Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Plant Biotechnol J ; 12(8): 1098-107, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24975464

ABSTRACT

Previously, our group engineered a plant-derived monoclonal antibody (MAb pE16) that efficiently treated West Nile virus (WNV) infection in mice. In this study, we developed a pE16 variant consisting of a single-chain variable fragment (scFv) fused to the heavy chain constant domains (CH) of human IgG (pE16scFv-CH). pE16 and pE16scFv-CH were expressed and assembled efficiently in Nicotiana benthamiana ∆XF plants, a glycosylation mutant lacking plant-specific N-glycan residues. Glycan analysis revealed that ∆XF plant-derived pE16scFv-CH (∆XFpE16scFv-CH) and pE16 (∆XFpE16) both displayed a mammalian glycosylation profile. ∆XFpE16 and ∆XFpE16scFv-CH demonstrated equivalent antigen-binding affinity and kinetics, and slightly enhanced neutralization of WNV in vitro compared with the parent mammalian cell-produced E16 (mE16). A single dose of ∆XFpE16 or ∆XFpE16scFv-CH protected mice against WNV-induced mortality even 4 days after infection at equivalent rates as mE16. This study provides a detailed tandem comparison of the expression, structure and function of a therapeutic MAb and its single-chain variant produced in glycoengineered plants. Moreover, it demonstrates the development of anti-WNV MAb therapeutic variants that are equivalent in efficacy to pE16, simpler to produce, and likely safer to use as therapeutics due to their mammalian N-glycosylation. This platform may lead to a more robust and cost-effective production of antibody-based therapeutics against WNV infection and other infectious, inflammatory or neoplastic diseases.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Nicotiana/metabolism , Single-Chain Antibodies/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antibodies, Monoclonal/immunology , Gene Expression , Glycosylation , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests , Plantibodies/immunology , Surface Plasmon Resonance , Viral Envelope Proteins/immunology
2.
Virol J ; 7: 95, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20462412

ABSTRACT

Antibodies generated against West Nile virus (WNV) during infection are essential for controlling dissemination. Recent studies have demonstrated that epitopes in all three domains of the flavivirus envelope protein (E) are targets for neutralizing antibodies, with determinants in domain III (DIII) eliciting antibodies with strong inhibitory properties. In order to increase the magnitude and quality of the antibody response against the WNV E protein, DNA vaccines with derivatives of the WNV E gene (full length E, truncated E, or DIII region, some in the context of the pre-membrane [prM] gene) were conjugated to the molecular adjuvant P28. The P28 region of the complement protein C3d is the minimum CR2-binding domain necessary for the adjuvant activity of C3d. Delivery of DNA-based vaccines by gene gun and intramuscular routes stimulated production of IgG antibodies against the WNV DIII region of the E protein. With the exception of the vaccine expressing prM/E given intramuscularly, only mice that received DNA vaccines by gene gun produced protective neutralizing antibody titers (FRNT80 titer >1/40). Correspondingly, mice vaccinated by the gene gun route were protected to a greater level from lethal WNV challenge. In general, mice vaccinated with P28-adjuvated vaccines produced higher IgG titers than mice vaccinated with non-adjuvanted vaccines.


Subject(s)
Complement C3d/chemistry , Complement C3d/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Chlorocebus aethiops , Complement C3d/administration & dosage , Complement C3d/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Vero Cells , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , West Nile Fever/virology , West Nile virus/chemistry , West Nile virus/genetics
3.
PLoS Pathog ; 6(2): e1000790, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20168989

ABSTRACT

Immunity to one of the four dengue virus (DV) serotypes can increase disease severity in humans upon subsequent infection with another DV serotype. Serotype cross-reactive antibodies facilitate DV infection of myeloid cells in vitro by promoting virus entry via Fcgamma receptors (FcgammaR), a process known as antibody-dependent enhancement (ADE). However, despite decades of investigation, no in vivo model for antibody enhancement of dengue disease severity has been described. Analogous to human infants who receive anti-DV antibodies by transplacental transfer and develop severe dengue disease during primary infection, we show here that passive administration of anti-DV antibodies is sufficient to enhance DV infection and disease in mice using both mouse-adapted and clinical DV isolates. Antibody-enhanced lethal disease featured many of the hallmarks of severe dengue disease in humans, including thrombocytopenia, vascular leakage, elevated serum cytokine levels, and increased systemic viral burden in serum and tissue phagocytes. Passive transfer of a high dose of serotype-specific antibodies eliminated viremia, but lower doses of these antibodies or cross-reactive polyclonal or monoclonal antibodies all enhanced disease in vivo even when antibody levels were neutralizing in vitro. In contrast, a genetically engineered antibody variant (E60-N297Q) that cannot bind FcgammaR exhibited prophylactic and therapeutic efficacy against ADE-induced lethal challenge. These observations provide insight into the pathogenesis of antibody-enhanced dengue disease and identify a novel strategy for the design of therapeutic antibodies against dengue.


Subject(s)
Antibodies, Viral/immunology , Antibody-Dependent Enhancement/immunology , Dengue/immunology , Immunoglobulin Fc Fragments/immunology , Animals , Antibodies, Neutralizing/immunology , Cell Separation , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunization, Passive , Immunohistochemistry , Mice , Neutralization Tests , Surface Plasmon Resonance , Viral Load
4.
Cell Host Microbe ; 6(4): 381-91, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19837377

ABSTRACT

Virus neutralization is governed by the number of antibodies that bind a virion during the cellular entry process. Cellular and serum factors that interact with antibodies have the potential to modulate neutralization potency. Although the addition of serum complement can increase the neutralizing activity of antiviral antibodies in vitro, the mechanism and significance of this augmented potency in vivo remain uncertain. Herein, we show that the complement component C1q increases the potency of antibodies against West Nile virus by modulating the stoichiometric requirements for neutralization. The addition of C1q does not result in virolysis but instead reduces the number of antibodies that must bind the virion to neutralize infectivity. For IgG subclasses that bind C1q avidly, this reduced stoichiometric threshold falls below the minimal number of antibodies required for antibody-dependent enhancement (ADE) of infection of cells expressing Fc-gamma receptors (CD32) and explains how C1q restricts the ADE of flavivirus infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Complement C1q/immunology , West Nile virus/immunology , Animals , Cell Line , Humans , Immunoglobulin G/immunology , Neutralization Tests
5.
Virology ; 393(1): 11-5, 2009 Oct 10.
Article in English | MEDLINE | ID: mdl-19744691

ABSTRACT

Although the interactions of complement and viruses have been widely studied, the function of C5 and the membrane attack complex in the context of viral infection or antibody-mediated neutralization remains controversial. Using C5-depleted or -deficient human or mouse sera, we show that C5 does not contribute to the antibody-dependent or -independent neutralization of West Nile virus (WNV) in cell culture. Consistent with this, C5 neither contributed to protection against WNV pathogenesis nor augmented the neutralizing efficacy of complement-fixing anti-WNV neutralizing antibodies in mice. Although previous studies established that activation of the classical, lectin, and alternative complement pathways restricts WNV infection, our results show little effect of C5 and by inference the terminal lytic complement components. Overall, these results enhance our mechanistic understanding of how complement controls flavivirus infections.


Subject(s)
Complement C5/immunology , West Nile Fever/immunology , West Nile Fever/pathology , West Nile virus/immunology , West Nile virus/pathogenicity , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Humans , Male , Mice , Neutralization Tests , Survival Analysis , Viral Plaque Assay
6.
Front Biosci (Landmark Ed) ; 14(8): 3024-34, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273254

ABSTRACT

West Nile encephalitis virus (WNV) is a small, enveloped, mosquito-transmitted, positive-polarity RNA virus of the Flaviviridae family. This virus is closely related to other arthropod-borne viruses that cause human disease including Dengue, Yellow fever, and Japanese encephalitis viruses. WNV cycles in nature between mosquitoes and birds, but also infects human, horses, and other vertebrates. In humans, WNV disseminates to the central nervous system (CNS) and causes severe disease primarily in the immunocompromised and elderly. Experimental studies have made significant progress in dissecting the viral and host factors that determine the pathogenesis and outcome of WNV infection. This review will focus on the interactions between WNV and the protective and pathogenic host immune responses.


Subject(s)
West Nile Fever/immunology , West Nile virus/immunology , Adaptation, Physiological , Aged , Animals , Central Nervous System/virology , Humans , Immunity, Innate
7.
Vaccine ; 26 Suppl 8: I100-7, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19388173

ABSTRACT

The complement system is a family of serum and cell surface proteins that recognize pathogen-associated molecular patterns, altered-self ligands, and immune complexes. Activation of the complement cascade triggers several antiviral functions including pathogen opsonization and/or lysis, and priming of adaptive immune responses. In this review, we will examine the role of complement activation in protection and/or pathogenesis against infection by Flaviviruses, with an emphasis on experiments with West Nile and Dengue viruses.


Subject(s)
Complement System Proteins/physiology , Flavivirus Infections/immunology , Complement Activation , Flavivirus Infections/etiology , Humans , Immunity, Innate
8.
Cell Host Microbe ; 2(6): 417-26, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18078693

ABSTRACT

Severe dengue virus infection can occur in humans with pre-existing antibodies against the virus. This observation led to the hypothesis that a subneutralizing antibody level in vivo can increase viral burden and cause more severe disease. Indeed, antibody-dependent enhancement of infection (ADE) in vitro has been described for multiple viruses, including the flaviviruses dengue virus and West Nile virus. Here, we demonstrate that the complement component C1q restricts ADE by anti-flavivirus IgG antibodies in an IgG subclass-specific manner in cell culture and in mice. IgG subclasses that avidly bind C1q induced minimal ADE in the presence of C1q. These findings add a layer of complexity for the analysis of humoral immunity and flavivirus infection.


Subject(s)
Antibodies, Viral/immunology , Complement C1q/immunology , Flavivirus Infections/immunology , Flavivirus/immunology , Immunoglobulin G/immunology , Animals , Antibody Specificity , Flavivirus/pathogenicity , Flavivirus Infections/virology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virulence
9.
J Exp Med ; 203(5): 1371-81, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16651386

ABSTRACT

West Nile virus (WNV) causes a severe infection of the central nervous system in several vertebrate animals including humans. Prior studies have shown that complement plays a critical role in controlling WNV infection in complement (C) 3(-/-) and complement receptor 1/2(-/-) mice. Here, we dissect the contributions of the individual complement activation pathways to the protection from WNV disease. Genetic deficiencies in C1q, C4, factor B, or factor D all resulted in increased mortality in mice, suggesting that all activation pathways function together to limit WNV spread. In the absence of alternative pathway complement activation, WNV disseminated into the central nervous system at earlier times and was associated with reduced CD8+ T cell responses yet near normal anti-WNV antibody profiles. Animals lacking the classical and lectin pathways had deficits in both B and T cell responses to WNV. Finally, and somewhat surprisingly, C1q was required for productive infection in the spleen but not for development of adaptive immune responses after WNV infection. Our results suggest that individual pathways of complement activation control WNV infection by priming adaptive immune responses through distinct mechanisms.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Complement Activation/immunology , Complement System Proteins/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Complement System Proteins/deficiency , Humans , Mice , Mice, Knockout , Spleen/immunology , Spleen/pathology , Spleen/virology , West Nile Fever/pathology , West Nile Fever/virology
10.
J Virol ; 79(12): 7466-77, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919902

ABSTRACT

Infection with West Nile virus (WNV) causes a severe infection of the central nervous system (CNS) with higher levels of morbidity and mortality in the elderly and the immunocompromised. Experiments with mice have begun to define how the innate and adaptive immune responses function to limit infection. Here, we demonstrate that the complement system, a major component of innate immunity, controls WNV infection in vitro primarily in an antibody-dependent manner by neutralizing virus particles in solution and lysing WNV-infected cells. More decisively, mice that genetically lack the third component of complement or complement receptor 1 (CR1) and CR2 developed increased CNS virus burdens and were vulnerable to lethal infection at a low dose of WNV. Both C3-deficient and CR1- and CR2-deficient mice also had significant deficits in their humoral responses after infection with markedly reduced levels of specific anti-WNV immunoglobulin M (IgM) and IgG. Overall, these results suggest that complement controls WNV infection, in part through its ability to induce a protective antibody response.


Subject(s)
Antibodies, Viral/blood , Complement Activation , West Nile Fever/immunology , West Nile virus/immunology , West Nile virus/pathogenicity , Animals , Antibodies, Viral/immunology , Brain/immunology , Brain/virology , Cell Line , Complement C3/genetics , Cricetinae , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , Neutralization Tests , Receptors, Complement/genetics , West Nile Fever/virology
11.
Nat Med ; 11(5): 522-30, 2005 May.
Article in English | MEDLINE | ID: mdl-15852016

ABSTRACT

Neutralization of West Nile virus (WNV) in vivo correlates with the development of an antibody response against the viral envelope (E) protein. Using random mutagenesis and yeast surface display, we defined individual contact residues of 14 newly generated monoclonal antibodies against domain III of the WNV E protein. Monoclonal antibodies that strongly neutralized WNV localized to a surface patch on the lateral face of domain III. Convalescent antibodies from individuals who had recovered from WNV infection also detected this epitope. One monoclonal antibody, E16, neutralized 10 different strains in vitro, and showed therapeutic efficacy in mice, even when administered as a single dose 5 d after infection. A humanized version of E16 was generated that retained antigen specificity, avidity and neutralizing activity. In postexposure therapeutic trials in mice, a single dose of humanized E16 protected mice against WNV-induced mortality, and may therefore be a viable treatment option against WNV infection in humans.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotherapy , Viral Envelope Proteins/immunology , West Nile Fever/therapy , West Nile virus/immunology , Animals , Cloning, Molecular , DNA Primers , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Mice , Mice, Inbred C57BL , Mutagenesis , Neutralization Tests , Plasmids/genetics , Viral Envelope Proteins/metabolism , West Nile Fever/immunology , Yeasts
12.
Viral Immunol ; 16(3): 259-78, 2003.
Article in English | MEDLINE | ID: mdl-14583143

ABSTRACT

WNV continues to spread throughout the Western Hemisphere as virus activity in insects and animals has been reported in the United States, Canada, Mexico, and the Caribbean islands. West Nile virus (WNV) infects the central nervous system and causes severe disease primarily in humans who are immunocompromised or elderly. In this review, we discuss the mechanisms by which the immune system limits dissemination of WNV infection. Recent experimental studies in animals suggest important roles for both the innate and the adaptive immune responses in controlling WNV infection. Interferons, antibody, complement components and CD8+ T cells coordinate protection against severe infection and disease. These findings are analyzed in the context of recent approaches to vaccine development and immunotherapy against WNV.


Subject(s)
West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antibodies, Viral/biosynthesis , Complement Activation , Dendritic Cells/immunology , Humans , Immunity, Cellular , Immunity, Innate , Immunotherapy , In Vitro Techniques , Interferons/biosynthesis , Killer Cells, Natural/immunology , Macrophages/immunology , Models, Immunological , T-Lymphocytes/immunology , Viral Vaccines/isolation & purification , West Nile Fever/therapy , West Nile virus/growth & development , West Nile virus/pathogenicity
13.
J Acquir Immune Defic Syndr ; 34(1): 7-19, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14501788

ABSTRACT

Interactions between HIV-1 and dendritic cells (DCs) play an important role in the initial establishment and spread of infection and development of antiviral immunity. We used chemically inactivated aldrithiol-2 (AT-2) simian immunodeficiency virus (SIV) with functional envelope glycoproteins to study virus interactions with DCs and developed an in vitro system to evaluate the quality of SIV antigen (Ag) presentation by DCs to T cells. AT-2 SIV interacts authentically with T cells and DCs and thus allows assessment of natural SIV-specific responses. CD4+ and CD8+ T cells from blood or lymph nodes of SIV-infected macaques released interferon-gamma (IFN gamma) and proliferated in response to a variety of AT-2 SIV isolates. Responses did not vary significantly as a function of the quantitative envelope glycoprotein content of the virions. Presentation of Ags derived from AT-2 SIV by DCs was more potent than presentation by comparably Ag-loaded monocytes. Interestingly, SIV-pulsed mature DCs stimulated both CD4+ and CD8+ T-cell responses, whereas immature DCs primarily stimulated CD4+ T cells. Further studies using AT-2 inactivated virus may help to define better the details of the virus-DC interactions critical for infection versus induction of antiviral immune responses.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Simian Immunodeficiency Virus/immunology , 2,2'-Dipyridyl/pharmacology , Animals , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/virology , Disulfides/pharmacology , Female , Leukocytes, Mononuclear/immunology , Lymph Nodes/immunology , Macaca mulatta , Male , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology , Vaccines, Inactivated/immunology
14.
J Immunol ; 169(8): 4172-82, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370346

ABSTRACT

Immature dendritic cells (DCs), unlike mature DCs, require the viral determinant nef to drive immunodeficiency virus (SIV and HIV) replication in coculture with CD4(+) T cells. Since immature DCs may capture and get infected by virus during mucosal transmission, we hypothesized that Nef associated with the virus or produced during early replication might modulate DCs to augment virus dissemination. Adenovirus vectors expressing nef were used to introduce nef into DCs in the absence of other immunodeficiency virus determinants to examine Nef-induced changes that might activate immature DCs to acquire properties of mature DCs and drive virus replication. Nef expression by immature human and macaque DCs triggered IL-6, IL-12, TNF-alpha, CXCL8, CCL3, and CCL4 release, but without up-regulating costimulatory and other molecules characteristic of mature DCs. Coincident with this, nef-expressing immature DCs stimulated stronger autologous CD4(+) T cell responses. Both SIV and HIV nef-expressing DCs complemented defective SIVmac239 delta nef, driving replication in autologous immature DC-T cell cultures. In contrast, if DCs were activated after capturing delta nef, virus growth was not exacerbated. This highlights one way in which nef-defective virus-bearing immature DCs that mature while migrating to draining lymph nodes could induce stronger immune responses in the absence of overwhelming productive infection (unlike nef-containing wild-type virus). Therefore, Nef expressed in immature DCs signals a distinct activation program that promotes virus replication and T cell recruitment but without complete DC maturation, thereby lessening the likelihood that wild-type virus-infected immature DCs would activate virus-specific immunity, but facilitating virus dissemination.


Subject(s)
Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/virology , Gene Products, nef/biosynthesis , Gene Products, nef/physiology , Immunophenotyping , Adenoviridae/immunology , Animals , Cell Differentiation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , HIV-1/immunology , Humans , Lymphocyte Activation , Macaca mulatta , Male , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication/immunology , nef Gene Products, Human Immunodeficiency Virus
15.
J Immunol Methods ; 260(1-2): 219-34, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11792391

ABSTRACT

The macaque-simian immunodeficiency virus (SIV) system is one of the best animal models available to study the role of dendritic cells (DCs) in transmission and pathogenesis of HIV, as well as to test DC-based vaccine and therapeutic strategies. To better define and optimize this system, the responsiveness of macaque monocyte-derived DCs to a variety of maturation stimuli was examined. Characteristic immunophenotypic and functional DC maturation induced by standard monocyte conditioned medium (MCM) was compared to the activation induced by a panel of stimuli including soluble CD40L, LPS, Poly I:C, PGE(2)/TNFalpha, and a cocktail mixture of PGE(2)/TNFalpha/IL-1beta/IL-6. Immunophenotypic analysis confirmed that all stimuli induced stable up-regulation of CD25, CD40, CD80, CD83, CD86, HLA-DR, DC-LAMP (CD208), and DEC-205 (CD205). In general, macaque DCs exhibited weaker responses to LPS and Poly I:C than human DCs, and soluble CD40L stimulation induced variable expression of CD25. Interestingly, while the endocytic capacity of CD40L-matured cells was down-modulated comparably to DCs matured with MCM or the cocktail, the T cell stimulatory activity was not enhanced to the same extent. The particularly reproducible and potent T cell stimulatory capacity of cocktail-treated DCs correlated with a more homogenous mature DC phenotype, consistently high levels of IL-12 production, and better viability upon reculture compared to DCs activated by other stimuli. Furthermore, cocktail-matured DCs efficiently captured and presented inactivated SIV to SIV-primed T cells in vitro. Thus, the cocktail represents a particularly potent and useful stimulus for the generation of efficacious immunostimulatory macaque DCs.


Subject(s)
Dendritic Cells/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Biological Assay , Humans , Immunity, Cellular , Immunophenotyping , Macaca mulatta , T-Lymphocytes/immunology
16.
Proc Natl Acad Sci U S A ; 99(3): 1568-73, 2002 Feb 05.
Article in English | MEDLINE | ID: mdl-11818554

ABSTRACT

Here, we describe the isolation and characterization of the rhesus macaque homolog for human DC-SIGN, a dendritic cell-specific C-type lectin. mac-DC-SIGN is 92% identical to hu-DC-SIGN. mac-DC-SIGN preserves the virus transmission function of hu-DC-SIGN, capturing and efficiently transducing simian and human immunodeficiency virus to target CD4(+) T cells. Surprisingly, however, mac-DC-SIGN plays no discernable role in the ability of rhesus macaque dendritic cells to capture and transmit primate lentiviruses. Expression and neutralization analyses suggest that this process is DC-SIGN independent in macaque, although the participation of other lectin molecules cannot be ruled out. The ability of primate lentiviruses to effectively use human and rhesus dendritic cells in virus transmission without the cells becoming directly infected suggests that these viruses have taken advantage of a conserved dendritic cell mechanism in which DC-SIGN family molecules are significant contributors but not the only participants.


Subject(s)
Antigens, CD , Antigens, Differentiation , Dendritic Cells/virology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/physiology , Acquired Immunodeficiency Syndrome/transmission , Acquired Immunodeficiency Syndrome/virology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cell Adhesion , Cell Adhesion Molecules/physiology , Cell Line , Cells, Cultured , Conserved Sequence , Flow Cytometry , HIV-1/physiology , Humans , Macaca mulatta , Molecular Sequence Data , RNA, Viral/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Simian Acquired Immunodeficiency Syndrome/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...