51
|
Serrano-Gómez D, Sierra-Filardi E, Martínez-Nuñez RT, Caparrós E, Delgado R, Muñoz-Fernández MA, Abad MA, Jimenez-Barbero J, Leal M, Corbí AL. Structural requirements for multimerization of the pathogen receptor dendritic cell-specific ICAM3-grabbing non-integrin (CD209) on the cell surface. J Biol Chem 2007; 283:3889-903. [PMID: 18073208 DOI: 10.1074/jbc.m706004200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The myeloid C-type lectin dendritic cell-specific ICAM3-grabbing non-integrin (DC-SIGN, CD209) recognizes oligosaccharide ligands on clinically relevant pathogens (HIV, Mycobacterium, and Aspergillus). Alternative splicing and genomic polymorphism generate DC-SIGN mRNA variants, which have been detected at sites of pathogen entrance and transmission. We present evidence that DC-SIGN neck variants are expressed on dendritic and myeloid cells at the RNA and protein levels. Structural analysis revealed that multimerization of DC-SIGN within a cellular context depends on the lectin domain and the number and arrangement of the repeats within the neck region, whose glycosylation negatively affects oligomer formation. Naturally occurring DC-SIGN neck variants differ in multimerization competence in the cell membrane, exhibit altered sugar binding ability, and retain pathogen-interacting capacity, implying that pathogen-induced cluster formation predominates over the basal multimerization capability. Analysis of DC-SIGN neck polymorphisms indicated that the number of allelic variants is higher than previously thought and that multimerization of the prototypic molecule is modulated in the presence of allelic variants with a different neck structure. Our results demonstrate that the presence of allelic variants or a high level of expression of neck domain splicing isoforms might influence the presence and stability of DC-SIGN multimers on the cell surface, thus providing a molecular explanation for the correlation between DC-SIGN polymorphisms and altered susceptibility to HIV-1 and other pathogens.
Collapse
Affiliation(s)
- Diego Serrano-Gómez
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Redline RW. Villitis of unknown etiology: noninfectious chronic villitis in the placenta. Hum Pathol 2007; 38:1439-46. [PMID: 17889674 DOI: 10.1016/j.humpath.2007.05.025] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 05/30/2007] [Indexed: 11/22/2022]
Abstract
Villitis of unknown etiology (VUE) is an important pattern of placental injury occurring predominantly in term placentas. Although overlapping with infectious villitis, its clinical and histologic characteristics are distinct. It is a common lesion, affecting 5% to 15% of all placentas. When low-grade lesions affecting less than 10 villi per focus are excluded, VUE is an important cause of intrauterine growth restriction and recurrent reproductive loss. Involvement of large fetal vessels in the placenta (obliterative fetal vasculopathy) in cases of VUE is a strong risk factor for neonatal encephalopathy and cerebral palsy. Although the etiology of the eliciting antigen is unknown, many other characteristics of the immune response have been clarified. VUE is caused by maternal T lymphocytes, predominantly CD8-positive, that inappropriately gain access to the villous stroma. Fetal antigen-presenting cells (Hofbauer cells) expand and are induced to express class II major histocompatibility complex molecules. Maternal monocyte-macrophages in the perivillous space likely amplify the immune response. Although much speculation exists that VUE represents a host-versus-graft reaction analogous to transplant rejection, other eliciting antigens have not been excluded. Irrespective of target antigen or antigens, the pathophysiologic implications of having activated maternal lymphocytes within vascularized fetal tissues are not trivial.
Collapse
Affiliation(s)
- Raymond W Redline
- Department of Pathology, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
53
|
Rathore A, Chatterjee A, Sivarama P, Yamamoto N, Dhole TN. Role of homozygous DC-SIGNR 5/5 tandem repeat polymorphism in HIV-1 exposed seronegative North Indian individuals. J Clin Immunol 2007; 28:50-7. [PMID: 17876530 PMCID: PMC7086598 DOI: 10.1007/s10875-007-9131-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2007] [Accepted: 08/21/2007] [Indexed: 12/05/2022]
Abstract
Despite multiple sexual exposures to HIV-1 virus, some individuals remain HIV-1 seronegative. Although several genetic factors have been related to HIV-1 resistance, the homozygosity for a mutation in CCR5 gene (the 32-bp deletion, i.e., CCR5-Delta32 allele) is presently considered the most relevant one. The C-type lectins, DC-SIGN (present on dendritic cells and macrophages) and DC-SIGNR (present on endothelial cells in liver and lymph nodes) efficiently bind and transmit HIV-1 to susceptible cell in trans, thereby augmenting the infection. A potential association of the DC-SIGN and DC-SIGNR neck domain repeat polymorphism and risk of HIV-1 infection is currently under debate. To determine the influence of host genetic factors on HIV-1 resistance, we conducted genetic risk association study in HIV-1-exposed seronegative (n = 47) individuals, HIV-1 seronegative (n = 262) healthy control, and HIV-1-infected seropositive patients (n = 168) for polymorphism in neck domain of DC-SIGN and DC-SIGNR genes. The DC-SIGN and DC-SIGNR genotypes were identified by polymerase chain reaction method in DNA extracted from peripheral blood and confirmed by sequencing. Fisher exact or χ2 test was used for static analysis. DC-SIGN genotype and allele distribution was fairly similar in HIV-1-exposed seronegative, HIV-1 seropositive, and HIV-1 seronegative control. There was no statistical significance in the differences in the distribution of DC-SIGN genotypes. A total of 13 genotypes were found in DC-SIGNR neck repeat region polymorphism. Among all the genotypes, only 5/5 homozygous showed significant reduced risk of HIV-1 infection in HIV-1-exposed seronegative individuals (p = 0.009). A unique genotype 8/5 heterozygous was also found in HIV-1 seropositive individual, which is not reported elsewhere.
Collapse
Affiliation(s)
- Anurag Rathore
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebarelli Road, Lucknow, 226014 India
| | - Animesh Chatterjee
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebarelli Road, Lucknow, 226014 India
| | - P. Sivarama
- AIDS Counseling and Treatment Center, Belgaum, India
| | - Naohiko Yamamoto
- Department of International Health, Nagoya University School of Medicine, Nagoya, Japan
| | - Tapan N. Dhole
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebarelli Road, Lucknow, 226014 India
| |
Collapse
|
54
|
Bai H, Zhang L, Ma L, Dou XG, Feng GH, Zhao GZ. Relationship of hepatitis B virus infection of placental barrier and hepatitis B virus intra-uterine transmission mechanism. World J Gastroenterol 2007; 13:3625-30. [PMID: 17659715 PMCID: PMC4146804 DOI: 10.3748/wjg.v13.i26.3625] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the mechanism of intra-uterine transmission, the HBV infection status of placental tissue and in vitro cultured placental trophoblastic cells was tested through in vivo and in vitro experiments.
METHODS: A variety of methods, such as ELISA, RT-PCR, IHC staining and immunofluorescent staining were employed to test the HBV marker positive pregnant women's placenta and in vitro cultured placental trophoblastic cells.
RESULTS: The HBV DNA levels in pregnant women's serum and fetal cord blood were correlated. For those cord blood samples positive for HBV DNA, their maternal blood levels of HBV DNA were at a high level. The HBsAg IHC staining positive cells could be seen in the placental tissues and the presence of HBV DNA detected. After co-incubating the trophoblastic cells and HBV DNA positive serum in vitro, the expressions of both HBsAg and HBV DNA could be detected.
CONCLUSION: The mechanism of HBV intra-uterine infection may be due to that HBV breaches the placental barrier and infects the fetus.
Collapse
Affiliation(s)
- Han Bai
- Department of Infectious Diseases, Shengjing Hospital Affiliated to CMU, Shenyang 110004, Liaoning Province, China
| | | | | | | | | | | |
Collapse
|
55
|
Barreiro LB, Neyrolles O, Babb CL, van Helden PD, Gicquel B, Hoal EG, Quintana-Murci L. Length variation of DC-SIGN and L-SIGN neck-region has no impact on tuberculosis susceptibility. Hum Immunol 2007; 68:106-12. [PMID: 17321900 PMCID: PMC7132702 DOI: 10.1016/j.humimm.2006.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 10/12/2006] [Accepted: 10/31/2006] [Indexed: 11/01/2022]
Abstract
The C-type lectins DC-SIGN and L-SIGN are important pathogen-recognition receptors of the human innate immune system. Both lectins have been shown to interact with a vast range of infectious agents, including Mycobacterium tuberculosis, the etiologic agent of tuberculosis in humans. In addition, DC-SIGN and L-SIGN possess a neck region, made up of a variable number of 23 amino acid tandem repeats, which plays a crucial role in the tetramerization of these proteins and support of the carbohydrate recognition domain. The length of the neck region, which shows variable levels of polymorphism, can critically influence the pathogen binding properties of these two receptors. We therefore investigated the impact of the DC-SIGN and L-SIGN neck-region length variation on the outcome of tuberculosis by screening this polymorphism in a large cohort of Coloured South African origin. The analyses of 711 individuals, including 351 tuberculosis patients and 360 healthy controls, revealed that none of the DC-SIGN and L-SIGN neck-region variants or genotypes seems to influence the individual susceptibility to develop tuberculosis.
Collapse
Affiliation(s)
- Luis B Barreiro
- CNRS FRE2849, Unit of Human Evolutionary Genetics, Institut Pasteur, Paris, France
| | | | | | | | | | | | | |
Collapse
|
56
|
Roulet V, Satie AP, Ruffault A, Le Tortorec A, Denis H, Guist'hau O, Patard JJ, Rioux-Leclerq N, Gicquel J, Jégou B, Dejucq-Rainsford N. Susceptibility of human testis to human immunodeficiency virus-1 infection in situ and in vitro. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:2094-103. [PMID: 17148672 PMCID: PMC1762481 DOI: 10.2353/ajpath.2006.060191] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Semen represents the main vector for human immunodeficiency virus (HIV) dissemination worldwide and has been shown to harbor replication-competent virus despite otherwise effective highly active anti-retroviral therapy, which achieves undetectable viral load in plasma. Despite this, the origin of seminal HIV particles remains unclear, as does the question of whether the male genital tract organs contribute virus to semen. Here we investigated the presence of HIV receptors within the human testis using immunohistochemistry and quantitative real-time polymerase chain reaction. We also analyzed the infectivity of a dual tropic HIV-1 strain in an organotypic culture, as well as the impact of viral exposure on testosterone production. Our study establishes that CXCR4+, CCR5+, CD4+, and DC-SIGN+ cells are present within the interstitial tissue of human testis and that these molecules persist throughout our organotypic culture. Our data also reveal that the human testis is permissive to HIV-1 and supports productive infection, leaving testosterone production apparently unaffected. Infected cells appeared to be testicular macrophages located within the interstitial tissue. That the testis itself represents a potential source of virus in semen could play a role in preventing viral eradication from semen because this organ constitutes a pharmacological sanctuary for many current antiretrovirals.
Collapse
Affiliation(s)
- Vanessa Roulet
- INSERM U625-GERHM, Campus Scientifique de Beaulieu, Avenue du Général Leclerc, 35 042 Rennes Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Greene W, Kuhne K, Ye F, Chen J, Zhou F, Lei X, Gao SJ. Molecular biology of KSHV in relation to AIDS-associated oncogenesis. Cancer Treat Res 2007; 133:69-127. [PMID: 17672038 PMCID: PMC2798888 DOI: 10.1007/978-0-387-46816-7_3] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
KSHV has been established as the causative agent of KS, PEL, and MCD, malignancies occurring more frequently in AIDS patients. The aggressive nature of KSHV in the context of HIV infection suggests that interactions between the two viruses enhance pathogenesis. KSHV latent infection and lytic reactivation are characterized by distinct gene expression profiles, and both latency and lytic reactivation seem to be required for malignant progression. As a sophisticated oncogenic virus, KSHV has evolved to possess a formidable repertoire of potent mechanisms that enable it to target and manipulate host cell pathways, leading to increased cell proliferation, increased cell survival, dysregulated angiogenesis, evasion of immunity, and malignant progression in the immunocompromised host. Worldwide, approximately 40.3 million people are currently living with HIV infection. Of these, a significant number are coinfected with KSHV. The complex interplay between the two viruses dramatically elevates the risk for development of KSHV-induced malignancies, KS, PEL, and MCD. Although HAART significantly reduces HIV viral load, the entire T-cell repertoire and immune function may not be completely restored. In fact, clinically significant immune deficiency is not necessary for the induction of KSHV-related malignancy. Because of variables such as lack of access to therapy noncompliance with prescribed treatment, failure to respond to treatment and the development of drug-resistant strains of HIV, KSHV-induced malignancies will continue to present as major health concerns.
Collapse
Affiliation(s)
- Whitney Greene
- Tiumor Virology Program, Children's Cancer Research Institute, Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|
58
|
Wu L, KewalRamani VN. Dendritic-cell interactions with HIV: infection and viral dissemination. Nat Rev Immunol 2006; 6:859-68. [PMID: 17063186 PMCID: PMC1796806 DOI: 10.1038/nri1960] [Citation(s) in RCA: 421] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dendritic cells (DCs) are crucial for the generation and the regulation of adaptive immunity. Because DCs have a pivotal role in marshalling immune responses, HIV has evolved ways to exploit DCs, thereby facilitating viral dissemination and allowing evasion of antiviral immunity. Defining the mechanisms that underlie cell-cell transmission of HIV and understanding the role of DCs in this process should help us in the fight against HIV infection. This Review highlights the latest advances in our understanding of the interactions between DCs and HIV, focusing on the mechanisms of DC-mediated viral dissemination.
Collapse
Affiliation(s)
- Li Wu
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
59
|
Wang JH, Janas AM, Olson WJ, KewalRamani VN, Wu L. CD4 coexpression regulates DC-SIGN-mediated transmission of human immunodeficiency virus type 1. J Virol 2006; 81:2497-507. [PMID: 17151103 PMCID: PMC1865928 DOI: 10.1128/jvi.01970-06] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DCs) potently stimulate the cell-cell transmission of human immunodeficiency virus type 1 (HIV-1). However, the mechanisms that underlie DC transmission of HIV-1 to CD4(+) T cells are not fully understood. DC-SIGN, a C-type lectin, efficiently promotes HIV-1 trans infection. DC-SIGN is expressed in monocyte-derived DCs (MDDCs), macrophage subsets, activated B lymphocytes, and various mucosal tissues. MDDC-mediated HIV-1 transmission to CD4(+) T cells involves DC-SIGN-dependent and -independent mechanisms. DC-SIGN transmission of HIV-1 depends on the donor cell type. HIV-1 Nef can upregulate DC-SIGN expression and promote DC-T-cell clustering and HIV-1 spread. Nef also downregulates CD4 expression; however, the effect of the CD4 downmodulation on DC-mediated HIV-1 transmission has not been examined. Here, we report that CD4 expression levels correlate with inefficient HIV-1 transmission by monocytic cells expressing DC-SIGN. Expression of CD4 on Raji B cells strongly impaired DC-SIGN-mediated HIV-1 transmission to T cells. By contrast, enhanced HIV-1 transmission was observed when CD4 molecules on MDDCs and DC-SIGN-CD4-expressing cell lines were blocked with specific antibodies. Coexpression of CD4 and DC-SIGN in Raji cells promoted the internalization and intracellular retention of HIV-1. Interestingly, internalized HIV-1 particles were sorted and confined to late endosomal compartments that were positive for CD63 and CD81. Furthermore, in HIV-1-infected MDDCs, significant downregulation of CD4 by Nef expression correlated with enhanced viral transmission. These results suggest that CD4, which is present at various levels in DC-SIGN-positive primary cells, is a key regulator of HIV-1 transmission.
Collapse
Affiliation(s)
- Jian-Hua Wang
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, BSB 203, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
60
|
Chaipan C, Soilleux EJ, Simpson P, Hofmann H, Gramberg T, Marzi A, Geier M, Stewart EA, Eisemann J, Steinkasserer A, Suzuki-Inoue K, Fuller GL, Pearce AC, Watson SP, Hoxie JA, Baribaud F, Pöhlmann S. DC-SIGN and CLEC-2 mediate human immunodeficiency virus type 1 capture by platelets. J Virol 2006; 80:8951-60. [PMID: 16940507 PMCID: PMC1563896 DOI: 10.1128/jvi.00136-06] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets can engulf human immunodeficiency virus type 1 (HIV-1), and a significant amount of HIV-1 in the blood of infected individuals is associated with these cells. However, it is unclear how platelets capture HIV-1 and whether platelet-associated virus remains infectious. DC-SIGN and other lectins contribute to capture of HIV-1 by dendritic cells (DCs) and facilitate HIV-1 spread in DC/T-cell cocultures. Here, we show that platelets express both the C-type lectin-like receptor 2 (CLEC-2) and low levels of DC-SIGN. CLEC-2 bound to HIV-1, irrespective of the presence of the viral envelope protein, and facilitated HIV-1 capture by platelets. However, a substantial fraction of the HIV-1 binding activity of platelets was dependent on DC-SIGN. A combination of DC-SIGN and CLEC-2 inhibitors strongly reduced HIV-1 association with platelets, indicating that these lectins are required for efficient HIV-1 binding to platelets. Captured HIV-1 was maintained in an infectious state over several days, suggesting that HIV-1 can escape degradation by platelets and might use these cells to promote its spread. Our results identify CLEC-2 as a novel HIV-1 attachment factor and provide evidence that platelets capture and transfer infectious HIV-1 via DC-SIGN and CLEC-2, thereby possibly facilitating HIV-1 dissemination in infected patients.
Collapse
Affiliation(s)
- Chawaree Chaipan
- Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Glückstrasse 6, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Donaghy H, Wilkinson J, Cunningham AL. HIV interactions with dendritic cells: has our focus been too narrow? J Leukoc Biol 2006; 80:1001-12. [PMID: 16923917 DOI: 10.1189/jlb.0306158] [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] [Indexed: 12/13/2022] Open
Abstract
Although few in number, dendritic cells (DCs) are heterogeneous, ubiquitous, and are crucial for protection against pathogens. In this review, the different DC subpopulations have been described and aspects of DC biology are discussed. DCs are important, not only in the pathogenesis of HIV, but also in the generation of anti-HIV immune responses. This review describes the roles that DC are thought to play in HIV pathogenesis, including uptake and transport of virus. We have also discussed the effects that the virus exerts on DCs such as infection and dysfunction. Then we proceed to focus on DC subsets in different organs and show how widespread the effects of HIV are on DC populations. It is clear that the small number of studies on tissue-derived DCs limits current research into the pathogenesis of HIV.
Collapse
Affiliation(s)
- Heather Donaghy
- Centre for Virus Research, Westmead Millennium Institute, Westmead Hosptial, Darcy Rd., Sydney, NSW 2145, Australia
| | | | | |
Collapse
|
62
|
Soilleux EJ, Sarno EN, Hernandez MO, Moseley E, Horsley J, Lopes UG, Goddard MJ, Vowler SL, Coleman N, Shattock RJ, Sampaio EP. DC-SIGN association with the Th2 environment of lepromatous lesions: cause or effect? J Pathol 2006; 209:182-9. [PMID: 16583355 DOI: 10.1002/path.1972] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The clinical spectrum of leprosy is related to patients' immune responses. Non-responsiveness towards Mycobacterium leprae (ML) seems to correlate with a Th2 cytokine profile. The reason for such a polarized immune response remains unclear. The C-type lectin, DC-SIGN, expressed by subsets of dendritic cells (DCs) and macrophages, has previously been associated with Th2 responses. Here we show abundant DC-SIGN expression in lepromatous but not borderline tuberculoid leprosy, in both HIV-positive and HIV-negative patients. Moreover, we demonstrate that DC-SIGN can act as an entry receptor for ML, as it does for M. tuberculosis, through the cell wall component lipoarabinomannan. DC-SIGN is expressed on virtually all ML-containing cells, providing further evidence for its role as a receptor. DC-SIGN may therefore be induced on macrophages in lepromatous leprosy and may then contribute to mycobacterial entry into these cells.
Collapse
Affiliation(s)
- E J Soilleux
- Department of Histopathology, Papworth Hospital, Papworth Everard, Cambridge CB3 8RE, and Nuffield Department of Clinical Laboratory Sciences, University of Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
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
|
64
|
Rappocciolo G, Jenkins FJ, Hensler HR, Piazza P, Jais M, Borowski L, Watkins SC, Rinaldo CR. DC-SIGN is a receptor for human herpesvirus 8 on dendritic cells and macrophages. THE JOURNAL OF IMMUNOLOGY 2006; 176:1741-9. [PMID: 16424204 DOI: 10.4049/jimmunol.176.3.1741] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human herpesvirus 8 (HHV-8) causes Kaposi's sarcoma and pleural effusion lymphoma. In this study, we show that dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN; CD209) is a receptor for HHV-8 infection of myeloid DCs and macrophages. DC-SIGN was required for virus attachment to these cells and DC-SIGN-expressing cell lines. HHV-8 binding and infection were blocked by anti-DC-SIGN mAb and soluble DC-SIGN, and mannan, a natural ligand for DC-SIGN. Infection of DCs and macrophages with HHV-8 led to production of viral proteins, with little production of viral DNA, similar to HHV-8 infection of vascular endothelial cells. Infection of DCs resulted in down-regulation of DC-SIGN, a decrease in endocytic activity, and an inhibition of Ag stimulation of CD8+ T cells. We propose that DC-SIGN serves as a portal for immune dysfunction and oncogenesis caused by HHV-8 infection.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/metabolism
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cell Line
- Cell Line, Transformed
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Herpesviridae Infections/immunology
- Herpesvirus 8, Human/metabolism
- Humans
- Integrin alpha3beta1/physiology
- K562 Cells
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Macrophages/metabolism
- Mannans/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
Collapse
Affiliation(s)
- Giovanna Rappocciolo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | |
Collapse
|
65
|
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
|
66
|
Breburda EE, Dambaeva SV, Slukvin II, Golos TG. Selective distribution and pregnancy-specific expression of DC-SIGN at the maternal–fetal interface in the rhesus macaque: DC-SIGN is a putative marker of the recognition of pregnancy. Placenta 2006; 27:11-21. [PMID: 16310033 DOI: 10.1016/j.placenta.2004.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2004] [Revised: 11/03/2004] [Accepted: 11/08/2004] [Indexed: 11/21/2022]
Abstract
We performed immunohistochemical analysis of DC-SIGN expression at the maternal-fetal interface at different stages of pregnancy in the rhesus monkey. Natural killer cells, monocytes and macrophages were observed in the nonpregnant endometrium, particularly in the luteal phase, and were increased in pregnant endometrium. No DC-SIGN+ cells were observed in the nonpregnant uterus. We observed decidual DC-SIGN+ cells within 1 week of implantation, and they increased in number during the first 5 weeks of gestation. DC-SIGN+ cells showed a clear differential distribution in the decidua in the first 2 weeks of pregnancy, being found only adjacent to the implantation site, in marked contrast to the widespread distribution of CD68+ macrophages and CD56+ NK cells throughout the endometrium. DC-SIGN+ cells also showed a more dendritic morphology than the general CD68+ cell population, and analysis of serial sections indicated an overlapping but not identical localization of these markers. Mature dendritic cells could not be detected as judged by total absence of immunostaining for CD83, CD86, DEC-205, or CD1a. DC-SIGN+ cells were defined as MHC class II+ and CD14+ by flow cytometry. We conclude that DC-SIGN expression is an early response by the primate maternal immune system to the implanting embryo. The selectively distributed population of DC-SIGN+ decidual leukocytes may represent a morphologically and phenotypically distinct subpopulation of decidual macrophages of early pregnancy that could contribute to the establishment of maternal-fetal immune tolerance.
Collapse
Affiliation(s)
- E E Breburda
- National Primate Research Center, University of Wisconsin Medical School, University of Wisconsin-Madison, 1223 Capitol Court, Madison, WI 53715-1299, USA
| | | | | | | |
Collapse
|
67
|
Abstract
Multiple mechanisms underlie the surprising willingness of mothers to tolerate genetically different fetal tissues during pregnancy. Chief among these is the choice of HLA-G, a gene with few alleles, rather than the highly polymorphic HLA-A and -B genes, for expression by the placental cells that interface directly with maternal blood and tissues. Novel aspects of this major histocompatibility complex class Ib gene include alternative splicing to permit production of membrane and soluble isoforms, deletions that dampen responses to interferons, and a shortened cytoplasmic tail that affects expression at the cell surface. Placental cells migrating into the maternal uterus synthesize both membrane and soluble isoforms, which interact with inhibitory receptors on leukocytes such as ILT2 and ILT4. Cytotoxic T lymphocytes either die or reduce production of one of their major coreceptor/activator cell surface molecules, CD8; natural killer cells are immobilized and mononuclear phagocytes are programmed into suppressive modes characterized by high production of anti-inflammatory cytokines. The idea that placental HLA-G proteins facilitate semiallogeneic pregnancy by inhibiting maternal immune responses to foreign (paternal) antigens via these actions on immune cells is now well established, and the postulate that the recombinant counterparts of these proteins may be used as powerful tools for preventing immune rejection of transplanted organs is gaining in popularity.
Collapse
Affiliation(s)
- Joan S Hunt
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas 66160-7400, USA.
| | | | | | | |
Collapse
|
68
|
Granelli-Piperno A, Pritsker A, Pack M, Shimeliovich I, Arrighi JF, Park CG, Trumpfheller C, Piguet V, Moran TM, Steinman RM. Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin/CD209 is abundant on macrophages in the normal human lymph node and is not required for dendritic cell stimulation of the mixed leukocyte reaction. THE JOURNAL OF IMMUNOLOGY 2005; 175:4265-73. [PMID: 16177066 PMCID: PMC1378112 DOI: 10.4049/jimmunol.175.7.4265] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The C-type lectin dendritic cell-specific ICAM 3-grabbing nonintegrin (DC-SIGN)/CD209 efficiently binds several pathogens, including HIV-1. DC-SIGN is expressed on monocyte-derived DCs in culture, and importantly, it is able to sequester HIV-1 within cells and facilitate transmission of virus to CD4+ T cells. To investigate DC-SIGN function, we have generated new mAbs. We report in this study that these and prior anti-DC-SIGN mAbs primarily label macrophages in the medullary sinuses of noninflamed human lymph node. In contrast, expression is not detected on most DCs in the T cell area, except for occasional cells. We also noted that IL-4 alone can induce expression of DC-SIGN in CD14+ monocytes and circulating blood DCs. However, blockade of DC-SIGN with Abs and DC-SIGN small interfering RNA did not result in a major reduction in the capacity of these DCs to transfer HIV to T cells, confirming significant DC-SIGN-independent mechanisms. The blocking approaches did reduce HIV-1 transmission by DC-SIGN-transfected cells by >90%. DC-SIGN blockade also did not reduce the ability of DCs to stimulate T cell proliferation in the MLR. These results indicate that DC-SIGN has the potential to contribute to macrophage function in normal human lymph node, and that DCs do not require DC-SIGN to transmit HIV or to initiate T cell responses.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Cell Adhesion Molecules/antagonists & inhibitors
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cell Line
- Clone Cells
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Dogs
- HIV-1/immunology
- Humans
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/metabolism
- Lymphocyte Culture Test, Mixed
- Macrophages/metabolism
- Mice
- Mice, Inbred BALB C
- RNA, Small Interfering/metabolism
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/metabolism
- T-Lymphocytes/virology
- Transfection
Collapse
Affiliation(s)
- Angela Granelli-Piperno
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
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
|
70
|
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
|
71
|
Neil SJD, Aasa-Chapman MMI, Clapham PR, Nibbs RJ, McKnight A, Weiss RA. The promiscuous CC chemokine receptor D6 is a functional coreceptor for primary isolates of human immunodeficiency virus type 1 (HIV-1) and HIV-2 on astrocytes. J Virol 2005; 79:9618-24. [PMID: 16014924 PMCID: PMC1181543 DOI: 10.1128/jvi.79.15.9618-9624.2005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The role of coreceptors other than CCR5 and CXCR4 in the pathogenesis of human immunodeficiency virus (HIV) disease is controversial. Here we show that a promiscuous CC chemokine receptor, D6, can function as a coreceptor for various primary dual-tropic isolates of HIV type 1 (HIV-1) and HIV-2. Furthermore, D6 usage is common among chimeric HIV-1 constructs bearing the gp120 proteins of isolates from early seroconverting patients. D6 mRNA and immunoreactivity were demonstrated to be expressed in HIV-1 target cells such as macrophages, peripheral blood mononuclear cells, and primary astrocytes. In primary astrocytes, an RNA interference-mediated knockdown of D6 expression inhibited D6-tropic isolate infection. D6 usage may account for some previous observations of alternative receptor tropism for primary human cells. Thus, D6 may be an important receptor for HIV pathogenesis in the brain and for the early dissemination of virus in the host.
Collapse
Affiliation(s)
- Stuart J D Neil
- Wohl Virion Centre, Division of Infection and Immunity, University College London, UK
| | | | | | | | | | | |
Collapse
|
72
|
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
|
73
|
Abstract
Maternal antigen presenting cells, which are macrophages and dendritic cells, are scattered throughout human decidualized endometrium during all stages of pregnancy. These powerful, multi-functional leukocytes reside in close proximity to uterine glandular epithelium, uterine blood vessels, and HLA-G-producing invasive cytotrophoblast cells. Macrophages and dendritic cells, which express the HLA-G receptors, ILT2 and ILT4, play major roles in driving innate and adaptive immune responses, altering the behavior of local stromal cells, shaping the cytokine microenvironment, and protecting the tissue from infection. Therefore, encounters between decidual antigen presenting cells and HLA-G molecules are likely to influence uterine and placental homeostasis as well as local maternal immune responses to the fetus during pregnancy.
Collapse
Affiliation(s)
- R H McIntire
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | |
Collapse
|
74
|
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
|
75
|
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
|
76
|
Abstract
In pregnancy, the endometrium represents the hosting surface for invading semi-allogeneic cells. A very fine tuning of cellular interactions is indispensible for the successful symbiosis of mother and fetus. Such interactions include a variety of immune cells, which are present in extremely high concentrations, and trophoblast cells. A wide spectrum of soluble and surface molecules is involved in homing, activation and switching of lymphocyte functions, but also in regulation of embryo implantation and control of trophoblast invasion. This mini-review provides a brief overview of immunological features of the endometrium.
Collapse
|
77
|
Rieger L, Honig A, Sütterlin M, Kapp M, Dietl J, Ruck P, Kämmerer U. Antigen-presenting cells in human endometrium during the menstrual cycle compared to early pregnancy. JOURNAL OF THE SOCIETY FOR GYNECOLOGIC INVESTIGATION 2004; 11:488-93. [PMID: 15458747 DOI: 10.1016/j.jsgi.2004.05.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Human endometrium and early pregnancy decidua harbor a considerable and diverse population of antigen-presenting cells (APC). Changes in the number and distribution of macrophages and dendritic cells (DC) could point to a possible role of these immunocompetent cells in implantation and success of early pregnancy. METHODS Uterine tissue was obtained from 22 women undergoing hysterectomy for bleeding disorders or dysmenorrhea and from 11 women undergoing legal abortion. Tissue was investigated with antibodies against CD14, CD68, CD83, DC-SIGN, Ki-67, and human leukocyte antigen (HLA)-DR using single and double immunohistochemical staining techniques. RESULTS The number of CD14(+) cells was stable during all phases of the menstrual cycle and early pregnancy. In comparison to nonpregnant endometrium, DC-SIGN(+) cells showed a higher proliferation rate and were found associated in clusters with CD56(+) natural killer (NK) cells in early pregnancy. In the late secretory phase of the menstrual cycle, numbers of CD83(+) (P <.01) cells were significantly higher than in other endometrial phases and early pregnancy. HLA-DR(+) expression was significantly increased in early pregnancy but remained unchanged throughout the menstrual cycle. CONCLUSION The presence of DC-SIGN(+) cells during the menstrual cycle and their proliferation in early pregnancy suggests an important role of these cells with regard to the balance between defense against pathogens and tolerance of the fetal allograft. Whether the increase of CD83(+) mature DC and CD68(+) macrophages in the late secretory phase is caused by hormonal stimuli and/or is due to changes of the cytokine/chemokine micromilieu remains to be investigated.
Collapse
Affiliation(s)
- L Rieger
- Department of Obstetrics and Gynecology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
78
|
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
|
79
|
Gieseler RK, Marquitan G, Hahn MJ, Perdon LA, Driessen WHP, Sullivan SM, Scolaro MJ. DC-SIGN-specific liposomal targeting and selective intracellular compound delivery to human myeloid dendritic cells: implications for HIV disease. Scand J Immunol 2004; 59:415-24. [PMID: 15140050 DOI: 10.1111/j.0300-9475.2004.01431.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Myeloid dendritic cells (MyDCs), prime stimulators of antigen-specific immunity, can serve as one of the major reservoirs for human immunodeficiency virus type-1 (HIV-1). Utilizing mature monocyte-derived MyDCs generated with granulocyte/macrophage colony-stimulating factor, interleukin-4, and tumour necrosis factor-alpha as an in vitro model, we here present the first proof of concept for liposomal compound delivery to these cells by specifically addressing CD209, i.e. DC-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN), a MyDC-associated C-type lectin implicated in the transmission of HIV-1 to T helper cells. By employing a liposomally entrapped tracer, calcein, we demonstrate by flow cytometry and mathematics a superior targeting efficacy for DC-SIGN, as compared with select other MyDC markers (CD1a, CD4, CD45R0, and CD83). Fluorescence microscopy reveals time-dependent surface binding and intracellular uptake of DC-SIGN-specific liposomes by both immature and mature MyDCs. This pilot study implies that liposomal targeting to CD209 and related C-type lectins may afford therapeutic intracellular drug delivery to MyDCs and other reservoir and nonreservoir cells susceptible to infection with HIV-1.
Collapse
Affiliation(s)
- R K Gieseler
- Laboratories of Immunology and Molecular Biology, LTBH Medical Research Institute, Robertson Building, Beverly Hills, CA 90211, USA.
| | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
Mechanism of maternal retroviral transmission remains an unsolved problem. The current investigation is a part of our ongoing research on vertical transmission of MoMuLV-TB ts1 in BALB/c mice. A total of 270 adult mice and 165 fetuses were used. Forty-four experimental mice were injected with 0.1 mL of 4.0 x 10(6) ffu/mL of ts1 virus at 72 h after birth; 24 controls were injected with DMEM. Almost half of the females went through two rounds of pregnancies. In the first round, 135 experimental and 57 control pups were produced. Forty-three experimental and 20 control pups were followed until they developed clinical symptoms. The second round of pregnancy produced a total of 46 mid-gestational and 119 full-term fetuses. PCR, and light and electron microscopy were performed to evaluate viral transmission. Overall, 99% vertical transmission occurred in pups of infected mothers. Twelve percent of mid-gestational and 39% full-term fetuses were PCR positive. We have established that, if mothers are infected with ts1 virus at 72 h after birth, then nearly 100% vertical transmission occurs, via in utero, intrapartum, or breast milk. Thirty-nine percent transmission occurred in utero alone. This is an excellent model to study the transplacental and post-gestational transmission of retroviruses, such as ts1.
Collapse
Affiliation(s)
- Joan Duggan
- Department of Physiology and Molecular Medicine, Medical College of Ohio, Toledo, Ohio 43614-5804, USA
| | | | | |
Collapse
|
81
|
Slukvin II, Breburda EE, Golos TG. Dynamic Changes in Primate Endometrial Leukocyte Populations: Differential Distribution of Macrophages and Natural Killer Cells at the Rhesus Monkey Implantation Site and in Early Pregnancy. Placenta 2004; 25:297-307. [PMID: 15028422 DOI: 10.1016/j.placenta.2003.08.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2003] [Revised: 07/24/2003] [Accepted: 08/02/2003] [Indexed: 10/26/2022]
Abstract
The distribution of uterine leukocytes during the periimplantation period cannot be readily evaluated in human pregnancy. Using immunohistochemistry we examined the distribution of macrophages, natural killer (NK) cells, and T cells in the non-pregnant endometrium and in the decidua at early stages of implantation and pregnancy in the rhesus monkey. CD68+ macrophages, CD56+ lymphocytes and CD3+ T cells were present in the proliferative and secretory endometrium. The number of macrophages and CD56+ lymphocytes dramatically increased at implantation (day 14-15 of pregnancy) and continued to be high in early pregnancy decidua. Macrophages were conspicuously more numerous in proximity to implantation site (decidua basalis) as compared to sites peripheral to the developing placenta (decidua parietalis), and were found in close association with cytotrophoblasts adjacent to the decidua, as well as around arteries invaded by extravillous cytotrophoblasts. In contrast to macrophages, CD56+ lymphocytes were more evenly distributed throughout the decidua. Few CD3+ T cells were seen in pregnancy, being scattered in the endometrial stroma with occasional aggregate formation. The distribution of uterine leukocytes vis-à-vis trophoblasts at the rhesus monkey implantation site and in early pregnancy suggests different roles for macrophages and uterine NK cells in the response to trophoblast invasion.
Collapse
Affiliation(s)
- I I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin, 1223 Capitol Court, Madison, WI 53715-1299, USA
| | | | | |
Collapse
|
82
|
Soilleux EJ, Coleman N. Expression of DC-SIGN in human foreskin may facilitate sexual transmission of HIV. J Clin Pathol 2004; 57:77-8. [PMID: 14693841 PMCID: PMC1770180 DOI: 10.1136/jcp.57.1.77] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
This study demonstrates that the human immunodeficiency virus (HIV) binding C-type lectin DC-SIGN is coexpressed with CD4 and CCR5 on dendritic cells/macrophages in human foreskin. It is hypothesised that DC-SIGN may contribute to the sexual transmission of HIV in the foreskin, by enabling infection of permissive cells in cis and/or in trans.
Collapse
|
83
|
Pereira L, Maidji E, McDonagh S, Genbacev O, Fisher S. Human cytomegalovirus transmission from the uterus to the placenta correlates with the presence of pathogenic bacteria and maternal immunity. J Virol 2004; 77:13301-14. [PMID: 14645586 PMCID: PMC296088 DOI: 10.1128/jvi.77.24.13301-13314.2003] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prenatal cytomegalovirus infection may cause pregnancy complications such as intrauterine growth restriction and birth defects. How virus from the mother traverses the placenta is unknown. PCR analysis of biopsy specimens of the maternal-fetal interface revealed that DNA sequences from cytomegalovirus were commonly found with those of herpes simplex viruses and pathogenic bacteria. Cytomegalovirus DNA and infected cell proteins were found more often in the decidua than in the placenta, suggesting that the uterus functions as a reservoir for infection. In women with low neutralizing titers, cytomegalovirus replicated in diverse decidual cells and placental trophoblasts and capillaries. In women with intermediate to high neutralizing titers, decidual infection was suppressed and the placenta was spared. Overall, cytomegalovirus virions and maternal immunoglobulin G were detected in syncytiotrophoblasts, villus core macrophages, and dendritic cells. These results suggest that the outcome of cytomegalovirus infection depends on the presence of other pathogens and coordinated immune responses to viral replication at the maternal-fetal interface.
Collapse
Affiliation(s)
- Lenore Pereira
- Departments of Stomatology, University of California-San Francisco, San Francisco, California 94143-0512, USA.
| | | | | | | | | |
Collapse
|
84
|
Klimstra WB, Nangle EM, Smith MS, Yurochko AD, Ryman KD. DC-SIGN and L-SIGN can act as attachment receptors for alphaviruses and distinguish between mosquito cell- and mammalian cell-derived viruses. J Virol 2003; 77:12022-32. [PMID: 14581539 PMCID: PMC254289 DOI: 10.1128/jvi.77.22.12022-12032.2003] [Citation(s) in RCA: 182] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
C-type lectins such as DC-SIGN and L-SIGN, which bind mannose-enriched carbohydrate modifications of host and pathogen proteins, have been shown to bind glycoproteins of several viruses and facilitate either cis or trans infection. DC-SIGN and L-SIGN are expressed in several early targets of arbovirus infection, including dendritic cells (DCs) and cells of the reticuloendothelial system. In the present study, we show that DC-SIGN and L-SIGN can function as attachment receptors for Sindbis (SB) virus, an arbovirus of the Alphavirus genus. Human monocytic THP-1 cells stably transfected with DC-SIGN or L-SIGN were permissive for SB virus replication, while untransfected controls were essentially nonpermissive. The majority of control THP-1 cells were permissive when attachment and entry steps were eliminated through electroporation of virus transcripts. Infectivity for the DC-SIGN/L-SIGN-expressing cells was largely blocked by yeast mannan, EDTA, or a DC-SIGN/L-SIGN-specific monoclonal antibody. Infection of primary human DCs by SB virus was also dependent upon SIGN expression by similar criteria. Furthermore, production of virus particles in either C6/36 mosquito cells or CHO mammalian cells under conditions that limited complex carbohydrate content greatly increased SB virus binding to and infection of THP-1 cells expressing these lectins. C6/36-derived virus also was much more infectious for primary human DCs than CHO-derived virus. These results suggest that (i) lectin molecules such as DC-SIGN and L-SIGN may represent common attachment receptor molecules for arthropod-borne viruses, (ii) arbovirus particles produced in and delivered by arthropod vectors may preferentially target vertebrate host cells bearing these or similar lectin molecules, and (iii) a cell line has been identified that can productively replicate alphaviruses but is deficient in attachment receptors.
Collapse
Affiliation(s)
- William B Klimstra
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA.
| | | | | | | | | |
Collapse
|
85
|
Liu H, Yu W, Liou LY, Rice AP. Isolation and characterization of the human DC-SIGN and DC-SIGNR promoters. Gene 2003; 313:149-59. [PMID: 12957386 DOI: 10.1016/s0378-1119(03)00674-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
DC-SIGN is a C-type lectin expressed on the surface of dendritic cells (DCs) that is used by a number of human pathogens to disseminate infection in the host. In the human genome, there is a gene closely related to DC-SIGN, termed DC-SIGNR (also L-SIGN, DC-SIGN2), which likely arose through gene duplication. DC-SIGN protein and RNA expression is largely restricted to DCs and some specialized macrophages in lung and placenta, while DC-SIGNR expression is largely restricted to lymph nodes and liver sinusoidal endothelial cells. To begin to investigate the cell type-restricted expression of these closely related genes, we isolated the human DC-SIGN and DC-SIGNR promoters. They were found to be relatively weak promoters that express similarly in plasmid transfection assays in several transformed cell lines, suggesting that the cis-regulatory elements that confer cell-type restricted expression of these two genes are located outside of the promoters. The DC-SIGN gene contains four major transcriptional start sites at +1, +271, +364, and +435, with the +364 site being the most abundantly expressed in DCs. The DC-SIGN promoter is contained within nucleotides +251 to +487. AP-1, Sp1, Ets-1, and NF-kappaB binding sites in the DC-SIGN promoter appear to be important for function. Thus, despite its highly restricted pattern of expression, the DC-SIGN promoter has features common to promoters that are active in other cell types.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
86
|
Abstract
Dendritic cells (DCs) in the pregnant human uterine mucosa have been poorly characterized, although they are likely to regulate immune responses to both placental trophoblast cells and uterine infections. In this study an HLA-DR+, CD11c+ lin- (CD3-, CD19-, CD56-, CD14-) population has been identified by three-color flow cytometry. The cell isolates were prepared either by collagenase digestion or mechanically from first-trimester decidual tissue. The decidual DCs comprised approximately 1.7% of CD45+ cells in the isolates and had the phenotype of immature myeloid DCs. No CD1a+ Langerhans cells or CD123+ plasmacytoid DCs were detected. The decidual DCs were DC-SIGN-, DEC-205+, CD40+. Two subsets could be distinguished on the basis of relative expression of HLA-DR, which also differed in expression of DC-activation markers. The DCs were identified in situ by immunohistology by DEC-205 staining. Cells with dendritic processes were found scattered through both the decidua basalis (in which trophoblast cells are infiltrating) and the decidua parietalis. They were also visible in endothelial-lined spaces. This is the first study to identify and describe the phenotype and distribution of human decidual DCs.
Collapse
Affiliation(s)
- Lucy Gardner
- Research Group in Human Reproductive Immunobiology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | | |
Collapse
|
87
|
Lozach PY, Lortat-Jacob H, de Lacroix de Lavalette A, Staropoli I, Foung S, Amara A, Houles C, Fieschi F, Schwartz O, Virelizier JL, Arenzana-Seisdedos F, Altmeyer R. DC-SIGN and L-SIGN are high affinity binding receptors for hepatitis C virus glycoprotein E2. J Biol Chem 2003; 278:20358-66. [PMID: 12609975 DOI: 10.1074/jbc.m301284200] [Citation(s) in RCA: 289] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The hepatitis C virus (HCV) genome codes for highly mannosylated envelope proteins, which are naturally retained in the endoplasmic reticulum. We found that the HCV envelope glycoprotein E2 binds the dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) and the related liver endothelial cell lectin L-SIGN through high-mannose N-glycans. Competing ligands such as mannan and an antibody directed against the carbohydrate recognition domains (CRD) abrogated binding. While no E2 interaction with distant monomeric CRDs on biosensor chips could be detected, binding is observed if CRDs are closely seeded (Kd = 48 nm) and if the CRD is part of the oligomeric-soluble extracellular domain of DC-SIGN (Kd = 30 nm). The highest affinity is seen for plasma membrane-expressed DC-SIGN and L-SIGN (Kd = 3 and 6 nm, respectively). These results indicate that several high-mannose N-glycans in a structurally defined cluster on E2 bind to several subunits of the oligomeric lectin CRD. High affinity interaction of viral glycoproteins with oligomeric lectins might represent a strategy by which HCV targets to and concentrates in the liver and infects dendritic cells.
Collapse
Affiliation(s)
- Pierre-Yves Lozach
- Unité d'Immunologie Virale, Institut Pasteur, 28, rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Soilleux EJ, Rous B, Love K, Vowler S, Morris LS, Fisher C, Coleman N. Myxofibrosarcomas Contain Large Numbers of Infiltrating Immature Dendritic Cells. Am J Clin Pathol 2003. [DOI: 10.1309/jeb7dghh01j11vum] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
89
|
Pöhlmann S, Zhang J, Baribaud F, Chen Z, Leslie GJ, Lin G, Granelli-Piperno A, Doms RW, Rice CM, McKeating JA. Hepatitis C virus glycoproteins interact with DC-SIGN and DC-SIGNR. J Virol 2003; 77:4070-80. [PMID: 12634366 PMCID: PMC150620 DOI: 10.1128/jvi.77.7.4070-4080.2003] [Citation(s) in RCA: 310] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
DC-SIGN and DC-SIGNR are two closely related membrane-associated C-type lectins that bind human immunodeficiency virus (HIV) envelope glycoprotein with high affinity. Binding of HIV to cells expressing DC-SIGN or DC-SIGNR can enhance the efficiency of infection of cells coexpressing the specific HIV receptors. DC-SIGN is expressed on some dendritic cells, while DC-SIGNR is localized to certain endothelial cell populations, including hepatic sinusoidal endothelial cells. We found that soluble versions of the hepatitis C virus (HCV) E2 glycoprotein and retrovirus pseudotypes expressing chimeric forms of both HCV E1 and E2 glycoproteins bound efficiently to DC-SIGN and DC-SIGNR expressed on cell lines and primary human endothelial cells but not to other C-type lectins tested. Soluble E2 bound to immature and mature human monocyte-derived dendritic cells (MDDCs). Binding of E2 to immature MDDCs was dependent on DC-SIGN interactions, while binding to mature MDDCs was partly independent of DC-SIGN, suggesting that other cell surface molecules may mediate HCV glycoprotein interactions. HCV interactions with DC-SIGN and DC-SIGNR may contribute to the establishment or persistence of infection both by the capture and delivery of virus to the liver and by modulating dendritic cell function.
Collapse
Affiliation(s)
- Stefan Pöhlmann
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
Defining the mechanisms of HIV-1 entry has enabled the rational design of strategies aimed at interfering with the process. This article delineates what is currently understood about HIV-1 entry, as a window through which to understand what will likely be the next major group of antiretroviral therapeutics. These exciting new approaches offer the promise of adding viral entry to reverse transcription and protein processing as steps to block in the viral life cycle. Several principles learned with other antiretroviral drugs are sure to be valid for entry antagonists, whereas other considerations may be unique to this group of agents. There is no agent to which HIV-1 has not been able to acquire resistance and this is likely to remain the case. Multiple rounds of viral replication are required to generate the genetic diversity that forms the basis of resistance. Combination therapy in which replication is maximally suppressed will remain a cornerstone of treatment with entry inhibitors, as with other agents. Furthermore, the coreceptor specificity of some entry and fusion inhibitors argues that combinations will likely be needed to broaden the effective range of susceptible viral variants. Finally, the targeting of multiple steps within the entry process has the potential for synergy. The fusion inhibitor T20 and CXCR4 antagonist AMD3100 are synergistic in vitro at blocking infection of PBMC with clinical isolates [115] and T20 combined with the CD4 inhibitor PRO 542 have synergistic in vitro effects, with more than 10-fold greater inhibition of R5, X4, and R5X4 strains than either agent alone [116]. Entry antagonists raise other, unique issues. As discussed previously, the theoretic concern exists that blocking CCR5 could enhance the emergence of CXCR4-using variants and possibly accelerate disease. So far, in vitro selection for variants resistant to the CCR5 antagonist SCH-C in PBMC (which express both CCR5 and CXCR4) has resulted in mutants that were resistant to the blocker but still used CCR5. Alternatively, because many HIV-1 strains have the capacity to use several other chemokine or orphan receptors for entry, blocking both CCR5 and CXCR could lead to a variant that uses one of these other molecules in place of the principal coreceptors, although data in vitro so far suggest that this is unlikely [13,14]. This new class of antiviral drugs offers great promise but also novel concerns, and careful analysis of viruses that arise with their use in vivo is essential.
Collapse
Affiliation(s)
- Linda D Starr-Spires
- Pulmonary, Allergy and Critical Care Division, University of Pennsylvania School of Medicine, 522 Johnson Pavilion, 36th and Hamilton Walk, Philadelphia, PA 19104-6060, USA
| | | |
Collapse
|
91
|
Kämmerer U, Eggert AO, Kapp M, McLellan AD, Geijtenbeek TBH, Dietl J, van Kooyk Y, Kämpgen E. Unique appearance of proliferating antigen-presenting cells expressing DC-SIGN (CD209) in the decidua of early human pregnancy. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:887-96. [PMID: 12598322 PMCID: PMC1868095 DOI: 10.1016/s0002-9440(10)63884-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intact human pregnancy can be regarded as an immunological paradox in that the maternal immune system accepts the allogeneic embryo without general immunosuppression. Because dendritic cell (DC) subsets could be involved in peripheral tolerance, the uterine mucosa (decidua) was investigated for DC populations. Here we describe the detailed immunohistochemical and functional characterization of HLA-DR-positive antigen-presenting cells (APCs) in early pregnancy decidua. In contrast to classical macrophages and CD83(+) DCs, which were found in comparable numbers in decidua and nonpregnant endometrium, only decidua harbored a significant population of HLA-DR(+)/DC-SIGN(+) APCs further phenotyped as CD14(+)/CD4(+)/CD68(+/-)/CD83(-)/CD25(-). These cells exhibited a remarkable proliferation rate (9.2 to 9.8% of all CD209(+) cells) by double staining with Ki67 and proliferating cell nuclear antigen. Unique within the DC-family, the majority of DC-SIGN(+) decidual APCs were observed in situ to have intimate contact with CD56(+)/CD16(-)/ICAM-3(+) decidual natural killer cells, another pregnancy-restricted cell population. In vitro, freshly isolated CD14(+)/DC-SIGN(+) decidual cells efficiently took up antigen, but could not stimulate naive allogeneic T cells at all. Treatment with an inflammatory cytokine cocktail resulted in down-regulation of antigen uptake capacity and evolving capacity to effectively stimulate resting T cells. Fluorescence-activated cell sorting analysis confirmed the maturation of CD14(+)/DC-SIGN(+) decidual cells into CD25(+)/CD83(+) mature DCs. In summary, this is the first identification of a uterine immature DC population expressing DC-SIGN, that appears only in pregnancy-associated tissue, has a high proliferation rate, and a conspicuous association with a natural killer subset.
Collapse
Affiliation(s)
- Ulrike Kämmerer
- Department of Obstetrics and Gynecology, the University Medical School Würzburg, Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Soilleux EJ, Coleman N. Transplacental transmission of HIV: a potential role for HIV binding lectins. Int J Biochem Cell Biol 2003; 35:283-7. [PMID: 12531239 DOI: 10.1016/s1357-2725(02)00132-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although the majority of vertical transmission of HIV occurs around the time of birth, 1.5-2% of pregnancies in HIV-positive women appear to result in the vertical transmission of HIV across the placenta. HIV infection of a number of placental cell types has been demonstrated, but the exact mechanisms of intrauterine vertical transmission remain obscure. The recent discovery of the HIV binding lectins dendritic cell-specific ICAM-grabbing non-integrin (DC-SIGN) and DC-SIGN-related molecule (DC-SIGNR) provides one possible explanation. Cells expressing these lectins are able to adsorb the virus and mediate high efficiency HIV infection of other cell types. Both lectins are expressed by the placenta, with DC-SIGN expression also being present on maternal cells intimately associated with the placenta. This review focuses on possible mechanisms by which these lectins may potentiate the intrauterine vertical transmission of HIV.
Collapse
Affiliation(s)
- Elizabeth J Soilleux
- Medical Research Council Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 2XY, UK.
| | | |
Collapse
|
93
|
Bashirova AA, Wu L, Cheng J, Martin TD, Martin MP, Benveniste RE, Lifson JD, KewalRamani VN, Hughes A, Carrington M. Novel member of the CD209 (DC-SIGN) gene family in primates. J Virol 2003; 77:217-27. [PMID: 12477827 PMCID: PMC140574 DOI: 10.1128/jvi.77.1.217-227.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two CD209 family genes identified in humans, CD209 (DC-SIGN) and CD209L (DC-SIGNR/L-SIGN), encode C-type lectins that serve as adhesion receptors for ICAM-2 and ICAM-3 and participate in the transmission of human and simian immunodeficiency viruses (HIV and SIV, respectively) to target cells in vitro. Here we characterize the CD209 gene family in nonhuman primates and show that recent evolutionary alterations have occurred in this family across primate species. All of the primate species tested, specifically, Old World monkeys (OWM) and apes, have orthologues of human CD209. In contrast, CD209L is missing in OWM but present in apes. A third family member, that we have named CD209L2, was cloned from rhesus monkey cDNA and subsequently identified in OWM and apes but not in humans. Rhesus CD209L2 mRNA was prominently expressed in the liver and axillary lymph nodes, although preliminary data suggest that levels of expression may vary among individuals. Despite a high level of sequence similarity to both human and rhesus CD209, rhesus CD209L2 was substantially less effective at binding ICAM-3 and poorly transmitted HIV type 1 and SIV to target cells relative to CD209. Our data suggest that the CD209 gene family has undergone recent evolutionary processes involving duplications and deletions, the latter of which may be tolerated because of potentially redundant functional activities of the molecules encoded by these genes.
Collapse
Affiliation(s)
- Arman A Bashirova
- Science Application International Corporation at Frederick, Laboratory of Genomic Diversity, National Cancer Institute at Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Soilleux EJ, Morris LS, Trowsdale J, Coleman N, Boyle JJ. Human atherosclerotic plaques express DC-SIGN, a novel protein found on dendritic cells and macrophages. J Pathol 2002; 198:511-6. [PMID: 12434421 DOI: 10.1002/path.1205] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The association of autoimmune phenomena with atherosclerosis suggests that plaques may contain specialized antigen-presenting cells, dendritic cells (DCs). DC-SIGN is a C-type lectin expressed by DCs. This study assessed whether human atherosclerotic plaques expressed DC-SIGN and several other macrophage/DC markers. Plaques from human coronary and carotid arteries and aorta contained DC-SIGN-immunoreactive cells. Double-labelling showed co-expression of DC-SIGN and macrophage/DC lineage markers CD14, CD68, HLA-DR, and S100. There was no immunoreactivity for the DC activation markers CD83 or CMRF-44. Since DC-SIGN mediates adhesion to T-lymphocytes and endocytosis, its expression in atherosclerotic plaques may have functional implications. Activated DCs migrate quickly from areas of inflammation to regional lymph nodes, possibly explaining the paucity of activated DCs in atherosclerotic plaques. In conclusion, this study has shown that DC-SIGN is expressed in atherosclerosis.
Collapse
Affiliation(s)
- E J Soilleux
- Medical Research Council Cancer Centre Unit, Hutchinson/MRC Research Centre, Cambridge, UK
| | | | | | | | | |
Collapse
|
95
|
Yu Kimata MT, Cella M, Biggins JE, Rorex C, White R, Hicks S, Wilson JM, Patel PG, Allan JS, Colonna M, Kimata JT. Capture and transfer of simian immunodeficiency virus by macaque dendritic cells is enhanced by DC-SIGN. J Virol 2002; 76:11827-36. [PMID: 12414925 PMCID: PMC136877 DOI: 10.1128/jvi.76.23.11827-11836.2002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2002] [Accepted: 08/26/2002] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DCs) are among the first cells encountered by human and simian immunodeficiency virus (HIV and SIV) following mucosal infection. Because these cells efficiently capture and transmit virus to T cells, they may play a major role in mediating HIV and SIV infection. Recently, a C-type lectin protein present on DCs, DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN), was shown to efficiently bind and present HIV and SIV to CD4(+), coreceptor-positive cells in trans. However, the significance of DC-SIGN for virus transmission and pathogenesis in vivo remains unclear. Because SIV infection of macaques may represent the best model to study the importance of DC-SIGN in HIV infection, we cloned and characterized pig-tailed macaque DC-SIGN and generated monoclonal antibodies (MAbs) against it. We demonstrate that, like human DC-SIGN, pig-tailed macaque DC-SIGN (ptDC-SIGN) is expressed on DCs and macrophages but not on monocytes, T cells, or B cells. Moderate levels of ptDC-SIGN expression were detected on the surface of DCs, and low-level expression was found on macrophages. Additionally, we show that ptDC-SIGN efficiently binds and transmits replication-competent SIVmne variants to CD4(+), coreceptor-positive cells. Moreover, transmission of virus between pig-tailed macaque DCs and CD4(+) T cells is largely ptDC-SIGN dependent. Interestingly, MAbs directed against ptDC-SIGN vary in the capacity to block transmission of different SIVmne variants. These data demonstrate that ptDC-SIGN plays a central role in transmitting virus from macaque DCs to T cells, and they suggest that SIVmne variants may differ in their interactions with ptDC-SIGN. Thus, SIVmne infection of pig-tailed macaques may provide an opportunity to investigate the significance of DC-SIGN in primate lentiviral infections.
Collapse
Affiliation(s)
- Monica T Yu Kimata
- Department of Virology and Immunology, Southwest Foundation for Biomedical Research, San Antonio, Texas 78227, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Gruber A, Chalmers AS, Popov S, Ruprecht RM. Functional aspects of binding of monoclonal antibody DCN46 to DC-SIGN on dendritic cells. Immunol Lett 2002; 84:103-8. [PMID: 12270546 DOI: 10.1016/s0165-2478(02)00144-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dendritic cell (DC)-specific ICAM-3 grabbing nonintegrin (DC-SIGN) is a DC-specific antigen that plays an important role in the induction of primary immune responses as well as during HIV infection. In the present study, we analyzed the effect of binding of monoclonal antibody DCN46 to DC surface, expressed DC-SIGN on DC function. DC-SIGN antibody treated, immature DC were able to differentiate into mature DC and had the same capacity as untreated DC to induce primary and secondary immune responses. In combination with flow cytometric cell sorting, DC-SIGN antibody treatment of DC yielded highly pure and functional DC. Given the apparent lack of functional effect of monoclonal antibody DCN46 on DC, the latter antibody may prove useful for research on and clinical use of highly pure and functional DC.
Collapse
Affiliation(s)
- Andreas Gruber
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 44 Binney Street, JFB-809, Boston, MA 02115-6084, USA.
| | | | | | | |
Collapse
|
97
|
Halary F, Amara A, Lortat-Jacob H, Messerle M, Delaunay T, Houlès C, Fieschi F, Arenzana-Seisdedos F, Moreau JF, Déchanet-Merville J. Human cytomegalovirus binding to DC-SIGN is required for dendritic cell infection and target cell trans-infection. Immunity 2002; 17:653-64. [PMID: 12433371 DOI: 10.1016/s1074-7613(02)00447-8] [Citation(s) in RCA: 286] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cytomegalovirus (CMV) infection is characterized by host immunosuppression and multiorganic involvement. CMV-infected dendritic cells (DC) were recently shown to display reduced immune functions, but their role in virus dissemination is not clear. In this report, we demonstrated that CMV could be captured by DC through binding on DC-SIGN and subsequently transmitted to permissive cells. Moreover, blocking DC-SIGN by specific antibodies inhibited DC infection by primary CMV isolates and expression of DC-SIGN or its homolog DC-SIGNR rendered susceptible cells permissive to CMV infection. We demonstrated that CMV envelope glycoprotein B is a viral ligand for DC-SIGN and DC-SIGNR. These results provide new insights into the molecular interactions contributing to cell infection by CMV and extend DC-SIGN implication in virus propagation.
Collapse
Affiliation(s)
- Franck Halary
- Laboratoire d'immunologie, CNRS UMR 5540, Université Bordeaux 2, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Santos PR, Petitprez K, Butor C. Gene for chinese rhesus macaque DC-SIGN predicts the existence of A but not B isoforms of the protein. AIDS Res Hum Retroviruses 2002; 18:977-81. [PMID: 12230940 DOI: 10.1089/088922202760265641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We report here the gene for DC-SIGN from Chinese rhesus macaques. DC-SIGN is a C-type lectin expressed by dendritic cells (DCs). It is involved in the interaction of DCs with T cells, and in transmission to T cells of HIV-1 and SIV. Alternative splicing in human DC-SIGN yields A and B isoforms of the protein. The overall organization of the rhesus macaque gene is similar to that of the human gene. Translation of B isoforms cannot occur because of a point substitution. The coding sequence shows that we have cloned a fourth allele for rhesus macaque DC-SIGN. This allele shows high homology to the other rhesus macaque alleles. However, at the protein level, the homology is highest with the pigtail macaque protein. This suggests a convergent evolution of DC-SIGN in macaques living in China. The importance of DC-SIGN variability in the immune response remains to be investigated.
Collapse
Affiliation(s)
- Patrícia R Santos
- Département d'Immunologie, Institut Cochin, INSERM U567, CNRS UMR8104, Université Paris V-René Descartes, 75014 Paris, France.
| | | | | |
Collapse
|
99
|
Baribaud F, Pöhlmann S, Leslie G, Mortari F, Doms RW. Quantitative expression and virus transmission analysis of DC-SIGN on monocyte-derived dendritic cells. J Virol 2002; 76:9135-42. [PMID: 12186897 PMCID: PMC136426 DOI: 10.1128/jvi.76.18.9135-9142.2002] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2002] [Accepted: 06/13/2002] [Indexed: 01/16/2023] Open
Abstract
The C-type lectins DC-SIGN and DC-SIGNR efficiently bind human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) strains and can transmit bound virus to adjacent CD4-positive cells. DC-SIGN also binds efficiently to the Ebola virus glycoprotein, enhancing Ebola virus infection. DC-SIGN is thought to be responsible for the ability of dendritic cells (DCs) to capture HIV and transmit it to T cells, thus promoting HIV dissemination in vitro and perhaps in vivo as well. To investigate DC-SIGN function and expression levels on DCs, we characterized a panel of monoclonal antibodies (MAbs) directed against the carbohydrate recognition domain of DC-SIGN. Using quantitative fluorescence-activated cell sorter technology, we found that DC-SIGN is highly expressed on immature monocyte-derived DCs, with at least 100,000 copies and often in excess of 250,000 copies per DC. There was modest variation (three- to fourfold) in DC-SIGN expression levels between individuals and between DCs isolated from the same individual at different times. Several MAbs efficiently blocked virus binding to cell lines expressing human or rhesus DC-SIGN, preventing HIV and SIV transmission. Interactions with Ebola virus pseudotypes were also blocked efficiently. Despite their ability to block virus-DC-SIGN interactions on cell lines, these antibodies only inhibited transmission of virus from DCs by approximately 50% or less. These results indicate that factors other than DC-SIGN may play important roles in the ability of DCs to capture and transmit HIV.
Collapse
Affiliation(s)
- Frédéric Baribaud
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
100
|
van Kooyk Y, Geijtenbeek TBH. A novel adhesion pathway that regulates dendritic cell trafficking and T cell interactions. Immunol Rev 2002; 186:47-56. [PMID: 12234361 DOI: 10.1034/j.1600-065x.2002.18605.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Dendritic cells (DC) are present in essentially every tissue, where they operate at the interface of innate and acquired immunity by recognizing pathogens and presenting pathogen-derived peptides to T cells. Cell-cell interactions between DC, T cells and endothelial cells are crucial to all immunological processes. Recently, several C-type lectin receptors have been characterized that are abundantly expressed on the surface of DC. It is now becoming clear that these lectin receptors serve not only as antigen-receptors recognizing pathogens, but they may also function as adhesion receptors and/or signaling molecules. In particular the DC specific C-type lectin DC-SIGN (CD209) regulates adhesion processes, such as DC trafficking by interacting with ICAM-2 and T cell synapse formation, upon binding of ICAM-3. C-type lectins such as DC-SIGN contain a lectin domain that recognizes in a Ca2+-dependent manner carbohydrates such as mannose-containing structures presented on the glycoproteins ICAM-2 and ICAM-3. Although the integrin LFA-1 is a counter-receptor for both ICAM-2 and ICAM-3, on DC, DC-SIGN is the high affinity adhesion receptor for ICAM-2/-3. Here we discuss how the heterogeneity of mannose-residues exposed on cellular proteins and pathogens regulates specific binding of a repertoire of DC-expressed C-type lectins that contribute to the diversity of immune responses created by DC.
Collapse
Affiliation(s)
- Yvette van Kooyk
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|