Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Heart Lung Transplant ; 41(8): 1044-1054, 2022 08.
Article in English | MEDLINE | ID: mdl-35691796

ABSTRACT

BACKGROUND: Long-term survival of lung transplants lags behind other solid organs due to early onset of a fibrotic form of chronic rejection known as chronic lung allograft dysfunction (CLAD). Preventing CLAD is difficult as multiple immunologic and physiologic insults contribute to its development. Targeting fibroblast activation, which is the final common pathway leading to CLAD, offers the opportunity to ameliorate fibrosis irrespective of the initiating insult. Thy-1 is a surface glycoprotein that controls fibroblast differentiation and activation. METHODS: To study the role of Thy-1 in CLAD, we utilized the minor antigen mismatched C57BL/6 (B6wild-type) or B6Thy-1-/-→C57BL/10 (B10) model of murine orthotopic lung transplantation with postoperative bacterial infection modeled by intratracheal lipopolysaccharide (LPS) administration. The effects of LPS on Thy-1 expression, proliferation, and gene expression were assessed in fibroblasts in vitro and the therapeutic potential of Thy-1 replacement was assessed in vivo. RESULTS: More severe CLAD was evident in B6Thy-1-/- →B10 grafts compared to B6wild-type →B10 grafts. LPS further accentuated fibrosis in B6wild-type →B10 grafts with some, but limited, effects on B6Thy-1-/- →B10 grafts. LPS contributed to Thy-1 loss from Thy-1(+) fibroblasts in vitro due to a decrease in mRNA expression. In addition, LPS promoted proliferation and upregulation of multiple inflammatory pathways in Thy-1(-) fibroblasts by gene expression analysis. Most importantly, replacement of Thy-1 through exogenous administration ameliorated the fibrotic phenotype post-LPS mediated modeling of infection. CONCLUSIONS: Our findings suggest that the loss of Thy-1 on fibroblasts is a previously unrecognized cause of CLAD and its replacement may offer therapeutic applications for amelioration of this disease post-transplantation in the setting of infectious stress responses.


Subject(s)
Lipopolysaccharides , Lung Transplantation , Allografts , Animals , Fibrosis , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Mice , Mice, Inbred C57BL , Stromal Cells
2.
JCI Insight ; 7(10)2022 05 23.
Article in English | MEDLINE | ID: mdl-35603788

ABSTRACT

Cytokine therapy is limited by undesirable off-target side effects as well as terminal differentiation and exhaustion of chronically stimulated T cells. Here, we describe the signaling properties of a potentially unique cytokine by design, where T cell surface binding and signaling are separated between 2 different families of receptors. This fusion protein cytokine, called OMCPmutIL-2, bound with high affinity to the cytotoxic lymphocyte-defining immunoreceptor NKG2D but signaled through the common γ chain cytokine receptor. In addition to precise activation of cytotoxic T cells due to redirected binding, OMCPmutIL-2 resulted in superior activation of both human and murine CD8+ T cells by improving their survival and memory cell generation and decreasing exhaustion. This functional improvement was the direct result of altered signal transduction based on the reorganization of surface membrane lipid rafts that led to Janus kinase-3-mediated phosphorylation of the T cell receptor rather than STAT/AKT signaling intermediates. This potentially novel signaling pathway increased CD8+ T cell response to low-affinity antigens, activated nuclear factor of activated T cells transcription factors, and promoted mitochondrial biogenesis. OMCPmutIL-2 thus outperformed other common γ chain cytokines as a catalyst for in vitro CD8+ T cell expansion and in vivo CD8+ T cell-based immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Cytokines , Animals , Cytokines/metabolism , Humans , Immunotherapy , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Cytokine/metabolism
3.
J Immunol ; 207(1): 333-343, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34155069

ABSTRACT

Ex vivo expansion followed by reinfusion of tumor-infiltrating leukocytes (TILs) has been used successfully for the treatment of multiple malignancies. Most protocols rely on the use of the cytokine IL-2 to expand TILs prior to reinfusion. In addition, TIL administration relies on systemic administration of IL-2 after reinfusion to support transferred cell survival. The use of IL-2, however, can be problematic because of its preferential expansion of regulatory T and myeloid cells as well as its systemic side effects. In this study, we describe the use of a novel IL-2 mutant retargeted to NKG2D rather than the high-affinity IL-2R for TIL-mediated immunotherapy in a murine model of malignant melanoma. We demonstrate that the NKG2D-retargeted IL-2 (called OMCPmutIL-2) preferentially expands TIL-resident CTLs, such as CD8+ T cells, NK cells, and γδT cells, whereas wild-type IL-2 provides a growth advantage for CD4+Foxp3+ T cells as well as myeloid cells. OMCPmutIL-2-expanded CTLs express higher levels of tumor-homing receptors, such as LFA-1, CD49a, and CXCR3, which correlate with TIL localization to the tumor bed after i.v. injection. Consistent with this, OMCPmutIL-2-expanded TILs provided superior tumor control compared with those expanded in wild-type IL-2. Our data demonstrate that adoptive transfer immunotherapy can be improved by rational retargeting of cytokine signaling to NKG2D-expressing CTLs rather than indiscriminate expansion of all TILs.


Subject(s)
Adoptive Transfer , Interleukin-2/immunology , Leukocytes/immunology , Melanoma/immunology , Melanoma/therapy , NK Cell Lectin-Like Receptor Subfamily K/genetics , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/immunology , Signal Transduction/immunology
4.
Biochem Mol Biol Educ ; 49(1): 115-128, 2021 01.
Article in English | MEDLINE | ID: mdl-33176069

ABSTRACT

The rapid development of molecular biotechnology presents a curricular challenge for educators trying to provide students with relevant coursework. A comprehensive biology education should also include opportunities for students to develop intellectual and technical skills through authentic research experiences. Integrating relevant and interesting research projects into their classes, however, can be a challenging task for instructors. To address these varied demands, we redesigned our existing molecular cloning course to incorporate an independent research project assessing calcium signaling. In the revised course, students use traditional and recombination-based cloning strategies to generate bacterial and mammalian expression vectors encoding CaMPARI, a novel fluorescent calcium indicator. Bacterially-expressed CaMPARI is used in protein quantification and purification assays. Students must also design their own research project evaluating the effect of chemotherapeutic agents on calcium signaling in a mammalian system. Revised and novel labs were designed to be modular, facilitating their integration into the course over 2 years. End-of-semester student evaluations were compared between years revealing a significant difference in students' perception of the course's difficulty between years. This change in attitude highlights potential pedagogical considerations that must be examined when introducing new material and activities into existing courses. Since calcium signaling is important for cellular process across diverse species, instructors may be able to develop research projects within their respective areas of interest. Integration of authentic research experiences into the curriculum is challenging; however, the framework described here provides a versatile structure that can be adapted to merge diverse instructor interests with evolving educational needs.


Subject(s)
Biotechnology/education , Research , Animals , Calcium/metabolism , Cloning, Molecular , Curriculum , Fluorescent Dyes/chemistry , Humans , Proteins/analysis , Students
5.
Elife ; 72018 12 21.
Article in English | MEDLINE | ID: mdl-30575523

ABSTRACT

A recurrent theme in viral immune evasion is the sabotage of MHC-I antigen presentation, which brings virus the concomitant issue of 'missing-self' recognition by NK cells that use inhibitory receptors to detect surface MHC-I proteins. Here, we report that rodent herpesvirus Peru (RHVP) encodes a Qa-1 like protein (pQa-1) via RNA splicing to counteract NK activation. While pQa-1 surface expression is stabilized by the same canonical peptides presented by murine Qa-1, pQa-1 is GPI-anchored and resistant to the activity of RHVP pK3, a ubiquitin ligase that targets MHC-I for degradation. pQa-1 tetramer staining indicates that it recognizes CD94/NKG2A receptors. Consistently, pQa-1 selectively inhibits NKG2A+ NK cells and expression of pQa-1 can protect tumor cells from NK control in vivo. Collectively, these findings reveal an innovative NK evasion strategy wherein RHVP encodes a modified Qa-1 mimic refractory to MHC-I sabotage and capable of specifically engaging inhibitory receptors to circumvent NK activation.


Subject(s)
Cytotoxicity, Immunologic/immunology , Herpesviridae/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Base Sequence , Cytotoxicity, Immunologic/genetics , HEK293 Cells , Herpesviridae/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Mimicry/genetics , Molecular Mimicry/immunology , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Protein Binding/immunology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
6.
J Virol ; 92(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30282715

ABSTRACT

HSV virus-cell and cell-cell fusion requires multiple interactions between four essential virion envelope glycoproteins, gD, gB, gH, and gL, and between gD and a cellular receptor, nectin-1 or herpesvirus entry mediator (HVEM). Current models suggest that binding of gD to receptors induces a conformational change that leads to activation of gH/gL and consequent triggering of the prefusion form of gB to promote membrane fusion. Since protein-protein interactions guide each step of fusion, identifying the sites of interaction may lead to the identification of potential therapeutic targets that block this process. We have previously identified two "faces" on gD: one for receptor binding and the other for its presumed interaction with gH/gL. We previously separated the gD monoclonal antibodies (MAbs) into five competition communities. MAbs from two communities (MC2 and MC5) neutralize virus infection and block cell-cell fusion but do not block receptor binding, suggesting that they block binding of gD to gH/gL. Using a combination of classical epitope mapping of gD mutants with fusion and entry assays, we identified two residues (R67 and P54) on the presumed gH/gL interaction face of gD that allowed for fusion and viral entry but were no longer sensitive to inhibition by MC2 or MC5, yet both were blocked by other MAbs. As neutralizing antibodies interfere with essential steps in the fusion pathway, our studies strongly suggest that these key residues block the interaction of gD with gH/gL.IMPORTANCE Virus entry and cell-cell fusion mediated by HSV require gD, gH/gL, gB, and a gD receptor. Neutralizing antibodies directed against any of these proteins bind to residues within key functional sites and interfere with an essential step in the fusion pathway. Thus, the epitopes of these MAbs identify critical, functional sites on their target proteins. Unlike many anti-gD MAbs, which block binding of gD to a cellular receptor, two, MC2 and MC5, block a separate, downstream step in the fusion pathway which is presumed to be the activation of the modulator of fusion, gH/gL. By combining epitope mapping of a panel of gD mutants with fusion and virus entry assays, we have identified residues that are critical in the binding and function of these two MAbs. This new information helps to define the site of the presumptive interaction of gD with gH/gL, of which we have limited knowledge.


Subject(s)
Antibodies, Neutralizing/pharmacology , Simplexvirus/physiology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/pharmacology , Binding Sites/drug effects , Cell Line , Chlorocebus aethiops , Epitope Mapping , Mice , Models, Molecular , Protein Binding , Protein Conformation , Vero Cells , Viral Envelope Proteins/genetics , Virus Internalization/drug effects
7.
Oncoimmunology ; 6(2): e1265721, 2017.
Article in English | MEDLINE | ID: mdl-28344875

ABSTRACT

The use of high-dose interleukin-2 (IL-2) has fallen out of favor due to severe life-threatening side effects. We have recently described a unique way of directly targeting IL-2 to cytotoxic lymphocytes using a virally encoded immune evasion protein and an IL-2 mutant that avoids off-target side effects such as activation of regulatory T cells and vascular endothelium.

8.
Mol Cell Proteomics ; 16(4 suppl 1): S263-S276, 2017 04.
Article in English | MEDLINE | ID: mdl-28237943

ABSTRACT

Human cytomegalovirus (HCMV) is a significant cause of disease in immune-compromised adults and immune naïve newborns. No vaccine exists to prevent HCMV infection, and current antiviral therapies have toxic side effects that limit the duration and intensity of their use. There is thus an urgent need for new strategies to treat HCMV infection. Repurposing existing drugs as antivirals is an attractive approach to limit the time and cost of new antiviral drug development. Virus-induced changes in infected cells are often driven by changes in cellular kinase activity, which led us to hypothesize that defining the complement of kinases (the kinome), whose abundance or expression is altered during infection would identify existing kinase inhibitors that could be repurposed as new antivirals. To this end, we applied a kinase capture technique, multiplexed kinase inhibitor bead-mass spectrometry (MIB-MS) kinome, to quantitatively measure perturbations in >240 cellular kinases simultaneously in cells infected with a laboratory-adapted (AD169) or clinical (TB40E) HCMV strain. MIB-MS profiling identified time-dependent increases and decreases in MIB binding of multiple kinases including cell cycle kinases, receptor tyrosine kinases, and mitotic kinases. Based on the kinome data, we tested the antiviral effects of kinase inhibitors and other compounds, several of which are in clinical use or development. Using a novel flow cytometry-based assay and a fluorescent reporter virus we identified three compounds that inhibited HCMV replication with IC50 values of <1 µm, and at doses that were not toxic to uninfected cells. The most potent inhibitor of HCMV replication was OTSSP167 (IC50 <1.2 nm), a MELK inhibitor, blocked HCMV early gene expression and viral DNA accumulation, resulting in a >3 log decrease in virus replication. These results show the utility of MIB-MS kinome profiling for identifying existing kinase inhibitors that can potentially be repurposed as novel antiviral drugs.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Protein Kinase Inhibitors/pharmacology , Cells, Cultured , Cytomegalovirus/metabolism , Drug Repositioning , Humans , Mass Spectrometry/methods , Structure-Activity Relationship , Virus Replication/drug effects
9.
Nat Commun ; 7: 12878, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27650575

ABSTRACT

Despite over 20 years of clinical use, IL-2 has not fulfilled expectations as a safe and effective form of tumour immunotherapy. Expression of the high affinity IL-2Rα chain on regulatory T cells mitigates the anti-tumour immune response and its expression on vascular endothelium is responsible for life threatening complications such as diffuse capillary leak and pulmonary oedema. Here we describe the development of a recombinant fusion protein comprised of a cowpox virus encoded NKG2D binding protein (OMCP) and a mutated form of IL-2 with poor affinity for IL-2Rα. This fusion protein (OMCP-mutIL-2) potently and selectively activates IL-2 signalling only on NKG2D-bearing cells, such as natural killer (NK) cells, without broadly activating IL-2Rα-bearing cells. OMCP-mutIL-2 provides superior tumour control in several mouse models of malignancy and is not limited by mouse strain-specific variability of NK function. In addition, OMCP-mutIL-2 lacks the toxicity and vascular complications associated with parental wild-type IL-2.


Subject(s)
Interleukin-2/pharmacology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation/drug effects , Genes, MHC Class I , Humans , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit , Killer Cells, Natural/metabolism , Male , Mice , Mutation , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms, Experimental/drug therapy , Orthopoxvirus , Protein Binding , Recombinant Proteins
10.
Virology ; 448: 185-95, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24314649

ABSTRACT

Herpes simplex virus entry is initiated by glycoprotein D (gD) binding to a cellular receptor, such as HVEM or nectin-1. gD is activated by receptor-induced displacement of the C-terminus from the core of the glycoprotein. Binding of HVEM requires the formation of an N-terminal hairpin loop of gD; once formed this loop masks the nectin-1 binding site on the core of gD. We found that HVEM and nectin-1 exhibit non-reciprocal competition for binding to gD. The N-terminus of gD does not spontaneously form a stable hairpin in the absence of receptor and HVEM does not appear to rely on a pre-existing hairpin for binding to gD(3C-38C) mutants. However, HVEM function is affected by mutations that impair optimal hairpin formation. Furthermore, nectin-1 induces a new conformation of the N-terminus of gD. We conclude that the conformation of the N-terminus of gD is actively modified by the direct action of both receptors.


Subject(s)
Cell Adhesion Molecules/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Viral Envelope Proteins/chemistry , Amino Acid Motifs , Cell Adhesion Molecules/genetics , Cell Line , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/genetics , Humans , Nectins , Protein Binding , Protein Conformation , Receptors, Tumor Necrosis Factor, Member 14/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
11.
J Virol ; 87(2): 840-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115291

ABSTRACT

The NKG2D receptor is expressed on the surface of NK, T, and macrophage lineage cells and plays an important role in antiviral and antitumor immunity. To evade NKG2D recognition, herpesviruses block the expression of NKG2D ligands on the surface of infected cells using a diverse repertoire of sabotage methods. Cowpox and monkeypox viruses have taken an alternate approach by encoding a soluble NKG2D ligand, the orthopoxvirus major histocompatibility complex (MHC) class I-like protein (OMCP), which can block NKG2D-mediated cytotoxicity. This approach has the advantage of targeting a single conserved receptor instead of numerous host ligands that exhibit significant sequence diversity. Here, we show that OMCP binds the NKG2D homodimer as a monomer and competitively blocks host ligand engagement. We have also determined the 2.25-Å-resolution crystal structure of OMCP from the cowpox virus Brighton Red strain, revealing a truncated MHC class I-like platform domain consisting of a beta sheet flanked with two antiparallel alpha helices. OMCP is generally similar in structure to known host NKG2D ligands but has notable variations in regions typically used to engage NKG2D. Additionally, the determinants responsible for the 14-fold-higher affinity of OMCP for human than for murine NKG2D were mapped to a single loop in the NKG2D ligand-binding pocket.


Subject(s)
Cowpox virus/chemistry , NK Cell Lectin-Like Receptor Subfamily K/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Animals , Cowpox virus/pathogenicity , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Molecular Sequence Data , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Protein Binding , Protein Structure, Quaternary , Sequence Homology, Amino Acid , Viral Proteins/metabolism
12.
J Virol ; 86(3): 1563-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22130533

ABSTRACT

As the receptor-binding protein of herpes simplex virus (HSV), gD plays an essential role in virus entry. In its native state, the last 56 amino acids of the ectodomain C terminus (C-term) occlude binding to its receptors, herpesvirus entry mediator (HVEM) and nectin-1. Although it is clear that movement of the C-term must occur to permit receptor binding, we believe that this conformational change is also a key event for triggering later steps leading to fusion. Specifically, gD mutants containing disulfide bonds that constrain the C-term are deficient in their ability to trigger fusion following receptor binding. In this report, we show that two newly made monoclonal antibodies (MAbs), MC2 and MC5, have virus-neutralizing activity but do not block binding of gD to either receptor. In contrast, all previously characterized neutralizing anti-gD MAbs block binding of gD to a receptor(s). Interestingly, instead of blocking receptor binding, MC2 significantly enhances the affinity of gD for both receptors. Several nonneutralizing MAbs (MC4, MC10, and MC14) also enhanced gD-receptor binding. While MC2 and MC5 recognized different epitopes on the core of gD, these nonneutralizing MAbs recognized the gD C-term. Both the neutralizing capacity and rate of neutralization of virus by MC2 are uniquely enhanced when MC2 is combined with MAb MC4, MC10, or MC14. We suggest that MC2 and MC5 prevent gD from performing a function that triggers later steps leading to fusion and that the epitope for MC2 is normally occluded by the C-term of the gD ectodomain.


Subject(s)
Antibodies, Monoclonal/immunology , Neutralization Tests , Simplexvirus/immunology , Biosensing Techniques , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , Immunoprecipitation , Models, Molecular , Protein Conformation , Simplexvirus/chemistry
13.
J Exp Med ; 205(4): 981-92, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18391063

ABSTRACT

Nonliving antiviral vaccines traditionally target proteins expressed at the surface of the virion with the hope of inducing neutralizing antibodies. Orthopoxviruses (OPVs), such as the human smallpox virus and the mouse-equivalent ectromelia virus (ECTV; an agent of mousepox), encode immune response modifiers (IRMs) that can increase virulence by decreasing the host immune response. We show that one of these IRMs, the type I interferon (IFN) binding protein (bp) of ECTV, is essential for ECTV virulence and is a natural target of the antibody response. More strikingly, we demonstrate that immunization with recombinant type I IFN bp protects mice from lethal mousepox. Collectively, our experiments have important implications for our understanding of the role of IRMs in OPV virulence and of type I IFNs in OPV infections. Furthermore, our work provides proof of concept that effective antiviral vaccines can be made to prevent disease by targeting virulence factors as an alternative to the traditional approach that attempts to prevent infection by virus neutralization.


Subject(s)
Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Vaccination , Viral Proteins/immunology , Animals , Antibody Formation/immunology , Ectromelia virus/isolation & purification , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Genetic Complementation Test , Immunity, Innate/immunology , Immunocompetence , Interferon Type I/immunology , Mice , Open Reading Frames/genetics , Protein Binding , Receptors, Interferon/deficiency , Recombinant Proteins , Tissue Culture Techniques , Virulence
14.
J Virol ; 82(2): 700-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18032483

ABSTRACT

Glycoprotein D (gD) is the receptor binding protein of herpes simplex virus (HSV) and binds to at least two distinct protein receptors, herpesvirus entry mediator (HVEM) and nectin-1. While both receptor binding regions are found within the first 234 amino acids, a crystal structure shows that the C terminus of the gD ectodomain normally occludes the receptor binding sites. Receptor binding must therefore displace the C terminus, and this conformational change is postulated to be required for inducing fusion via gB and gH/gL. When cysteine residues are introduced at positions 37 and 302 of gD, a disulfide bond is formed that stabilizes the C terminus and prevents binding to either receptor. We speculated that if disulfide bonds were engineered further upstream, receptor binding might be separated from the induction of fusion. To test this, we made five additional double cysteine mutants, each potentially introducing a disulfide bond between the ectodomain C terminus and the core of the gD ectodomain. The two mutants predicted to impose the greatest constraint were unable to bind receptors or mediate cell-cell fusion. However, the three mutants with the most flexible C terminus bound well to both HVEM and nectin-1. Two of these mutants were impaired in cell-cell fusion and null-virus complementation. Importantly, a third mutant in this group was nonfunctional in both assays. This mutant clearly separates the role of gD in triggering fusion from its role in receptor binding. Based upon the properties of the panel of mutants we conclude that fusion requires greater flexibility of the gD ectodomain C terminus than does receptor binding.


Subject(s)
Herpesvirus 1, Human/physiology , Viral Envelope Proteins/physiology , Virus Attachment , Virus Internalization , Amino Acid Substitution , Disulfides/metabolism , Herpesvirus 1, Human/genetics , Mutagenesis, Site-Directed , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Viral Envelope Proteins/genetics
15.
EMBO J ; 24(23): 4144-53, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16292345

ABSTRACT

Herpes simplex virus (HSV) entry into cells requires binding of the envelope glycoprotein D (gD) to one of several cell surface receptors. The 50 C-terminal residues of the gD ectodomain are essential for virus entry, but not for receptor binding. We have determined the structure of an unliganded gD molecule that includes these C-terminal residues. The structure reveals that the C-terminus is anchored near the N-terminal region and masks receptor-binding sites. Locking the C-terminus in the position observed in the crystals by an intramolecular disulfide bond abolished receptor binding and virus entry, demonstrating that this region of gD moves upon receptor binding. Similarly, a point mutant that would destabilize the C-terminus structure was nonfunctional for entry, despite increased affinity for receptors. We propose that a controlled displacement of the gD C-terminus upon receptor binding is an essential feature of HSV entry, ensuring the timely activation of membrane fusion.


Subject(s)
Receptors, Virus/physiology , Simplexvirus/physiology , Simplexvirus/pathogenicity , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Dimerization , Herpes Simplex/metabolism , Ligands , Protein Structure, Tertiary , Simplexvirus/metabolism , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...