Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Biol Open ; 11(9)2022 09 15.
Article in English | MEDLINE | ID: mdl-35929543

ABSTRACT

Enterovirus 71 (EV71) is one of the causative agents of hand-foot-and-mouth disease, which in some circumstances could lead to severe neurological diseases. Despite of its importance for human health, little is known about the early stages of EV71 infection. EV71 starts uncoating with its receptor, human scavenger receptor B2 (hSCARB2), at low pH. We show that EV71 was not targeted to lysosomes in human rhabdomyosarcoma cells overexpressing hSCARB2 and that the autophagic pathway is not essential for EV71 productive uncoating. Instead, EV71 was efficiently uncoated 30 min after infection in late endosomes (LEs) containing hSCARB2, mannose-6-phosphate receptor (M6PR), RAB9, bis(monoacylglycero)phosphate and lysosomal associated membrane protein 2 (LAMP2). Furthering the notion that mature LEs are crucial for EV71 uncoating, cation-dependent (CD)-M6PR knockdown impairs EV71 infection. Since hSCARB2 interacts with cation-independent (CI)-M6PR through M6P-binding sites and CD-M6PR also harbor a M6P-binding site, CD-M6PR is likely to play important roles in EV71 uncoating in LEs.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Animals , Cations/metabolism , Endosomes/metabolism , Enterovirus/metabolism , Enterovirus A, Human/metabolism , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Receptor, IGF Type 2/metabolism , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism
2.
Methods Mol Biol ; 2304: 157-172, 2021.
Article in English | MEDLINE | ID: mdl-34028716

ABSTRACT

High-resolution confocal imaging has provided new insights in the process of receptor-mediated endocytosis in variety of cell types. We describe here the protocol for investigating B cell receptor (BCR)-mediated internalization of membrane bound antigens using confocal microscopy. We describe the method to prepare plasma membrane sheets (PMS) in a small area, bind fluorescently tagged antigens to the PMS and activate B cells on the PMS. We also describe the method for analyzing antigen internalization using confocal microscopy and computational image analysis. This protocol is useful for the study of antigen internalization by B cells and can be applied for studying receptor-mediated endocytosis in other cells as well. The setup we describe here is especially useful for studying rare cell types when the number of cells available is limiting.


Subject(s)
Antigens/metabolism , B-Lymphocytes/immunology , Receptors, Antigen, B-Cell/immunology , Antigens/chemistry , Cell Membrane/immunology , Computational Biology , Endocytosis , Fluorescent Dyes/chemistry , HEK293 Cells , HLA-D Antigens/chemistry , HLA-D Antigens/metabolism , HLA-DR Antigens/chemistry , HLA-DR Antigens/metabolism , Humans , Lymphocyte Activation , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/metabolism , Microscopy, Confocal
3.
Biochem Biophys Res Commun ; 524(2): 424-430, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32007273

ABSTRACT

LGP85/LIMP-2 is a type III transmembrane glycoprotein of lysosomes, which traverses the membrane twice with an N-terminal uncleaved signal sequence and C-terminal hydrophobic domain. In addition to functioning as a receptor for a lysosomal enzyme ß-glucocerebrosidase and for several enteroviruses, LGP85 plays a key role in the biogenesis and maintenance of endosomal/lysosomal compartments (ELCs). Our previous studies have demonstrated that overexpression of rat LGP85 into COS cells results in the enlarged ELCs, from where membrane trafficking is impaired. We show here that rat LGP85 is polyubiquitinated at the N-terminal short cytoplasmic domain that comprises of only three amino acid residues, alanine, arginine, and cysteine. Replacement of either arginine or cysteine with alanine within the N-terminal cytoplasmic domain did not influence the ubiquitination of LGP85, thereby indicating that ubiquitin (Ub) is conjugated to the α-NH2 group of the N-terminal alanine residue. Furthermore, we were able to define a domain necessary for ubiquitination in a region ranging from the amino acids 156 to 255 within the lumenal domain of LGP85. This is the first report showing that the integral lysosomal membrane protein LGP85 is ubiquitinated.


Subject(s)
CD36 Antigens/metabolism , Lysosomal Membrane Proteins/metabolism , Ubiquitination , Animals , CD36 Antigens/chemistry , COS Cells , Chlorocebus aethiops , Lysosomal Membrane Proteins/chemistry , Lysosomes/metabolism , Protein Domains , Rats , Ubiquitinated Proteins/chemistry , Ubiquitinated Proteins/metabolism
4.
FEBS J ; 287(9): 1777-1797, 2020 05.
Article in English | MEDLINE | ID: mdl-31804757

ABSTRACT

Development of human leishmaniasis is dependent on the ability of intracellular Leishmania parasites to spread and enter macrophages. The mechanism through which free promastigotes and amastigotes bind and enter host macrophages has been previously investigated; however, little is known about intracellular trafficking and cell-to-cell spreading. In this study, the mechanism involved in the spreading of Leishmania aethiopica and Leishmania mexicana was investigated. A significant increase in phosphatidylserine (PS) exhibition, cytochrome C release, and active caspase-3 expression was detected (P < 0.05) during L. aethiopica, but not L. mexicana spreading. A decrease (P < 0.05) of protein kinase B (Akt) protein and BCL2-associated agonist of cell death (BAD) phosphorylation was also observed. The nuclear factor kappa-light-chain enhancer of activated B cells (NF-kB) signaling pathway and pro-apoptotic protein protein kinase C delta (PKC-δ) were downregulated while inhibition of caspase-3 activation prevented L. aethiopica spreading. Overall suggesting that L. aethiopica induces host cell's apoptosis during spreading in a caspase-3-dependent manner. The trafficking of amastigotes within macrophages following cell-to-cell spreading differed from that of axenic parasites and involved co-localization with lysosomal-associated membrane protein 1 (LAMP-1) within 10 min postinfection. Interestingly, following infection with axenic amastigotes and promastigotes, co-localization of parasites with LAMP-1-positive structures took place at 1 and 4 h, respectively, suggesting that the membrane coat and LAMP-1 protein were derived from the donor cell. Collectively, these findings indicate that host cell apoptosis, demonstrated by PS exhibition, caspase-3 activation, cytochrome C release, downregulation of Akt, BAD phosphorylation, NF-kB activation, and independent of PKC-δ expression, is involved in L. aethiopica spreading. Moreover, L. aethiopica parasites associate with LAMP-rich structures when taken up by neighboring macrophages.


Subject(s)
Caspase 3/metabolism , Leishmania/metabolism , Lysosomal Membrane Proteins/metabolism , NF-kappa B/metabolism , Protein Kinase C-delta , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Humans , Lysosomal Membrane Proteins/chemistry , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , THP-1 Cells
5.
Mol Brain ; 12(1): 20, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30867010

ABSTRACT

Lysosome-associated membrane protein 5 (LAMP5) is a mammalian ortholog of the Caenorhabditis elegans protein, UNC-46, which functions as a sorting factor to localize the vesicular GABA transporter UNC-47 to synaptic vesicles. In the mouse forebrain, LAMP5 is expressed in a subpopulation of GABAergic neurons in the olfactory bulb and the striato-nigral system, where it is required for fine-tuning of GABAergic synaptic transmission. Here we focus on the prominent expression of LAMP5 in the brainstem and spinal cord and suggest a role for LAMP5 in these brain regions. LAMP5 was highly expressed in several brainstem nuclei involved with auditory processing including the cochlear nuclei, the superior olivary complex, nuclei of the lateral lemniscus and grey matter in the spinal cord. It was localized exclusively in inhibitory synaptic terminals, as has been reported in the forebrain. In the absence of LAMP5, localization of the vesicular inhibitory amino acid transporter (VIAAT) was unaltered in the lateral superior olive and the ventral cochlear nuclei, arguing against a conserved role for LAMP5 in trafficking VIAAT. Lamp5 knockout mice showed no overt behavioral abnormality but an increased startle response to auditory and tactile stimuli. In addition, LAMP5 deficiency led to a larger intensity-dependent increase of wave I, II and V peak amplitude of auditory brainstem response. Our results indicate that LAMP5 plays a pivotal role in sensorimotor processing in the brainstem and spinal cord.


Subject(s)
Auditory Pathways , Lysosomal Membrane Proteins/metabolism , Neural Inhibition , Presynaptic Terminals/metabolism , Reflex, Startle , Rhombencephalon/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Behavior, Animal , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism
6.
Nat Microbiol ; 4(3): 414-419, 2019 03.
Article in English | MEDLINE | ID: mdl-30531980

ABSTRACT

Enterovirus 71 (EV71) is a common cause of hand, foot and mouth disease-a disease endemic especially in the Asia-Pacific region1. Scavenger receptor class B member 2 (SCARB2) is the major receptor of EV71, as well as several other enteroviruses responsible for hand, foot and mouth disease, and plays a key role in cell entry2. The isolated structures of EV71 and SCARB2 are known3-6, but how they interact to initiate infection is not. Here, we report the EV71-SCARB2 complex structure determined at 3.4 Å resolution using cryo-electron microscopy. This reveals that SCARB2 binds EV71 on the southern rim of the canyon, rather than across the canyon, as predicted3,7,8. Helices 152-163 (α5) and 183-193 (α7) of SCARB2 and the viral protein 1 (VP1) GH and VP2 EF loops of EV71 dominate the interaction, suggesting an allosteric mechanism by which receptor binding might facilitate the low-pH uncoating of the virus in the endosome/lysosome. Remarkably, many residues within the binding footprint are not conserved across SCARB2-dependent enteroviruses; however, a conserved proline and glycine seem to be key residues. Thus, although the virus maintains antigenic variability even within the receptor-binding footprint, the identification of binding 'hot spots' may facilitate the design of receptor mimic therapeutics less likely to quickly generate resistance.


Subject(s)
Enterovirus/metabolism , Host Microbial Interactions , Lysosomal Membrane Proteins/chemistry , Receptors, Scavenger/chemistry , Viral Proteins/chemistry , Cryoelectron Microscopy , Enterovirus/ultrastructure , Humans , Virus Attachment
7.
Acta Virol ; 62(1): 50-57, 2018.
Article in English | MEDLINE | ID: mdl-29521103

ABSTRACT

There is still no effective clinical antiviral drug against human enterovirus 71 (EV71) infection, which causes hand, foot and mouth disease (HFMD) in children. Scavenger receptor class B member 2 (SCARB2) is an important receptor of EV71 as it plays a vital role in the early steps of viral infection. In this study, recombinant SCARB2 protein was expressed and purified in a prokaryotic expression system, and was identified by western blot with a monoclonal antibody and mass spectrometry analysis. Detection of the sera from mice immunized with the recombinant SCARB2 protein using ELISA and western blot showed good immunogenicity of the recombinant protein. Furthermore, in the neutralization test cytopathic effect was significantly decreased when EV71 was incubated with the immune sera before infection. In summary, the SCARB2 protein was expressed successfully, and the immune sera showed obvious antiviral effect against EV71. This study provides useful information about the interaction mechanism between SCARB2 and EV71, and is also helpful for further clinical treatment research of HFMD.


Subject(s)
Enterovirus A, Human/metabolism , Lysosomal Membrane Proteins/immunology , Receptors, Scavenger/immunology , Animals , Antibodies, Viral , Base Sequence , Cell Line, Tumor , Enterovirus A, Human/chemistry , Enterovirus A, Human/immunology , Enzyme-Linked Immunosorbent Assay , Hand, Foot and Mouth Disease/metabolism , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/metabolism , Mice , Neutralization Tests , Protein Binding , Receptors, Scavenger/chemistry , Receptors, Scavenger/metabolism , Recombinant Proteins , Viral Vaccines/immunology
8.
Anal Chem ; 89(10): 5373-5381, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28414218

ABSTRACT

Enterovirus 71 (EV71) is a major public health threat that requires rapid point-of-care detection. Here, we developed a surface-enhanced Raman spectroscopy (SERS)-based scheme that utilized protein-induced aggregation of colloidal gold nanostars (AuNS) to rapidly detect EV71 without the need for fabricating a solid substrate, Raman labels or complicated sample handling. We used AuNS (hydrodynamic diameter, DH of 105.12 ± 1.13 nm) conjugated to recombinant scavenger receptor class B, member 2 (SCARB2) protein with known affinity to EV71. In the absence of EV71, AuNS-SCARB2 aggregated in biological media and produced four enhanced Raman peaks at 390, 510, 670, and 910 cm-1. In the presence of EV71, the three peaks at 510, 670, and 910 cm-1 disappeared, while the peak at 390 cm-1 diminished in intensity as the virus bound to AuNS-SCARB2 and prevented them from aggregation. These three peaks (510, 670, and 910 cm-1) were potential markers for specific detection of EV71 as their disappearance was not observable with a different dengue virus (DENV) as our control. Furthermore, the Raman measurements from colloidal SERS were more sensitive in probing the aggregation of AuNS-SCARB2 for detecting the presence of EV71 in protein-rich samples compared to UV-vis spectrum measurements. With this facile "anti-aggregation" approach, we were able to detect EV71 in protein-rich biological medium within 15 min with reasonable sensitivity of 107 pfu/mL and minimal sample preparation, making this translatable for point-of-care applications.


Subject(s)
Enterovirus A, Human/isolation & purification , Hand, Foot and Mouth Disease/diagnosis , Nanostructures/chemistry , Spectrum Analysis, Raman , Enterovirus A, Human/chemistry , Gold/chemistry , Hand, Foot and Mouth Disease/virology , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Protein Binding , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrophotometry
9.
Protein Cell ; 8(8): 590-600, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28447294

ABSTRACT

Entero virus 71 (EV71) causes hand, foot, and mouth disease (HFMD) and occasionally leads to severe neurological complications and even death. Scavenger receptor class B member 2 (SCARB2) is a functional receptor for EV71, that mediates viral attachment, internalization, and uncoating. However, the exact binding site of EV71 on SCARB2 is unknown. In this study, we generated a monoclonal antibody (mAb) that binds to human but not mouse SCARB2. It is named JL2, and it can effectively inhibit EV71 infection of target cells. Using a set of chimeras of human and mouse SCARB2, we identified that the region containing residues 77-113 of human SCARB2 contributes significantly to JL2 binding. The structure of the SCARB2-JL2 complex revealed that JL2 binds to the apical region of SCARB2 involving α-helices 2, 5, and 14. Our results provide new insights into the potential binding sites for EV71 on SCARB2 and the molecular mechanism of EV71 entry.


Subject(s)
Antibodies, Monoclonal/chemistry , Enterovirus A, Human/drug effects , Immunoglobulin Fab Fragments/chemistry , Lysosomal Membrane Proteins/chemistry , Receptors, Scavenger/chemistry , Receptors, Virus/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Binding Sites , Cell Line , Crystallography, X-Ray , Enterovirus A, Human/genetics , Enterovirus A, Human/growth & development , Enterovirus A, Human/immunology , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/immunology , Mice , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Scavenger/genetics , Receptors, Scavenger/immunology , Receptors, Virus/genetics , Receptors, Virus/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera , Thermodynamics
10.
PLoS Pathog ; 13(4): e1006337, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28448640

ABSTRACT

Cell entry of many enveloped viruses occurs by engagement with cellular receptors, followed by internalization into endocytic compartments and pH-induced membrane fusion. A previously unnoticed step of receptor switching was found to be critical during cell entry of two devastating human pathogens: Ebola and Lassa viruses. Our recent studies revealed the functional role of receptor switching to LAMP1 for triggering membrane fusion by Lassa virus and showed the involvement of conserved histidines in this switching, suggesting that other viruses from this family may also switch to LAMP1. However, when we investigated viruses that are genetically close to Lassa virus, we discovered that they cannot bind LAMP1. A crystal structure of the receptor-binding module from Morogoro virus revealed structural differences that allowed mapping of the LAMP1 binding site to a unique set of Lassa residues not shared by other viruses in its family, illustrating a key difference in the cell-entry mechanism of Lassa virus that may contribute to its pathogenicity.


Subject(s)
Arenaviridae Infections/virology , Arenaviruses, Old World/metabolism , Lassa Fever/virology , Lassa virus/metabolism , Lysosomal Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Arenaviruses, Old World/chemistry , Arenaviruses, Old World/genetics , Binding Sites , Humans , Lassa virus/chemistry , Lassa virus/genetics , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Membrane Fusion , Models, Molecular , Models, Structural , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment , Species Specificity
11.
J Cell Sci ; 129(11): 2135-43, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27165304

ABSTRACT

Danon disease is a rare, severe X-linked form of cardiomyopathy caused by deficiency of lysosome-associated membrane protein 2 (LAMP-2). Other clinical manifestations include skeletal myopathy, cognitive defects and visual problems. Although individuals with Danon disease have been clinically described since the early 1980s, the underlying molecular mechanisms involved in pathological progression remain poorly understood. LAMP-2 is known to be involved in autophagy, and a characteristic accumulation of autophagic vacuoles in the affected tissues further supports the idea that autophagy is disrupted in this disease. The LAMP2 gene is alternatively spliced to form three splice isoforms, which are thought to play different autophagy-related cellular roles. This Commentary explores findings from genetic, histological, functional and tissue expression studies that suggest that the specific loss of the LAMP-2B isoform, which is likely to be involved in macroautophagy, plays a crucial role in causing the Danon phenotype. We also compare findings from mouse and cellular models, which have allowed for further molecular characterization but have also shown phenotypic differences that warrant attention. Overall, there is a need to better functionally characterize the LAMP-2B isoform in order to rationally explore more effective therapeutic options for individuals with Danon disease.


Subject(s)
Autophagy , Cardiomyopathies/complications , Cardiomyopathies/pathology , Glycogen Storage Disease Type IIb/complications , Glycogen Storage Disease Type IIb/pathology , Amino Acid Sequence , Animals , Disease Models, Animal , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/metabolism , Phenotype
12.
J Biomol Screen ; 21(6): 556-66, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27048485

ABSTRACT

We previously developed an assay of cytotoxic T-lymphocyte lytic granule exocytosis based on externalization of LAMP-1/CD107A using nonphysiological stimuli to generate maximal levels of exocytosis. Here, we used polystyrene beads coated with anti-CD3 antibodies to stimulate cells. Light scatter let us distinguish cells that contacted beads from cells that had not, allowing comparison of signaling events and exocytosis from stimulated and unstimulated cells in one sample. Bead stimulation resulted in submaximal exocytosis, making it possible to detect compounds that either augment or inhibit lytic granule exocytosis. Coupled with the assay's ability to distinguish responses in cells that have and have not contacted a stimulatory bead, it is possible to detect three kinds of compounds: inhibitors, stimulators, which cause exocytosis, and augmenters, which enhance receptor-stimulated exocytosis. To validate the assay, we screened a set of synthetic compounds identified using our previous assay and a library of 320 extracts prepared from tunicate-associated bacteria. One of the extracts augmented exocytosis threefold. Activity-guided fractionation and structure elucidation revealed that this compound is the known PKC activator teleocidin A-1. We conclude that our modified assay is suitable for screening synthetic compound plates and natural product collections, and will be useful for identifying immunologically active small molecules.


Subject(s)
Biological Products/chemistry , Cytoplasmic Granules/chemistry , High-Throughput Screening Assays/methods , T-Lymphocytes, Cytotoxic/chemistry , Antibodies, Anti-Idiotypic/chemistry , Antibodies, Anti-Idiotypic/immunology , Apoptosis/drug effects , Bacteria/chemistry , Cell Extracts/chemistry , Cell Extracts/immunology , Cell Extracts/pharmacology , Cytoplasmic Granules/drug effects , Exocytosis/drug effects , Exocytosis/immunology , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lyngbya Toxins/chemistry , Lyngbya Toxins/genetics , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/immunology , Protein Kinase C/chemistry , Protein Kinase C/genetics , Signal Transduction/drug effects , T-Lymphocytes, Cytotoxic/immunology
13.
PLoS Pathog ; 12(2): e1005418, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26849049

ABSTRACT

Lassa virus is an enveloped, bi-segmented RNA virus and the most prevalent and fatal of all Old World arenaviruses. Virus entry into the host cell is mediated by a tripartite surface spike complex, which is composed of two viral glycoprotein subunits, GP1 and GP2, and the stable signal peptide. Of these, GP1 binds to cellular receptors and GP2 catalyzes fusion between the viral envelope and the host cell membrane during endocytosis. The molecular structure of the spike and conformational rearrangements induced by low pH, prior to fusion, remain poorly understood. Here, we analyzed the three-dimensional ultrastructure of Lassa virus using electron cryotomography. Sub-tomogram averaging yielded a structure of the glycoprotein spike at 14-Å resolution. The spikes are trimeric, cover the virion envelope, and connect to the underlying matrix. Structural changes to the spike, following acidification, support a viral entry mechanism dependent on binding to the lysosome-resident receptor LAMP1 and further dissociation of the membrane-distal GP1 subunits.


Subject(s)
Glycoproteins/metabolism , Lassa virus/metabolism , Lysosomal Membrane Proteins/metabolism , Protein Sorting Signals , Viral Envelope Proteins/metabolism , Animals , Chlorocebus aethiops , Glycoproteins/chemistry , Hydrogen-Ion Concentration , Lassa virus/chemistry , Lassa virus/ultrastructure , Lysosomal Membrane Proteins/chemistry , Models, Molecular , Molecular Conformation , Multiprotein Complexes , Protein Binding , Protein Structure, Tertiary , Vero Cells , Viral Envelope Proteins/chemistry , Virion , Virus Internalization
14.
Sci Rep ; 5: 15069, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26472258

ABSTRACT

Our previously reported gene atlasing of schistosome tissues revealed transcripts that were highly enriched in the digestive tract of Schistosoma mansoni. From these, we selected two candidates, Sm-LAMP and Sm-NPC2 for testing as vaccine targets. The two molecules were selected on the basis of relatively high expression in the gastrodermis, their potentially important biological function, divergence from homologous molecules of the host and possible apical membrane expression in the gastrodermis. Bacterially expressed recombinant peptides corresponding to regions excluding trans-membrane domains of the selected vaccine targets were used in blinded vaccine trials in CBA mice using alum-CpG as adjuvant. Vaccine trials using the recombinant insoluble Sm-LAMP protein showed 16-25% significant reduction in total worm burden. Faecal egg count reduction was 52% and 60% in two trials, respectively, with similar results for the solubly expressed protein. Liver egg burden was reduced significantly (20% and 38%) with an insoluble recombinant Sm-LAMP in two trials, but not with the soluble recombinant form. Parasite fecundity was not affected by either Sm-LAMP protein preparations in the trials. It is concluded that Sm-LAMP may provide limited protection towards S. mansoni infections but could be used in combination with other vaccine candidates, to provide more comprehensive protection.


Subject(s)
Antigens, Helminth/immunology , Lysosomal Membrane Proteins/immunology , Protozoan Vaccines/immunology , Schistosoma mansoni/immunology , Schistosomiasis/prevention & control , Amino Acid Sequence , Animals , Disease Models, Animal , Female , Gene Expression , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/classification , Lysosomal Membrane Proteins/genetics , Male , Mice , Molecular Sequence Data , Parasite Egg Count , Parasite Load , Phylogeny , Protein Transport , Recombinant Proteins , Schistosomiasis/parasitology
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(5): 723-7, 2015 Oct.
Article in Chinese | MEDLINE | ID: mdl-26419000

ABSTRACT

SCARB2 (scavenger receptor class B, member 2) is a lysosomal membrane glucoprotein, which is encoded by SCARB2 gene. It takes vital parts in the physiological and pathological processes including the transportation of beta-glucocerebrosidase to the lysosome, infection of EV71 and load-induced cardiac myocyte hypertrophy. This article has reviewed the molecular structure and functions of SCARB2 gene and its protein, as well as their relationship with diseases.


Subject(s)
Lysosomal Membrane Proteins/physiology , Receptors, Scavenger/physiology , Hand, Foot and Mouth Disease/genetics , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Myoclonic Epilepsies, Progressive/genetics , Parkinson Disease/genetics , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics
16.
J Virol ; 89(20): 10347-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26246576

ABSTRACT

UNLABELLED: As a recycling center, lysosomes are filled with numerous acid hydrolase enzymes that break down waste materials and invading pathogens. Recently, lysosomal cell death has been defined as "lysosomal membrane permeabilization and the consequent leakage of lysosome contents into cytosol." Here, we show that the neuraminidase (NA) of H5N1 influenza A virus markedly deglycosylates and degrades lysosome-associated membrane proteins (LAMPs; the most abundant membrane proteins of lysosome), which induces lysosomal rupture, and finally leads to cell death of alveolar epithelial carcinoma A549 cells and human tracheal epithelial cells. The NA inhibitors peramivir and zanamivir could effectively block the deglycosylation of LAMPs, inhibit the virus cell entry, and prevent cell death induced by the H5N1 influenza virus. The NA of seasonal H1N1 virus, however, does not share these characteristics. Our findings not only reveal a novel role of NA in the early stage of the H5N1 influenza virus life cycle but also elucidate the molecular mechanism of lysosomal rupture crucial for influenza virus induced cell death. IMPORTANCE: The integrity of lysosomes is vital for maintaining cell homeostasis, cellular defense and clearance of invading pathogens. This study shows that the H5N1 influenza virus could induce lysosomal rupture through deglycosylating lysosome-associated membrane proteins (LAMPs) mediated by the neuraminidase activity of NA protein. NA inhibitors such as peramivir and zanamivir could inhibit the deglycosylation of LAMPs and protect lysosomes, which also further interferes with the H5N1 influenza virus infection at early stage of life cycle. This work is significant because it presents new concepts for NA's function, as well as for influenza inhibitors' mechanism of action, and could partially explain the high mortality and high viral load after H5N1 virus infection in human beings and why NA inhibitors have more potent therapeutic effects for lethal avian influenza virus infections at early stage.


Subject(s)
Cell Membrane/enzymology , Lysosomal Membrane Proteins/metabolism , Lysosomes/enzymology , Neuraminidase/metabolism , Viral Proteins/metabolism , Acids, Carbocyclic , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/chemistry , Cyclopentanes/pharmacology , Cytosol/drug effects , Cytosol/enzymology , Cytosol/virology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Guanidines/pharmacology , Humans , Hydrolysis , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Lysosomal Membrane Proteins/chemistry , Lysosomes/drug effects , Lysosomes/virology , Protein Binding , Proteolysis , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Species Specificity , Virus Internalization/drug effects , Zanamivir/pharmacology
17.
J Virol ; 89(15): 7584-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972533

ABSTRACT

UNLABELLED: Lassa virus is a notorious human pathogen that infects many thousands of people each year in West Africa, causing severe viral hemorrhagic fevers and significant mortality. The surface glycoprotein of Lassa virus mediates receptor recognition through its GP1 subunit. Here we report the crystal structure of GP1 from Lassa virus, which is the first representative GP1 structure for Old World arenaviruses. We identify a unique triad of histidines that forms a binding site for LAMP1, a known lysosomal protein recently discovered to be a critical receptor for internalized Lassa virus at acidic pH. We demonstrate that mutation of this histidine triad, which is highly conserved among Old World arenaviruses, impairs LAMP1 recognition. Our biochemical and structural data further suggest that GP1 from Lassa virus may undergo irreversible conformational changes that could serve as an immunological decoy mechanism. Together with a variable region that we identify on the surface of GP1, those could be two distinct mechanisms that Lassa virus utilizes to avoid antibody-based immune response. IMPORTANCE: Structural data at atomic resolution for viral proteins is key for understanding their function at the molecular level and can facilitate novel avenues for combating viral infections. Here we used X-ray protein crystallography to decipher the crystal structure of the receptor-binding domain (GP1) from Lassa virus. This is a pathogenic virus that causes significant illness and mortality in West Africa. This structure reveals the overall architecture of GP1 domains from the group of viruses known as the Old World arenaviruses. Using this structural information, we elucidated the mechanisms for pH switch and binding of Lassa virus to LAMP1, a recently identified host receptor that is critical for successful infection. Lastly, our structural analysis suggests two novel immune evasion mechanisms that Lassa virus may utilize to escape antibody-based immune response.


Subject(s)
Lassa Fever/metabolism , Lassa virus/metabolism , Lysosomal Membrane Proteins/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Cell Line , Humans , Lassa Fever/genetics , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/genetics , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Alignment , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
18.
Biochem Biophys Res Commun ; 457(3): 334-40, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25576872

ABSTRACT

The lysosomal integral membrane protein type-2 (LIMP-2/SCARB2) has been identified as a receptor for enterovirus 71 uptake and mannose-6-phosphate-independent lysosomal trafficking of the acid hydrolase ß-glucocerebrosidase. Here we show that LIMP-2 undergoes proteolytic cleavage mediated by lysosomal cysteine proteases. Heterologous expression and in vitro studies suggest that cathepsin-F is mainly responsible for the lysosomal processing of wild-type LIMP-2. Furthermore, examination of purified lysosomes revealed that LIMP-2 undergoes proteolysis in vivo. Mutations in the gene encoding cathepsin-F (CTSF) have recently been associated with type-B-Kufs-disease, an adult form of neuronal ceroid-lipofuscinosis. In this study we show that disease-causing cathepsin-F mutants fail to cleave LIMP-2. Our findings provide evidence that LIMP-2 represents an in vivo substrate of cathepsin-F with relevance for understanding the pathophysiology of type-B-Kufs-disease.


Subject(s)
Cathepsin F/genetics , Cathepsin F/metabolism , Lysosomal Membrane Proteins/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Receptors, Scavenger/metabolism , Animals , CD36 Antigens/chemistry , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , HEK293 Cells , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Lysosomes/metabolism , Mice , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Proteolysis , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
19.
Protein Cell ; 5(9): 692-703, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24986489

ABSTRACT

Unlike the well-established picture for the entry of enveloped viruses, the mechanism of cellular entry of non-enveloped eukaryotic viruses remains largely mysterious. Picornaviruses are representative models for such viruses, and initiate this entry process by their functional receptors. Here we present the structural and functional studies of SCARB2, a functional receptor of the important human enterovirus 71 (EV71). SCARB2 is responsible for attachment as well as uncoating of EV71. Differences in the structures of SCARB2 under neutral and acidic conditions reveal that SCARB2 undergoes a pivotal pH-dependent conformational change which opens a lipid-transfer tunnel to mediate the expulsion of a hydrophobic pocket factor from the virion, a pre-requisite for uncoating. We have also identified the key residues essential for attachment to SCARB2, identifying the canyon region of EV71 as mediating the receptor interaction. Together these results provide a clear understanding of cellular attachment and initiation of uncoating for enteroviruses.


Subject(s)
Enterovirus A, Human/metabolism , Lysosomal Membrane Proteins/metabolism , Receptors, Scavenger/metabolism , Virion/metabolism , Virus Attachment , Acids/chemistry , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , HEK293 Cells , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Molecular Docking Simulation , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Sequence Homology, Amino Acid , Sf9 Cells , Static Electricity , Virion/genetics
20.
J Biochem ; 156(1): 39-49, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24695761

ABSTRACT

Lysosome-associated membrane protein-1 (LAMP-1) is a type I transmembrane protein with a short cytoplasmic tail that possesses a lysosome-targeting signal of GYQTI(382)-COOH. Wild-type (WT)-LAMP-1 was exclusively localized in high density lysosomes, and efficiency of LAMP-1's transport to lysosomes depends on its COOH-terminal amino acid residue. Among many different COOH-terminal amino acid substitution mutants of LAMP-1, a leucine-substituted mutant (I382L) displays the most efficient targeting to late endosomes and lysosomes [Akasaki et al. (2010) J. Biochem. 148: , 669-679]. In this study, we generated two human hepatoma cell lines (HepG2 cell lines) that stably express WT-LAMP-1 and I382L, and compared their intracellular distributions. The subcellular fractionation study using Percoll density gradient centrifugation revealed that WT-LAMP-1 had preferential localization in the high density secondary lysosomes where endogenous human LAMP-1 was enriched. In contrast, a major portion of I382L was located in a low density fraction. The low density fraction also contained approximately 80% of endogenous human LAMP-1 and significant amounts of endogenous ß-glucuronidase and LAMP-2, which probably represents occurrence of low density lysosomes in the I382L-expressing cells. Double immunofluorescence microscopic analyses distinguished I382L-containing intracellular vesicles from endogenous LAMP-1-containing lysosomes and early endosomes. Altogether, constitutive expression of I382L causes its aberrant intracellular localization and generation of low density lysosomes, indicating that the COOH-terminal isoleucine is critical for normal localization of LAMP-1 in the dense lysosomes.


Subject(s)
Leucine/genetics , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/chemistry , Lysosomes/metabolism , Mutation/genetics , Hep G2 Cells , Humans , Lysosomal Membrane Proteins/chemistry , Protein Transport , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL