, as proven by many examples in the past, progresses in retrovirology will undoubtedly also provide some priceless insights into cell biology.
Collapse
Affiliation(s)
- Sébastien Nisole
- Division of Virology, National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| | - Ali Saïb
- CNRS UPR9051, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75475 Paris cedex 10, France
| |
Collapse
|
197
|
Takada A, Fujioka K, Tsuiji M, Morikawa A, Higashi N, Ebihara H, Kobasa D, Feldmann H, Irimura T, Kawaoka Y. Human macrophage C-type lectin specific for galactose and N-acetylgalactosamine promotes filovirus entry. J Virol 2004; 78:2943-7. [PMID: 14990712 PMCID: PMC353724 DOI: 10.1128/jvi.78.6.2943-2947.2004] [Citation(s) in RCA: 208] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Filoviruses cause lethal hemorrhagic disease in humans and nonhuman primates. An initial target of filovirus infection is the mononuclear phagocytic cell. Calcium-dependent (C-type) lectins such as dendritic cell- or liver/lymph node-specific ICAM-3 grabbing nonintegrin (DC-SIGN or L-SIGN, respectively), as well as the hepatic asialoglycoprotein receptor, bind to Ebola or Marburg virus glycoprotein (GP) and enhance the infectivity of these viruses in vitro. Here, we demonstrate that a recently identified human macrophage galactose- and N-acetylgalactosamine-specific C-type lectin (hMGL), whose ligand specificity differs from DC-SIGN and L-SIGN, also enhances the infectivity of filoviruses. This enhancement was substantially weaker for the Reston and Marburg viruses than for the highly pathogenic Zaire virus. We also show that the heavily glycosylated, mucin-like domain on the filovirus GP is required for efficient interaction with this lectin. Furthermore, hMGL, like DC-SIGN and L-SIGN, is present on cells known to be major targets of filoviruses (i.e., macrophages and dendritic cells), suggesting a role for these C-type lectins in viral replication in vivo. We propose that filoviruses use different C-type lectins to gain cellular entry, depending on the cell type, and promote efficient viral replication.
Collapse
Affiliation(s)
- Ayato Takada
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Abstract
DC-SIGN, a dendritic Cell-specific adhesion receptor and a type II transmembrane mannose-binding C-type lectin, is very important in the function of DC, both in mediating naive T cell interactions through ICAM-3 and as a rolling receptor that mediates the DC-specific ICAM-2-dependent migration processes. It can be used by viral and bacterial pathogens including Human Immunodeficiency Virus (HIV), HCV, Ebola Virus, CMV and Mycobacterium tuberculosis to facilitate infection. Both DC-SIGN and DC-SIGNR can act either in cis, by concentrating virus on target cells, or in trans, by transmission of bound virus to a target cell expressing appropropriate entry receptors. Recent work showed that DC-SIGN are high-affinity binding receptors for HCV. Besides playing a role in entry into DC, HCV E2 interaction with DC-SIGN might also be detrimental for the interaction of DC with T cells during antigen presentation. The clinical strategies that target DC-SIGN may be successful in restricting HCV dissemination and pathogenesis as well as directing the migration of DCs to manipulate appropriate immune responses in autoimmunity and tumorigenic situations.
Collapse
Affiliation(s)
- Zhi-Hua Feng
- The Center of Diagnosis and Treatment for Infectious Diseases of PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China.
| | | | | | | | | |
Collapse
|
199
|
Geijtenbeek TBH, van Vliet SJ, Engering A, 't Hart BA, van Kooyk Y. Self- and nonself-recognition by C-type lectins on dendritic cells. Annu Rev Immunol 2004; 22:33-54. [PMID: 15032573 DOI: 10.1146/annurev.immunol.22.012703.104558] [Citation(s) in RCA: 373] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) are highly efficient antigen-presenting cells (APCs) that collect antigen in body tissues and transport them to draining lymph nodes. Antigenic peptides are loaded onto major histocompatibility complex (MHC) molecules for presentation to naive T cells, resulting in the induction of cellular and humoral immune responses. DCs take up antigen through phagocytosis, pinocytosis, and endocytosis via different groups of receptor families, such as Fc receptors for antigen-antibody complexes, C-type lectin receptors (CLRs) for glycoproteins, and pattern recognition receptors, such as Toll-like receptors (TLRs), for microbial antigens. Uptake of antigen by CLRs leads to presentation of antigens on MHC class I and II molecules. DCs are well equipped to distinguish between self- and nonself-antigens by the variable expression of cell-surface receptors such as CLRs and TLRs. In the steady state, DCs are not immunologically quiescent but use their antigen-handling capacities to maintain peripheral tolerance. DCs are continuously sampling and presenting self- and harmless environmental proteins to silence immune activation. Uptake of self-components in the intestine and airways are good examples of sites where continuous presentation of self- and foreign antigens occurs without immune activation. In contrast, efficient antigen-specific immune activation occurs upon encounter of DCs with nonself-pathogens. Recognition of pathogens by DCs triggers specific receptors such as TLRs that result in DC maturation and subsequently immune activation. Here we discuss the concept that cross talk between TLRs and CLRs, differentially expressed by subsets of DCs, accounts for the different pathways to peripheral tolerance, such as deletion and suppression, and immune activation.
Collapse
Affiliation(s)
- Teunis B H Geijtenbeek
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center Amsterdam, 1081 BT Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
200
|
Jochimsen F, Gruening W, Arnould T, Segal MS, Kruskall MS, Colgrove R, Walz G. Thrombotic microangiopathy associated with unusual viral sequences in HIV-1-positive patients. Nephrol Dial Transplant 2004; 19:1129-35. [PMID: 14993479 DOI: 10.1093/ndt/gfh085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Thrombotic microangiopathy (TMA) is a rare disorder caused by endothelial cell damage. TMA has been associated with the human immunodeficiency virus 1 (HIV-1) infection, yet only a minority of all HIV-1 patients develops TMA. Since HIV-1 has been shown to interact with endothelial cells, we investigated whether certain mutations in the HIV-1 envelope protein are associated with the development of TMA in HIV-1-infected patients. METHODS Plasma was obtained from nine HIV-1-positive patients with TMA. Viral loads were determined from the samples and compared with the clinical data. Viral envelope protein sequences from the regions known to be responsible for viral tropism were isolated, sequenced and compared with known HIV-1 isolates. The isolates were expressed as synthetic fusion proteins; binding of these fusion proteins to CD4+ cells as well as to endothelial cell lines was investigated. RESULTS The viral loads in patients with HIV/TMA were highly variable with no correlation to the clinical status. Most patients carried macrophage-tropic viral envelope protein sequences and an unusual insertion was found in the V2 variable region. The isolates showed increased CD4 binding, but a direct binding to endothelial cells was not observed. CONCLUSIONS Although TMA is generally diagnosed in patients with advanced HIV-1 infection, viral loads per se were not predictive of TMA in this study. While a direct interaction with endothelial cells was not detectable, specific viral envelope mutations were found in a region known to influence viral tropism. Hence, viral-specific factors might contribute to the pathogenesis of HIV-associated TMA.
Collapse
Affiliation(s)
- Friederike Jochimsen
- Beth Israel Deaconess Medical Center, Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|