Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 8(1): 1735, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170370

ABSTRACT

RNA viruses are characterized by a high mutation rate, a buffer against environmental change. Nevertheless, the means by which random mutation improves viral fitness is not well characterized. Here we report the X-ray crystal structure of the receptor-binding domain (RBD) of the human coronavirus, HCoV-229E, in complex with the ectodomain of its receptor, aminopeptidase N (APN). Three extended loops are solely responsible for receptor binding and the evolution of HCoV-229E and its close relatives is accompanied by changing loop-receptor interactions. Phylogenetic analysis shows that the natural HCoV-229E receptor-binding loop variation observed defines six RBD classes whose viruses have successively replaced each other in the human population over the past 50 years. These RBD classes differ in their affinity for APN and their ability to bind an HCoV-229E neutralizing antibody. Together, our results provide a model for alphacoronavirus adaptation and evolution based on the use of extended loops for receptor binding.


Subject(s)
Coronavirus 229E, Human/genetics , Coronavirus 229E, Human/physiology , Adaptation, Physiological/genetics , Amino Acid Sequence , CD13 Antigens/chemistry , CD13 Antigens/metabolism , Coronavirus 229E, Human/pathogenicity , Coronavirus Infections/virology , Crystallography, X-Ray , Evolution, Molecular , Genetic Variation , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Models, Biological , Models, Molecular , Phylogeny , Protein Interaction Domains and Motifs , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Surface Plasmon Resonance
2.
Nat Commun ; 8(1): 185, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28775322

ABSTRACT

Protein O-glucosyltransferase 1/Rumi-mediated glucosylation of Notch epidermal growth factor-like (EGF-like) domains plays an important role in Notch signaling. Protein O-glucosyltransferase 1 shows specificity for folded EGF-like domains, it can only glycosylate serine residues in the C1XSXPC2 motif, and it possesses an uncommon dual donor substrate specificity. Using several EGF-like domains and donor substrate analogs, we have determined the structures of human Protein O-glucosyltransferase 1 substrate/product complexes that provide mechanistic insight into the basis for these properties. Notably, we show that Protein O-glucosyltransferase 1's requirement for folded EGF-like domains also leads to its serine specificity and that two distinct local conformational states are likely responsible for its ability to transfer both glucose and xylose. We also show that Protein O-glucosyltransferase 1 possesses the potential to xylosylate a much broader range of EGF-like domain substrates than was previously thought. Finally, we show that Protein O-glucosyltransferase 1 has co-evolved with EGF-like domains of the type found in Notch.POGLUT1 is a protein-O-glucosyltransferase that transfers glucose and xylose to the EGF-like domains of Notch and other signaling receptors. Here the authors report the structure of human POGLUT1 in complexes with 3 different EGF-like domains and donor substrates and shed light on the enzyme's substrate specificity and catalytic mechanism.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glucosyltransferases/metabolism , Receptors, Notch/metabolism , Catalytic Domain , Glucosyltransferases/genetics , HEK293 Cells , Humans , Models, Molecular , Protein Conformation , Receptors, Notch/genetics
3.
Nat Chem Biol ; 13(7): 757-763, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530709

ABSTRACT

Protein O-fucosyltransferase 1 (POFUT1) fucosylates the epidermal growth factor (EGF)-like domains found in cell-surface and secreted glycoproteins including Notch and its ligands. Although Notch fucosylation is critical for development, and POFUT1 deficiency leads to human disease, how this enzyme binds and catalyzes the fucosylation of its diverse EGF-like domain substrates has not been determined. Reported here is the X-ray crystal structure of mouse POFUT1 in complex with several EGF-like domains, including EGF12 and EGF26 of Notch. Overall shape complementarity, interactions with invariant atoms of the fucosylation motif and flexible segments on POFUT1 all define its EGF-like-domain binding properties. Using large-scale structural and sequence analysis, we also show that POFUT1 binds EGF-like domains of the hEGF type and that the highly correlated presence of POFUT1 and fucosylatable hEGFs has accompanied animal evolution.


Subject(s)
Epidermal Growth Factor/chemistry , Fucosyltransferases/metabolism , Protein Domains , Receptors, Notch/metabolism , Animals , Crystallography, X-Ray , Humans , Mice , Models, Molecular
4.
Vaccine ; 30(20): 3061-7, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22406109

ABSTRACT

Despite the availability of live attenuated measles virus (MV) vaccines, a large number of measles-associated deaths occur among infants in developing countries. The development of a measles subunit vaccine may circumvent the limitations associated with the current live attenuated vaccines and eventually contribute to global measles eradication. Therefore, the goal of this study was to test the feasibility of producing the recombinant globular head domain of the MV hemagglutinin (H) protein by stably transfected human cells and to examine the ability of this recombinant protein to elicit MV-specific immune responses. The recombinant protein was purified from the culture supernatant of stably transfected HEK293T cells secreting a tagged version of the protein. Two subcutaneous immunizations with the purified recombinant protein alone resulted in the production of MV-specific serum IgG and neutralizing antibodies in mice. Formulation of the protein with adjuvants (polyphosphazene or alum) further enhanced the humoral immune response and in addition resulted in the induction of cell-mediated immunity as measured by the production of MV H-specific interferon gamma (IFN-γ) and interleukin 5 (IL-5) by in vitro re-stimulated splenocytes. Furthermore, the inclusion of polyphosphazene into the vaccine formulation induced a mixed Th1/Th2-type immune response. In addition, the purified recombinant protein retained its immunogenicity even after storage at 37°C for 2 weeks.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cell Line , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/isolation & purification , Humans , Interferon-gamma/metabolism , Interleukin-5/metabolism , Leukocytes, Mononuclear/immunology , Measles Vaccine/administration & dosage , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
5.
J Mol Biol ; 414(5): 798-811, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22056345

ABSTRACT

Leukocyte-type core 2 ß1,6-N-acetylglucosaminyltransferase (C2GnT-L) is an inverting, metal-ion-independent glycosyltransferase that catalyzes the formation of mucin-type core 2 O-glycans. C2GnT-L belongs to the GT-A fold, yet it lacks the metal ion binding DXD motif characteristic of other nucleoside disphosphate GT-A fold glycosyltransferases. To shed light on the basis for its metal ion independence, we have solved the X-ray crystal structure (2.3 Å resolution) of a mutant form of C2GnT-L (C217S) in complex with the nucleotide sugar product UDP and, using site-directed mutagenesis, examined the roles of R378 and K401 in both substrate binding and catalysis. The structure shows that C2GnT-L exists in an "open" conformation and a "closed" conformation and that, in the latter, R378 and K401 interact with the ß-phosphate moiety of the bound UDP. The two conformations are likely to be important in catalysis, but the conformational changes that lead to their interconversion do not resemble the nucleotide-sugar-mediated loop ordering observed in other GT-A glycosyltransferases. R378 and K401 were found to be important in substrate binding and/or catalysis, an observation consistent with the suggestion that they serve the same role played by metal ion in all of the other GT-A glycosyltransferases studied to date. Notably, R378 and K401 appear to function in a manner similar to that of the arginine and lysine residues contained in the RX(4-5)K motif found in the retaining GT-B glycosyltransferases.


Subject(s)
Leukocytes/enzymology , N-Acetylglucosaminyltransferases/chemistry , Amino Acid Motifs , HEK293 Cells , Humans , Models, Molecular , Mutation , N-Acetylglucosaminyltransferases/genetics , Protein Conformation
6.
J Mol Biol ; 388(4): 815-23, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19324051

ABSTRACT

The spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) is responsible for host cell attachment and fusion of the viral and host cell membranes. Within S the receptor binding domain (RBD) mediates the interaction with angiotensin-converting enzyme 2 (ACE2), the SARS-CoV host cell receptor. Both S and the RBD are highly immunogenic and both have been found to elicit neutralizing antibodies. Reported here is the X-ray crystal structure of the RBD in complex with the Fab of a neutralizing mouse monoclonal antibody, F26G19, elicited by immunization with chemically inactivated SARS-CoV. The RBD-F26G19 Fab complex represents the first example of the structural characterization of an antibody elicited by an immune response to SARS-CoV or any fragment of it. The structure reveals that the RBD surface recognized by F26G19 overlaps significantly with the surface recognized by ACE2 and, as such, suggests that F26G19 likely neutralizes SARS-CoV by blocking the virus-host cell interaction.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Peptidyl-Dipeptidase A , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibody Formation/immunology , Binding Sites , Cell Line , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Membrane Glycoproteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics
7.
J Exp Med ; 205(12): 2763-79, 2008 Nov 24.
Article in English | MEDLINE | ID: mdl-19001139

ABSTRACT

Progressive loss of T cell functionality is a hallmark of chronic infection with human immunodeficiency virus 1 (HIV-1). We have identified a novel population of dysfunctional T cells marked by surface expression of the glycoprotein Tim-3. The frequency of this population was increased in HIV-1-infected individuals to a mean of 49.4 +/- SD 12.9% of CD8(+) T cells expressing Tim-3 in HIV-1-infected chronic progressors versus 28.5 +/- 6.8% in HIV-1-uninfected individuals. Levels of Tim-3 expression on T cells from HIV-1-infected inviduals correlated positively with HIV-1 viral load and CD38 expression and inversely with CD4(+) T cell count. In progressive HIV-1 infection, Tim-3 expression was up-regulated on HIV-1-specific CD8(+) T cells. Tim-3-expressing T cells failed to produce cytokine or proliferate in response to antigen and exhibited impaired Stat5, Erk1/2, and p38 signaling. Blocking the Tim-3 signaling pathway restored proliferation and enhanced cytokine production in HIV-1-specific T cells. Thus, Tim-3 represents a novel target for the therapeutic reversal of HIV-1-associated T cell dysfunction.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Membrane Proteins/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antiretroviral Therapy, Highly Active , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Disease Progression , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , HIV Infections/drug therapy , HIV Infections/pathology , HIV Infections/physiopathology , HIV-1/pathogenicity , HLA Antigens , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins/genetics , Phenotype , Programmed Cell Death 1 Receptor , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes/cytology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
8.
Vaccine ; 25(1): 136-43, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-16919855

ABSTRACT

We studied the immunogenicity of an anti-SARS subunit vaccine comprised of the fragment of the SARS coronavirus (SARS-CoV) spike protein amino acids 318-510 (S318-510) containing the receptor-binding domain. The S protein fragment was purified from the culture supernatant of stably transformed HEK293T cells secreting a tagged version of the protein. The vaccine was given subcutaneously to 129S6/SvEv mice in saline, with alum adjuvant or with alum plus CpG oligodeoxynucleotides (ODN). Mice immunized with the adjuvanted antigen elicited strong antibody and cellular immune responses; furthermore, adding the CpG ODN to the alum resulted in increased IgG2a antibody titers and a higher number of INF-gamma-secreting murine splenocytes. Mice vaccinated with S318-510 deglycosylated by PNGase F (dgS318-510) showed a lower neutralizing antibody response but had similar numbers of INF-gamma-producing cells in the spleen. This finding suggests that carbohydrate is important for the immunogenicity of the S318-510 protein fragment and provide useful information for designing an effective and safe SARS subunit vaccine.


Subject(s)
Membrane Glycoproteins , Receptors, Virus/metabolism , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins , Viral Vaccines/immunology , Alum Compounds , Animals , Antibodies, Viral/blood , Cell Line , Female , Humans , Interferon-gamma/biosynthesis , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Oligodeoxyribonucleotides , Severe Acute Respiratory Syndrome/immunology , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Vaccines/administration & dosage
9.
J Biol Chem ; 281(36): 26693-701, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16829524

ABSTRACT

Leukocyte type core 2 beta1,6-N-acetylglucosaminyltransferase (C2GnT-L) is a key enzyme in the biosynthesis of branched O-glycans. It is an inverting, metal ion-independent family 14 glycosyltransferase that catalyzes the formation of the core 2 O-glycan (Galbeta1-3[GlcNAcbeta1-6]GalNAc-O-Ser/Thr) from its donor and acceptor substrates, UDP-GlcNAc and the core 1 O-glycan (Galbeta1-3GalNAc-O-Ser/Thr), respectively. Reported here are the x-ray crystal structures of murine C2GnT-L in the absence and presence of the acceptor substrate Galbeta1-3GalNAc at 2.0 and 2.7A resolution, respectively. C2GnT-L was found to possess the GT-A fold; however, it lacks the characteristic metal ion binding DXD motif. The Galbeta1-3GalNAc complex defines the determinants of acceptor substrate binding and shows that Glu-320 corresponds to the structurally conserved catalytic base found in other inverting GT-A fold glycosyltransferases. Comparison of the C2GnT-L structure with that of other GT-A fold glycosyltransferases further suggests that Arg-378 and Lys-401 serve to electrostatically stabilize the nucleoside diphosphate leaving group, a role normally played by metal ion in GT-A structures. The use of basic amino acid side chains in this way is strikingly similar to that seen in a number of metal ion-independent GT-B fold glycosyltransferases and suggests a convergence of catalytic mechanism shared by both GT-A and GT-B fold glycosyltransferases.


Subject(s)
Ions/chemistry , Metals/chemistry , N-Acetylglucosaminyltransferases/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Mice , Models, Molecular , Molecular Sequence Data , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Polysaccharides/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Substrate Specificity , Uridine Diphosphate Glucuronic Acid/metabolism
10.
J Mol Biol ; 360(1): 67-79, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16769084

ABSTRACT

The Golgi-resident glycosyltransferase, UDP-N-acetyl-d-glucosamine:alpha-3-d-mannoside beta-1,2-N-acetylglucosaminyltransferase I (GnT I), initiates the conversion of high-mannose oligosaccharides to complex and hybrid structures in the biosynthesis of N-linked glycans. Reported here are the X-ray crystal structures of GnT I in complex with UDP-CH2-GlcNAc (a non-hydrolyzable C-glycosidic phosphonate), UDP-2-deoxy-2-fluoro-glucose, UDP-glucose and UDP. Collectively, these structures provide evidence for the importance of the GlcNAc moiety and its N-acetyl group in donor substrate binding, as well as insight into the role played by the flexible 318-330 loop in substrate binding and product release. In addition, the UDP-CH2-GlcNAc complex reveals a well-defined glycerol molecule poised for nucleophilic attack on the C1 atom of the donor substrate analogue. The position and orientation of this glycerol molecule have allowed us to model the binding of the Manalpha1,3Manbeta1 moiety of the acceptor substrate and, based on the model, to suggest a rationalization for the main determinants of GnT I acceptor specificity.


Subject(s)
N-Acetylglucosaminyltransferases/chemistry , Animals , Catalysis , Crystallography, X-Ray , Glycosyltransferases/chemistry , Kinetics , Mannose/chemistry , Models, Chemical , Models, Molecular , Oligosaccharides/chemistry , Protein Binding , Protein Structure, Tertiary , Rabbits , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...