Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32699088

ABSTRACT

N-linked glycans commonly contribute to secretory protein folding, sorting, and signaling. For enveloped viruses, such as the influenza A virus (IAV), large N-linked glycans can also be added to prevent access to epitopes on the surface antigens hemagglutinin (HA or H) and neuraminidase (NA or N). Sequence analysis showed that in the NA head domain of H1N1 IAVs, three N-linked glycosylation sites are conserved and that a fourth site is conserved in H3N2 IAVs. Variable sites are almost exclusive to H1N1 IAVs of human origin, where the number of head glycosylation sites first increased over time and then decreased with and after the introduction of the 2009 pandemic H1N1 IAV of Eurasian swine origin. In contrast, variable sites exist in H3N2 IAVs of human and swine origin, where the number of head glycosylation sites has mainly increased over time. Analysis of IAVs carrying N1 and N2 mutants demonstrated that the N-linked glycosylation sites on the NA head domain are required for efficient virion incorporation and replication in cells and eggs. It also revealed that N1 stability is more affected by the head domain glycans, suggesting N2 is more amenable to glycan additions. Together, these results indicate that in addition to antigenicity, N-linked glycosylation sites can alter NA enzymatic stability and the NA amount in virions.IMPORTANCE N-linked glycans are transferred to secretory proteins upon entry into the endoplasmic reticulum lumen. In addition to promoting secretory protein maturation, enveloped viruses also utilize these large oligosaccharide structures to prevent access to surface antigen epitopes. Sequence analyses of the influenza A virus (IAV) surface antigen neuraminidase (NA or N) showed that the conservation of N-linked glycosylation sites on the NA enzymatic head domain differs by IAV subtype (H1N1 versus H3N2) and species of origin, with human-derived IAVs possessing the most variability. Experimental analyses verified that the N-linked glycosylation sites on the NA head domain contribute to virion incorporation and replication. It also revealed that the head domain glycans affect N1 stability more than N2, suggesting N2 is more accommodating to glycan additions. These results demonstrate that in addition to antigenicity, changes in N-linked glycosylation sites can alter other properties of viral surface antigens and virions.


Subject(s)
Influenza A virus/metabolism , Neuraminidase/chemistry , Neuraminidase/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Virus Replication/physiology , Animals , Antigens, Viral/metabolism , Cell Line , Dogs , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Models, Molecular , Mutation , Neuraminidase/genetics , Protein Folding , Swine , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/metabolism
2.
Nat Microbiol ; 5(8): 1064, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32546783

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nat Microbiol ; 4(12): 2565-2577, 2019 12.
Article in English | MEDLINE | ID: mdl-31451775

ABSTRACT

Influenza neuraminidase (NA) is a sialidase that contributes to viral mobility by removing the extracellular receptors for the haemagglutinin (HA) glycoprotein. However, it remains unclear why influenza NAs evolved to function as Ca2+-dependent tetramers that display variable stability. Here, we show that the Ca2+ ion located at the centre of the NA tetramer is a major stability determinant, as this Ca2+ ion is required for catalysis and its binding affinity varies between NAs. By examining NAs from 2009 pandemic-like H1N1 viruses, we traced the affinity variation to local substitutions that cause residues in the central Ca2+-binding pocket to reposition. A temporal analysis revealed that these local substitutions predictably alter the stability of the 2009 pandemic-like NAs and contribute to the tendency for the stability to vary up and down over time. In addition to the changes in stability, the structural plasticity of NA was also shown to support the formation of heterotetramers, which creates a mechanism for NA to obtain hybrid properties and propagate suboptimal mutants. Together, these results demonstrate how the structural restrictions for activity provide influenza NA with several mechanisms for adaptation and diversification.


Subject(s)
Influenza A virus/enzymology , Neuraminidase/chemistry , Neuraminidase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Binding Sites , Calcium , Carrier Proteins , Cell Line , Enzyme Stability , Genetic Heterogeneity , Genome, Viral , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A virus/genetics , Influenza A virus/growth & development , Influenza, Human/virology , Kinetics , Models, Molecular , Neuraminidase/genetics , Protein Conformation , Sequence Analysis, Protein , Viral Proteins/genetics , Virus Replication
4.
ACS Nano ; 13(6): 6689-6701, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31199124

ABSTRACT

Eukaryotic cells possess a dynamic network of membranes that vary in lipid composition. To perform numerous biological functions, cells modulate their shape and the lateral organization of proteins associated with membranes. The modulation is generally facilitated by physical cues that recruit proteins to specific regions of the membrane. Analyzing these cues is difficult due to the complexity of the membrane conformations that exist in cells. Here, we examine how different types of membrane proteins respond to changes in curvature and to lipid phases found in the plasma membrane. By using giant plasma membrane vesicles derived from transfected cells, the proteins were positioned in the correct orientation and the analysis was performed in plasma membranes with a biological composition. Nanoscale membrane curvatures were generated by extracting nanotubes from these vesicles with an optical trap. The viral membrane protein neuraminidase was not sensitive to curvature, but it did exhibit strong partitioning (coefficient of K = 0.16) disordered membrane regions. In contrast, the membrane repair protein annexin 5 showed a preference for nanotubes with a density up to 10-15 times higher than that on the more flat vesicle membrane. The investigation of nanoscale effects in isolated plasma membranes provides a quantitative platform for studying peripheral and integral membrane proteins in their natural environment.


Subject(s)
Extracellular Vesicles/chemistry , Membrane Proteins/metabolism , Annexin A5/chemistry , Annexin A5/metabolism , Extracellular Vesicles/metabolism , HEK293 Cells , Humans , Liposomes/chemistry , Liposomes/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Membrane Proteins/chemistry , Nanotubes/chemistry , Neuraminidase/chemistry , Neuraminidase/metabolism , Optical Tweezers , Protein Transport
5.
Front Immunol ; 9: 1581, 2018.
Article in English | MEDLINE | ID: mdl-30079062

ABSTRACT

Influenza viruses replicate within the nucleus of the host cell. This uncommon RNA virus trait provides influenza with the advantage of access to the nuclear machinery during replication. However, it also increases the complexity of the intracellular trafficking that is required for the viral components to establish a productive infection. The segmentation of the influenza genome makes these additional trafficking requirements especially challenging, as each viral RNA (vRNA) gene segment must navigate the network of cellular membrane barriers during the processes of entry and assembly. To accomplish this goal, influenza A viruses (IAVs) utilize a combination of viral and cellular mechanisms to coordinate the transport of their proteins and the eight vRNA gene segments in and out of the cell. The aim of this review is to present the current mechanistic understanding for how IAVs facilitate cell entry, replication, virion assembly, and intercellular movement, in an effort to highlight some of the unanswered questions regarding the coordination of the IAV infection process.

6.
J Cell Biol ; 216(8): 2283-2293, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28696227

ABSTRACT

A primary function of 5' regions in many secretory protein mRNAs is to encode an endoplasmic reticulum (ER) targeting sequence. In this study, we show how the regions coding for the ER-targeting sequences of the influenza glycoproteins NA and HA also function as translational regulatory elements that are controlled by the viral RNA-binding protein (RBP) NS1. The translational increase depends on the nucleotide composition and 5' positioning of the ER-targeting sequence coding regions and is facilitated by the RNA-binding domain of NS1, which can associate with ER membranes. Inserting the ER-targeting sequence coding region of NA into different 5' UTRs confirmed that NS1 can promote the translation of secretory protein mRNAs based on the nucleotides within this region rather than the resulting amino acids. By analyzing human protein mRNA sequences, we found evidence that this mechanism of using 5' coding regions and particular RBPs to achieve gene-specific regulation may extend to human-secreted proteins.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Influenza A Virus, H1N1 Subtype/enzymology , Neuraminidase/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , A549 Cells , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/enzymology , HEK293 Cells , HeLa Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Neuraminidase/genetics , Protein Binding , Protein Biosynthesis , Protein Domains , RNA, Messenger/genetics , RNA, Viral/genetics , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Transfection , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics
7.
Cell Rep ; 20(1): 251-263, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683318

ABSTRACT

Genome delivery to the proper cellular compartment for transcription and replication is a primary goal of viruses. However, methods for analyzing viral genome localization and differentiating genomes with high identity are lacking, making it difficult to investigate entry-related processes and co-examine heterogeneous RNA viral populations. Here, we present an RNA labeling approach for single-cell analysis of RNA viral replication and co-infection dynamics in situ, which uses the versatility of padlock probes. We applied this method to identify influenza A virus (IAV) infections in cells and lung tissue with single-nucleotide specificity and to classify entry and replication stages by gene segment localization. Extending the classification strategy to co-infections of IAVs with single-nucleotide variations, we found that the dependence on intracellular trafficking places a time restriction on secondary co-infections necessary for genome reassortment. Altogether, these data demonstrate how RNA viral genome labeling can help dissect entry and co-infections.


Subject(s)
Genome, Viral , Influenza A virus/physiology , RNA, Viral/metabolism , Single-Cell Analysis/methods , Staining and Labeling/methods , Virus Replication , Animals , Dogs , Epithelial Cells/virology , HEK293 Cells , Humans , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Proteins/metabolism
8.
Biochim Biophys Acta ; 1858(12): 2984-2992, 2016 12.
Article in English | MEDLINE | ID: mdl-27620332

ABSTRACT

We used the amphipathic styrene maleic acid (SMA) co-polymer to extract cytochrome c oxidase (CytcO) in its native lipid environment from S. cerevisiae mitochondria. Native nanodiscs containing one CytcO per disc were purified using affinity chromatography. The longest cross-sections of the native nanodiscs were 11nm×14nm. Based on this size we estimated that each CytcO was surrounded by ~100 phospholipids. The native nanodiscs contained the same major phospholipids as those found in the mitochondrial inner membrane. Even though CytcO forms a supercomplex with cytochrome bc1 in the mitochondrial membrane, cyt. bc1 was not found in the native nanodiscs. Yet, the loosely-bound Respiratory SuperComplex factors were found to associate with the isolated CytcO. The native nanodiscs displayed an O2-reduction activity of ~130 electrons CytcO-1s-1 and the kinetics of the reaction of the fully reduced CytcO with O2 was essentially the same as that observed with CytcO in mitochondrial membranes. The kinetics of CO-ligand binding to the CytcO catalytic site was similar in the native nanodiscs and the mitochondrial membranes. We also found that excess SMA reversibly inhibited the catalytic activity of the mitochondrial CytcO, presumably by interfering with cyt. c binding. These data point to the importance of removing excess SMA after extraction of the membrane protein. Taken together, our data shows the high potential of using SMA-extracted CytcO for functional and structural studies.


Subject(s)
Electron Transport Complex IV/isolation & purification , Lipids/analysis , Saccharomyces cerevisiae/enzymology , Catalytic Domain , Electron Transport Complex IV/chemistry , Maleates/pharmacology , Nanoparticles , Polystyrenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...