Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Traffic ; 25(1): e12927, 2024 01.
Article in English | MEDLINE | ID: mdl-38272446

ABSTRACT

Endoplasmic reticulum (ER) retention of misfolded glycoproteins is mediated by the ER-localized eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognizes a misfolded glycoprotein and flags it for ER retention by re-glucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease, even if the mutant glycoprotein retains activity ("responsive mutant"). Using confocal laser scanning microscopy, we investigated here the subcellular localization of the human Trop-2-Q118E, E227K and L186P mutants, which cause gelatinous drop-like corneal dystrophy (GDLD). Compared with the wild-type Trop-2, which is correctly localized at the plasma membrane, these Trop-2 mutants are retained in the ER. We studied fluorescent chimeras of the Trop-2 Q118E, E227K and L186P mutants in mammalian cells harboring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 genes. The membrane localization of the Trop-2 Q118E, E227K and L186P mutants was successfully rescued in UGGT1-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation would constitute a novel therapeutic strategy for the treatment of pathological conditions associated to misfolded membrane glycoproteins (whenever the mutation impairs but does not abrogate function), and it encourages the testing of modulators of ER glycoprotein folding quality control as broad-spectrum rescue-of-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.


Subject(s)
Protein Folding , Rare Diseases , Animals , Humans , Rare Diseases/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Endoplasmic Reticulum/metabolism , Mutation , Mammals/metabolism , Glucosyltransferases/metabolism
2.
Proc Natl Acad Sci U S A ; 119(15): e2119893119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385354

ABSTRACT

The emergence of SARS-CoV-2 triggering the COVID-19 pandemic ranks as arguably the greatest medical emergency of the last century. COVID-19 has highlighted health disparities both within and between countries and will leave a lasting impact on global society. Nonetheless, substantial investment in life sciences over recent decades has facilitated a rapid scientific response with innovations in viral characterization, testing, and sequencing. Perhaps most remarkably, this permitted the development of highly effective vaccines, which are being distributed globally at unprecedented speed. In contrast, drug treatments for the established disease have delivered limited benefits so far. Innovative and rapid approaches in the design and execution of large-scale clinical trials and repurposing of existing drugs have saved many lives; however, many more remain at risk. In this review we describe challenges and unmet needs, discuss existing therapeutics, and address future opportunities. Consideration is given to factors that have hindered drug development in order to support planning for the next pandemic challenge and to allow rapid and cost-effective development of new therapeutics with equitable delivery.


Subject(s)
COVID-19 Drug Treatment , Pandemics , COVID-19 Vaccines , Drug Development , Humans , Pandemics/prevention & control , SARS-CoV-2
3.
J Proteome Res ; 22(6): 1614-1629, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37219084

ABSTRACT

Japanese encephalitis virus is a leading cause of neurological infection in the Asia-Pacific region with no means of detection in more remote areas. We aimed to test the hypothesis of a Japanese encephalitis (JE) protein signature in human cerebrospinal fluid (CSF) that could be harnessed in a rapid diagnostic test (RDT), contribute to understanding the host response and predict outcome during infection. Liquid chromatography and tandem mass spectrometry (LC-MS/MS), using extensive offline fractionation and tandem mass tag labeling (TMT), enabled comparison of the deep CSF proteome in JE vs other confirmed neurological infections (non-JE). Verification was performed using data-independent acquisition (DIA) LC-MS/MS. 5,070 proteins were identified, including 4,805 human proteins and 265 pathogen proteins. Feature selection and predictive modeling using TMT analysis of 147 patient samples enabled the development of a nine-protein JE diagnostic signature. This was tested using DIA analysis of an independent group of 16 patient samples, demonstrating 82% accuracy. Ultimately, validation in a larger group of patients and different locations could help refine the list to 2-3 proteins for an RDT. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD034789 and 10.6019/PXD034789.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Humans , Encephalitis, Japanese/diagnosis , Chromatography, Liquid/methods , Proteomics/methods , Tandem Mass Spectrometry/methods , Proteome/analysis
4.
Proc Natl Acad Sci U S A ; 117(47): 29595-29601, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33154157

ABSTRACT

Mammalian protein N-linked glycosylation is critical for glycoprotein folding, quality control, trafficking, recognition, and function. N-linked glycans are synthesized from Glc3Man9GlcNAc2 precursors that are trimmed and modified in the endoplasmic reticulum (ER) and Golgi apparatus by glycoside hydrolases and glycosyltransferases. Endo-α-1,2-mannosidase (MANEA) is the sole endo-acting glycoside hydrolase involved in N-glycan trimming and is located within the Golgi, where it allows ER-escaped glycoproteins to bypass the classical N-glycosylation trimming pathway involving ER glucosidases I and II. There is considerable interest in the use of small molecules that disrupt N-linked glycosylation as therapeutic agents for diseases such as cancer and viral infection. Here we report the structure of the catalytic domain of human MANEA and complexes with substrate-derived inhibitors, which provide insight into dynamic loop movements that occur on substrate binding. We reveal structural features of the human enzyme that explain its substrate preference and the mechanistic basis for catalysis. These structures have inspired the development of new inhibitors that disrupt host protein N-glycan processing of viral glycans and reduce the infectivity of bovine viral diarrhea and dengue viruses in cellular models. These results may contribute to efforts aimed at developing broad-spectrum antiviral agents and help provide a more in-depth understanding of the biology of mammalian glycosylation.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Glycosylation/drug effects , Mannosidases/chemistry , Mannosidases/pharmacology , Animals , Bovine Virus Diarrhea-Mucosal Disease/drug therapy , Cattle , Cell Line , Dengue Virus/drug effects , Dogs , Glucosidases/metabolism , Humans , Madin Darby Canine Kidney Cells , Polysaccharides/metabolism , Secretory Pathway/drug effects
5.
Int J Mol Sci ; 25(1)2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38203525

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has claimed over 7 million lives worldwide, providing a stark reminder of the importance of pandemic preparedness. Due to the lack of approved antiviral drugs effective against coronaviruses at the start of the pandemic, the world largely relied on repurposed efforts. Here, we summarise results from randomised controlled trials to date, as well as selected in vitro data of directly acting antivirals, host-targeting antivirals, and immunomodulatory drugs. Overall, repurposing efforts evaluating directly acting antivirals targeting other viral families were largely unsuccessful, whereas several immunomodulatory drugs led to clinical improvement in hospitalised patients with severe disease. In addition, accelerated drug discovery efforts during the pandemic progressed to multiple novel directly acting antivirals with clinical efficacy, including small molecule inhibitors and monoclonal antibodies. We argue that large-scale investment is required to prepare for future pandemics; both to develop an arsenal of broad-spectrum antivirals beyond coronaviruses and build worldwide clinical trial networks that can be rapidly utilised.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics , Immunomodulating Agents , Antiviral Agents/therapeutic use
6.
Immunology ; 164(3): 587-601, 2021 11.
Article in English | MEDLINE | ID: mdl-34287854

ABSTRACT

Sepsis is a life-threatening condition involving a dysregulated immune response to infectious agents that cause injury to host tissues and organs. Current treatments are limited to early administration of antibiotics and supportive care. While appealing, the strategy of targeted inhibition of individual molecules in the inflammatory cascade has not proved beneficial. Non-targeted, systemic immunosuppression with steroids has shown limited efficacy and raises concern for secondary infection. Iminosugars are a class of small molecule glycomimetics with distinct inhibition profiles for glycan processing enzymes based on stereochemistry. Inhibition of host endoplasmic reticulum resident glycoprotein processing enzymes has demonstrated efficacy as a broad-spectrum antiviral strategy, but limited consideration has been given to the effects on host glycoprotein production and consequent disruption of signalling cascades. This work demonstrates that iminosugars inhibit dengue virus, bacterial lipopolysaccharide and fungal antigen-stimulated cytokine responses in human macrophages. In spite of decreased inflammatory mediator production, viral replication is suppressed in the presence of iminosugar. Transcriptome analysis reveals the key interaction of pathogen-induced endoplasmic reticulum stress, the resulting unfolded protein response and inflammation. Our work shows that iminosugars modulate these interactions. Based on these findings, we propose a new therapeutic role for iminosugars as treatment for sepsis-related inflammatory disorders associated with excess cytokine secretion.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Sepsis/drug therapy , Unfolded Protein Response/drug effects , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antigens, Fungal/immunology , Cells, Cultured , Dengue Virus/immunology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Lipopolysaccharides/immunology , Macrophages , Primary Cell Culture , Sepsis/immunology , Sepsis/microbiology , Toll-Like Receptor 4/metabolism , Unfolded Protein Response/immunology
7.
Plant Biotechnol J ; 19(10): 2027-2039, 2021 10.
Article in English | MEDLINE | ID: mdl-34002936

ABSTRACT

Chronic infection with hepatitis C virus (HCV) remains a leading cause of liver-related pathologies and a global health problem, currently affecting more than 71 million people worldwide. The development of a prophylactic vaccine is much needed to complement the effective antiviral treatment available and achieve HCV eradication. Current strategies focus on increasing the immunogenicity of the HCV envelope glycoprotein E2, the major target of virus-neutralizing antibodies, by testing various expression systems or manipulating the protein conformation and the N-glycosylation pattern. Here we report the first evidence of successful production of the full-length HCV E2 glycoprotein in Nicotiana benthamiana, by using the Agrobacterium-mediated transient expression technology. Molecular and functional analysis showed that the viral protein was correctly processed in plant cells and achieved the native folding required for binding to CD81, one of the HCV receptors. N-glycan analysis of HCV-E2 produced in N. benthamiana and mammalian cells indicated host-specific trimming of mannose residues and possibly, protein trafficking. Notably, the plant-derived viral antigen triggered a significant immune response in vaccinated mice, characterized by the presence of antibodies with HCV-neutralizing activity. Together, our study demonstrates that N. benthamiana is a viable alternative to costly mammalian cell cultures for the expression of complex viral antigens and supports the use of plants as cost-effective production platforms for the development of HCV vaccines.


Subject(s)
Hepacivirus , Viral Hepatitis Vaccines , Animals , Antibodies, Neutralizing , Hepatitis C Antibodies , Mice , Nicotiana , Viral Envelope Proteins/genetics
8.
Chem Rev ; 118(7): 3559-3607, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29488756

ABSTRACT

Membrane proteins perform a host of vital cellular functions. Deciphering the molecular mechanisms whereby they fulfill these functions requires detailed biophysical and structural investigations. Detergents have proven pivotal to extract the protein from its native surroundings. Yet, they provide a milieu that departs significantly from that of the biological membrane, to the extent that the structure, the dynamics, and the interactions of membrane proteins in detergents may considerably vary, as compared to the native environment. Understanding the impact of detergents on membrane proteins is, therefore, crucial to assess the biological relevance of results obtained in detergents. Here, we review the strengths and weaknesses of alkyl phosphocholines (or foscholines), the most widely used detergent in solution-NMR studies of membrane proteins. While this class of detergents is often successful for membrane protein solubilization, a growing list of examples points to destabilizing and denaturing properties, in particular for α-helical membrane proteins. Our comprehensive analysis stresses the importance of stringent controls when working with this class of detergents and when analyzing the structure and dynamics of membrane proteins in alkyl phosphocholine detergents.


Subject(s)
Cell Membrane/ultrastructure , Detergents/chemistry , Membrane Proteins/chemistry , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Animals , Biophysical Phenomena , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Magnetic Resonance Spectroscopy/methods , Micelles , Models, Molecular , Protein Conformation , Protein Folding , Protein Stability , Solubility
9.
Proc Natl Acad Sci U S A ; 114(32): 8544-8549, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739903

ABSTRACT

Glycoproteins traversing the eukaryotic secretory pathway begin life in the endoplasmic reticulum (ER), where their folding is surveyed by the 170-kDa UDP-glucose:glycoprotein glucosyltransferase (UGGT). The enzyme acts as the single glycoprotein folding quality control checkpoint: it selectively reglucosylates misfolded glycoproteins, promotes their association with ER lectins and associated chaperones, and prevents premature secretion from the ER. UGGT has long resisted structural determination and sequence-based domain boundary prediction. Questions remain on how this single enzyme can flag misfolded glycoproteins of different sizes and shapes for ER retention and how it can span variable distances between the site of misfold and a glucose-accepting N-linked glycan on the same glycoprotein. Here, crystal structures of a full-length eukaryotic UGGT reveal four thioredoxin-like (TRXL) domains arranged in a long arc that terminates in two ß-sandwiches tightly clasping the glucosyltransferase domain. The fold of the molecule is topologically complex, with the first ß-sandwich and the fourth TRXL domain being encoded by nonconsecutive stretches of sequence. In addition to the crystal structures, a 15-Å cryo-EM reconstruction reveals interdomain flexibility of the TRXL domains. Double cysteine point mutants that engineer extra interdomain disulfide bridges rigidify the UGGT structure and exhibit impaired activity. The intrinsic flexibility of the TRXL domains of UGGT may therefore endow the enzyme with the promiscuity needed to recognize and reglucosylate its many different substrates and/or enable reglucosylation of N-linked glycans situated at variable distances from the site of misfold.


Subject(s)
Glucosyltransferases/chemistry , Glucosyltransferases/physiology , Animals , Chaetomium/genetics , Chaetomium/metabolism , Crystallography, X-Ray/methods , Endoplasmic Reticulum/metabolism , Eukaryota/metabolism , Eukaryotic Cells/metabolism , Glucosyltransferases/metabolism , Glycoproteins/metabolism , Molecular Conformation , Protein Domains/physiology , Protein Folding , Protein Transport/physiology , Substrate Specificity
10.
Angew Chem Int Ed Engl ; 59(52): 23544-23548, 2020 12 21.
Article in English | MEDLINE | ID: mdl-32841477

ABSTRACT

The SARS-CoV-2 main protease (Mpro ) cleaves along the two viral polypeptides to release non-structural proteins required for viral replication. MPro is an attractive target for antiviral therapies to combat the coronavirus-2019 disease. Here, we used native mass spectrometry to characterize the functional unit of Mpro . Analysis of the monomer/dimer equilibria reveals a dissociation constant of Kd =0.14±0.03 µM, indicating MPro has a strong preference to dimerize in solution. We characterized substrate turnover rates by following temporal changes in the enzyme-substrate complexes, and screened small molecules, that bind distant from the active site, for their ability to modulate activity. These compounds, including one proposed to disrupt the dimer, slow the rate of substrate processing by ≈35 %. This information, together with analysis of the x-ray crystal structures, provides a starting point for the development of more potent molecules that allosterically regulate MPro activity.


Subject(s)
Coronavirus 3C Proteases/chemistry , Coronavirus Protease Inhibitors/chemistry , Models, Molecular , SARS-CoV-2/enzymology , Small Molecule Libraries/chemistry , Allosteric Regulation , Binding Sites , Biological Assay , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus Protease Inhibitors/pharmacology , Crystallography, X-Ray , Mass Spectrometry , Protein Binding , Protein Conformation , Protein Multimerization , SARS-CoV-2/physiology , Small Molecule Libraries/pharmacology , Substrate Specificity , Virus Replication
12.
Proc Natl Acad Sci U S A ; 113(32): E4630-8, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27462106

ABSTRACT

The biosynthesis of enveloped viruses depends heavily on the host cell endoplasmic reticulum (ER) glycoprotein quality control (QC) machinery. This dependency exceeds the dependency of host glycoproteins, offering a window for the targeting of ERQC for the development of broad-spectrum antivirals. We determined small-angle X-ray scattering (SAXS) and crystal structures of the main ERQC enzyme, ER α-glucosidase II (α-GluII; from mouse), alone and in complex with key ligands of its catalytic cycle and antiviral iminosugars, including two that are in clinical trials for the treatment of dengue fever. The SAXS data capture the enzyme's quaternary structure and suggest a conformational rearrangement is needed for the simultaneous binding of a monoglucosylated glycan to both subunits. The X-ray structures with key catalytic cycle intermediates highlight that an insertion between the +1 and +2 subsites contributes to the enzyme's activity and substrate specificity, and reveal that the presence of d-mannose at the +1 subsite renders the acid catalyst less efficient during the cleavage of the monoglucosylated substrate. The complexes with iminosugar antivirals suggest that inhibitors targeting a conserved ring of aromatic residues between the α-GluII +1 and +2 subsites would have increased potency and selectivity, thus providing a template for further rational drug design.


Subject(s)
Antiviral Agents/pharmacology , Endoplasmic Reticulum/enzymology , Glycoside Hydrolase Inhibitors/pharmacology , alpha-Glucosidases/chemistry , Animals , Catalysis , Crystallography, X-Ray , Mice , Protein Conformation , Protein Subunits , Scattering, Small Angle , Substrate Specificity
13.
Int J Mol Sci ; 20(19)2019 Sep 26.
Article in English | MEDLINE | ID: mdl-31561440

ABSTRACT

Claudins regulate paracellular permeability in different tissues. The claudin-binding domain of Clostridium perfringens enterotoxin (cCPE) is a known modulator of a claudin subset. However, it does not efficiently bind to claudin-1 (Cldn1). Cldn1 is a pharmacological target since it is (i) an essential co-receptor for hepatitis C virus (HCV) infections and (ii) a key element of the epidermal barrier limiting drug delivery. In this study, we investigated the potential of a Cldn1-binding cCPE mutant (i) to inhibit HCV entry into hepatocytes and (ii) to open the epidermal barrier. Inhibition of HCV infection by blocking of Cldn1 with cCPE variants was analyzed in the Huh7.5 hepatoma cell line. A model of reconstructed human epidermis was used to investigate modulation of the epidermal barrier by cCPE variants. In contrast to cCPEwt, the Cldn1-binding cCPE-S305P/S307R/S313H inhibited infection of Huh7.5 cells with HCV in a dose-dependent manner. In addition, TJ modulation by cCPE variant-mediated targeting of Cldn1 and Cldn4 opened the epidermal barrier in reconstructed human epidermis. cCPE variants are potent claudin modulators. They can be applied for mechanistic in vitro studies and might also be used as biologics for therapeutic claudin targeting including HCV treatment (host-targeting antivirals) and improvement of drug delivery.


Subject(s)
Claudins/metabolism , Enterotoxins/metabolism , Hepatocytes/metabolism , Skin/metabolism , Amino Acid Substitution , Cell Line, Tumor , Claudins/chemistry , Enterotoxins/chemistry , Enterotoxins/pharmacology , Epidermis/metabolism , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Models, Molecular , Molecular Conformation , Protein Binding , Skin/cytology , Virus Internalization/drug effects , Virus Replication
14.
J Proteome Res ; 17(3): 987-999, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29420040

ABSTRACT

Broadly neutralizing antibodies (bNAbs) that target the trimeric HIV-1 envelope glycoprotein spike (Env) are tools that can guide the design of recombinant Env proteins intended to engage the predicted human germline precursors of bNAbs (gl-bNAbs). The protein components of gl-bNAb epitopes are often masked by glycans, while mature bNAbs can evolve to accommodate or bypass these shielding glycans. The design of germline-targeting Env immunogens therefore includes the targeted deletion of specific glycan sites. However, the processing of glycans on Env trimers can be influenced by the density with which they are packed together, a highly relevant point given the essential contributions under-processed glycans make to multiple bNAb epitopes. We sought to determine the impact of the removal of 15 potential N-glycan sites (5 per protomer) from the germline-targeting soluble trimer, BG505 SOSIP.v4.1-GT1, using quantitative, site-specific N-glycan mass spectrometry analysis. We find that, compared with SOSIP.664, there was little overall change in the glycan profile but only subtle increases in the extent of processing at sites immediately adjacent to where glycans had been deleted. We conclude that multiple glycans can be deleted from BG505 SOSIP trimers without perturbing the overall integrity of the glycan shield.


Subject(s)
Antibodies, Neutralizing/chemistry , Epitopes/chemistry , HIV Antibodies/chemistry , HIV-1/metabolism , Polysaccharides/chemistry , Protein Processing, Post-Translational , env Gene Products, Human Immunodeficiency Virus/chemistry , Amino Acid Motifs , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites , CHO Cells , Carbohydrate Sequence , Cell Lineage/immunology , Cricetulus , Epitopes/genetics , Epitopes/immunology , Epitopes/metabolism , Gene Expression , Glycosylation , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Antibodies/metabolism , HIV-1/genetics , HIV-1/immunology , Polysaccharides/immunology , Polysaccharides/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism
17.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27807235

ABSTRACT

The formation of a correctly folded and natively glycosylated HIV-1 viral spike is dependent on protease cleavage of the gp160 precursor protein in the Golgi apparatus. Cleavage induces a compact structure which not only renders the spike capable of fusion but also limits further maturation of its extensive glycosylation. The redirection of the glycosylation pathway to preserve underprocessed oligomannose-type glycans is an important feature in immunogen design, as glycans contribute to or influence the epitopes of numerous broadly neutralizing antibodies. Here we present a quantitative site-specific analysis of a recombinant, trimeric mimic of the native HIV-1 viral spike (BG505 SOSIP.664) compared to the corresponding uncleaved pseudotrimer and the matched gp120 monomer. We present a detailed molecular map of a trimer-associated glycan remodeling that forms a localized subdomain of the native mannose patch. The formation of native trimers is a critical design feature in shaping the glycan epitopes presented on recombinant vaccine candidates. IMPORTANCE: The envelope spike of human immunodeficiency virus type 1 (HIV-1) is a target for antibody-based neutralization. For some patients infected with HIV-1, highly potent antibodies have been isolated that can neutralize a wide range of circulating viruses. It is a goal of HIV-1 vaccine research to elicit these antibodies by immunization with recombinant mimics of the viral spike. These antibodies have evolved to recognize the dense array of glycans that coat the surface of the viral molecule. We show how the structure of these glycans is shaped by steric constraints imposed upon them by the native folding of the viral spike. This information is important in guiding the development of vaccine candidates.


Subject(s)
HIV-1/metabolism , Mannose/metabolism , Models, Molecular , Protein Conformation , Protein Multimerization , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/metabolism , Glycosylation , Humans , Polysaccharides/metabolism , Protein Binding , Proteolysis , Structure-Activity Relationship
18.
Cell Microbiol ; 19(5)2017 05.
Article in English | MEDLINE | ID: mdl-28207988

ABSTRACT

Symptomatic dengue virus (DENV) infections range from mild fever to severe haemorrhagic disease and death. Host-viral interactions play a significant role in deciding the fate of the infection. The unfolded protein response (UPR) is a prosurvival cellular reaction induced in response to DENV-mediated endoplasmic reticulum stress. The UPR has complex interactions with the cellular autophagy machinery, apoptosis, and innate immunity. DENV has evolved to manipulate the UPR to facilitate its replication and to evade host immunity. Our knowledge of this intertwined network of events is continuously developing. A better understanding of the UPR mediated antiviral and proviral effects will shed light on dengue disease pathogenesis and may help development of anti-DENV therapeutics. This review summarizes the role of the UPR in viral replication, autophagy, and DENV-induced inflammation to describe how a host response contributes to DENV pathogenesis.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Unfolded Protein Response/immunology , Animals , Apoptosis , Autophagy , Dengue/pathology , Host-Pathogen Interactions , Humans , Immunity, Innate , Virus Replication
19.
Virol J ; 15(1): 193, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30587198

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has become a global threat with immediate need for accurate diagnostics, efficacious vaccines and therapeutics. Several ZIKV envelope (Env)-based vaccines have been developed recently. However, many commercially available ZIKV Env are based on the African lineage and produced in insect cells. Here, we sought to produce Asian-lineage ZIKV Env in mammalian cells for research and clinical applications. METHODS: We designed various gene expression constructs to optimize the production of ZIKV using prM-Env and full or C-terminal truncations of Env; with or without a rat CD4 fusion partner to allow large-scale production of soluble protein in mammalian HEK293 cells. Protein expression was verified by mass spectrometry and western-blot with a pan-flavivirus antibody, a ZIKV Env monoclonal antibody and with immune sera from adenoviral (ChAdOx1) ZIKV Env-vaccinated mice. The resulting Env-CD4 was used as a coating reagent for immunoassay (ELISA) using both mouse and human seropositive sera. RESULTS: Replacement of the C-terminus transmembrane Env domain by a rat CD4 and addition of prM supported optimal expression and secretion of Env. Binding between the antigens and the antibodies was similar to binding when using commercially available ZIKV Env reagents. Furthermore, antibodies from ZIKV patients bound ZIKV Env-CD4 in ELISA assays, whereas sera from healthy blood donors yielded minimal OD background. The serological outcomes of this assay correlated also with ZIKV neutralisation capacity in vitro. CONCLUSIONS: Results obtained from this study indicate the potential of the Asian-lineage Zika Env-CD4 and Env proteins in ELISA assays to monitor humoral immune responses in upcoming clinical trials as well as a sero-diagnostic tool in ZIKV infection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Recombinant Fusion Proteins/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/isolation & purification , Zika Virus/immunology , Animals , CD4 Antigens/genetics , Enzyme-Linked Immunosorbent Assay/methods , HEK293 Cells , Humans , Mexico , Mice , Neutralization Tests/methods , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Zika Virus/genetics
20.
Adv Exp Med Biol ; 1062: 277-301, 2018.
Article in English | MEDLINE | ID: mdl-29845540

ABSTRACT

The antiviral mechanism of action of iminosugars against many enveloped viruses, including dengue virus (DENV), HIV, influenza and hepatitis C virus, is believed to be mediated by inducing misfolding of viral N-linked glycoproteins through inhibition of host endoplasmic reticulum-resident α-glucosidase enzymes. This leads to reduced secretion and/or infectivity of virions and hence lower viral titres, both in vitro and in vivo. Free oligosaccharide analysis from iminosugar-treated cells shows that antiviral activity correlates with production of mono- and tri-glucosylated sugars, indicative of inhibition of ER α-glucosidases. We demonstrate that glucose-mimicking iminosugars inhibit isolated glycoprotein and glycolipid processing enzymes and that this inhibition also occurs in primary cells treated with these drugs. Galactose-mimicking iminosugars that have been tested do not inhibit glycoprotein processing but do inhibit glycolipid processing, and are not antiviral against DENV. By comparison, the antiviral activity of glucose-mimetic iminosugars that inhibit endoplasmic reticulum-resident α-glucosidases, but not glycolipid processing, demonstrates that inhibition of α-glucosidases is responsible for iminosugar antiviral activity against DENV. This monograph will review the investigations of many researchers into the mechanisms of action of iminosugars and the contribution of our current understanding of these mechanisms for optimising clinical delivery of iminosugars. The effects of iminosugars on enzymes other than glucosidases, the induction of ER stress and viral receptors will be also put into context. Data suggest that inhibition of α-glucosidases results in inhibited release of virus and is the primary antiviral mechanism of action of iminosugars against DENV.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Sugars/immunology , Animals , Dengue/enzymology , Dengue/genetics , Dengue/virology , Dengue Virus/genetics , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/virology , Humans , alpha-Glucosidases/genetics , alpha-Glucosidases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL