Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Chembiochem ; 22(21): 3037-3041, 2021 11 03.
Article En | MEDLINE | ID: mdl-34018291

The major capsid protein VP1 of JC Polyomavirus assembles into pentamers that serve as a model for studying viral entry of this potentially severe human pathogen. Previously, labeling of viral proteins utilized large fusion proteins or non-specific amine- or cysteine-functionalization with fluorescent dyes. Imaging of these sterically hindered fusion proteins or heterogeneously labeled virions limits reproducibility and could prevent the detection of subtle trafficking phenomena. Here we advance the π-clamp-mediated cysteine conjugation for site-selective fluorescent labeling of VP1-pentamers. We demonstrate a one-step synthesis of a probe consisting of a bio-orthogonal click chemistry handle bridged to a perfluoro-biphenyl π-clamp reactive electrophile by a polyethylene glycol linker. We expand the scope of the π-clamp conjugation by demonstrating selective labeling of an internal, surface exposed loop in VP1. Thus, the π-clamp conjugation offers a general method to selectively bioconjugate tags-of-interest to viral proteins without impeding their ability to bind and enter cells.


Capsid Proteins/metabolism , Cysteine/metabolism , JC Virus/metabolism , Small Molecule Libraries/metabolism , Capsid Proteins/chemistry , Cysteine/chemistry , JC Virus/chemistry , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry
2.
Microbiol Immunol ; 64(12): 783-791, 2020 Dec.
Article En | MEDLINE | ID: mdl-32965709

JC polyomavirus (JCPyV) is a common human pathogen that results in a chronic asymptomatic infection in healthy adults. Under conditions of immunosuppression, JCPyV spreads to the central nervous system and can cause the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML), a disease for which there are no vaccines or antiviral therapies. Retro-2 is a previously identified small molecule inhibitor that was originally shown to block retrograde transport of toxins such as ricin toxin from endosomes to the Golgi apparatus and endoplasmic reticulum (ER), and Retro-2.1 is a chemical analog of Retro-2 that has been shown to inhibit ricin intoxication of cells at low nanomolar concentrations. Retro-2 has previously been shown to prevent retrograde transport of JCPyV virions to the ER, but the effect of Retro-2.1 on JCPyV infectivity is unknown. Here it is shown that Retro-2.1 inhibits JCPyV with an EC50 of 3.9 µM. This molecule inhibits JCPyV infection at dosages that are not toxic to human tissue culture cells. Retro-2.1 was also tested against two other polyomaviruses, the human BK polyomavirus and simian virus 40, and was also shown to inhibit infection at similar concentrations. Viral uncoating studies demonstrate that Retro-2.1 inhibits BKPyV infectivity in a manner similar to Retro-2. These studies demonstrate that improved analogs of Retro-2 can inhibit infection at lower dosages than Retro-2 and further optimization of these compounds may lead to effective treatment options for those suffering from JCPyV infection and PML.


Benzamides/pharmacology , JC Virus/drug effects , Polyomavirus Infections/drug therapy , Thiophenes/pharmacology , Animals , BK Virus/drug effects , Chlorocebus aethiops , Humans , Leukoencephalopathy, Progressive Multifocal/virology , Polyomavirus/drug effects , Simian virus 40/drug effects , Vero Cells
3.
Curr Clin Microbiol Rep ; 4(3): 132-141, 2017 Sep.
Article En | MEDLINE | ID: mdl-28989857

PURPOSE OF REVIEW: JC polyomavirus (JCPyV) is a significant human pathogen that causes an asymptomatic infection in the kidney in the majority of the population. In immunosuppressed individuals, the virus can become reactivated and spread to the brain, causing the fatal, demyelinating disease progressive multifocal leukoencephalopathy (PML). There are currently limited treatment options for this fatal disease. Attachment to receptors and entry into host cells are the initiating events in JCPyV infection and therefore an attractive target for therapeutics to prevent or treat PML. This review provides the current understanding of JCPyV attachment and entry events and the potential therapeutics to target these areas. RECENT FINDINGS: JCPyV attachment and entry to host cells is mediated by α2,6-linked lactoseries tetrasaccharide c (LSTc) and 5-hydroxytryptamine receptors (5-HT2Rs), respectively, and subsequent trafficking to the endoplasmic reticulum is required for infection. Recently, vaccines, monoclonal antibodies, and small molecules have shown promise as anti-viral and PML therapies. SUMMARY: This review summarizes our current understanding of JCPyV attachment, entry, and trafficking and the development of potential PML therapeutics that inhibit these critical steps in JCPyV infection.

4.
Am J Pathol ; 185(8): 2246-58, 2015 Aug.
Article En | MEDLINE | ID: mdl-26056932

The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy, a rare demyelinating disease that occurs in the setting of prolonged immunosuppression. After initial asymptomatic infection, the virus establishes lifelong persistence in the kidney and possibly other extraneural sites. In rare instances, the virus traffics to the central nervous system, where oligodendrocytes, astrocytes, and glial precursors are susceptible to lytic infection, resulting in progressive multifocal leukoencephalopathy. The mechanisms by which the virus traffics to the central nervous system from peripheral sites remain unknown. Lactoseries tetrasaccharide c (LSTc), a pentasaccharide containing a terminal α2,6-linked sialic acid, is the major attachment receptor for polyomavirus. In addition to LSTc, type 2 serotonin receptors are required for facilitating virus entry into susceptible cells. We studied the distribution of virus receptors in kidney and brain using lectins, antibodies, and labeled virus. The distribution of LSTc, serotonin receptors, and virus binding sites overlapped in kidney and in the choroid plexus. In brain parenchyma, serotonin receptors were expressed on oligodendrocytes and astrocytes, but these cells were negative for LSTc and did not bind virus. LSTc was instead found on microglia and vascular endothelium, to which virus bound abundantly. Receptor distribution was not changed in the brains of patients with progressive multifocal leukoencephalopathy. Virus infection of oligodendrocytes and astrocytes during disease progression is LSTc independent.


Brain/metabolism , Choroid Plexus/metabolism , JC Virus , Kidney/metabolism , Polysaccharides/metabolism , Receptors, Serotonin, 5-HT2/metabolism , Receptors, Virus/metabolism , Sialic Acids/metabolism , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Female , Humans , Male , Middle Aged , Oligodendroglia/metabolism
5.
J Virol ; 89(12): 6364-75, 2015 Jun.
Article En | MEDLINE | ID: mdl-25855729

UNLABELLED: The human JC polyomavirus (JCPyV) establishes an asymptomatic, persistent infection in the kidneys of the majority of the population and is the causative agent of the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in immunosuppressed individuals. The Mad-1 strain of JCPyV, a brain isolate, was shown earlier to require α2,6-linked sialic acid on the lactoseries tetrasaccharide c (LSTc) glycan for attachment to host cells. In contrast, a JCPyV kidney isolate type 3 strain, WT3, has been reported to interact with sialic acid-containing gangliosides, but the role of these glycans in JCPyV infection has remained unclear. To help rationalize these findings and probe the effects of strain-specific differences on receptor binding, we performed a comprehensive analysis of the glycan receptor specificities of these two representative JCPyV strains using high-resolution X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, and correlated these data with the results of infectivity assays. We show here that capsid proteins of Mad-1 and WT3 JCPyV can both engage LSTc as well as multiple sialylated gangliosides. However, the binding affinities exhibit subtle differences, with the highest affinity observed for LSTc. Engagement of LSTc is a prerequisite for functional receptor engagement, while the more weakly binding gangliosides are not required for productive infection. Our findings highlight the complexity of virus-carbohydrate interactions and demonstrate that subtle differences in binding affinities, rather than the binding event alone, help determine tissue tropism and viral pathogenesis. IMPORTANCE: Viral infection is initiated by attachment to receptors on host cells, and this event plays an important role in viral disease. We investigated the receptor-binding properties of human JC polyomavirus (JCPyV), a virus that resides in the kidneys of the majority of the population and can cause the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in the brains of immunosuppressed individuals. JCPyV has been reported to interact with multiple carbohydrate receptors, and we sought to clarify how the interactions between JCPyV and cellular carbohydrate receptors influenced infection. Here we demonstrate that JCPyV can engage numerous sialylated carbohydrate receptors. However, the virus displays preferential binding to LSTc, and only LSTc mediates a productive infection. Our findings demonstrate that subtle differences in binding affinity, rather than receptor engagement alone, are a key determinant of viral infection.


Capsid Proteins/metabolism , JC Virus/physiology , Polysaccharides/metabolism , Receptors, Virus/metabolism , Sialic Acids/metabolism , Virus Attachment , Animals , Capsid Proteins/chemistry , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Mice , Receptors, Virus/chemistry
6.
J Virol ; 89(7): 3910-21, 2015 Apr.
Article En | MEDLINE | ID: mdl-25609820

UNLABELLED: JC polyomavirus (JCPyV) infection of immunocompromised individuals results in the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML). The viral capsid of JCPyV is composed primarily of the major capsid protein virus protein 1 (VP1), and pentameric arrangement of VP1 monomers results in the formation of a pore at the 5-fold axis of symmetry. While the presence of this pore is conserved among polyomaviruses, its functional role in infection or assembly is unknown. Here, we investigate the role of the 5-fold pore in assembly and infection of JCPyV by generating a panel of mutant viruses containing amino acid substitutions of the residues lining this pore. Multicycle growth assays demonstrated that the fitness of all mutants was reduced compared to that of the wild-type virus. Bacterial expression of VP1 pentamers containing substitutions to residues lining the 5-fold pore did not affect pentamer assembly or prevent association with the VP2 minor capsid protein. The X-ray crystal structures of selected pore mutants contained subtle changes to the 5-fold pore, and no other changes to VP1 were observed. Pore mutant pseudoviruses were not deficient in assembly, packaging of the minor capsid proteins, or binding to cells or in transport to the host cell endoplasmic reticulum. Instead, these mutant viruses were unable to expose VP2 upon arrival to the endoplasmic reticulum, a step that is critical for infection. This study demonstrated that the 5-fold pore is an important structural feature of JCPyV and that minor modifications to this structure have significant impacts on infectious entry. IMPORTANCE: JCPyV is an important human pathogen that causes a severe neurological disease in immunocompromised individuals. While the high-resolution X-ray structure of the major capsid protein of JCPyV has been solved, the importance of a major structural feature of the capsid, the 5-fold pore, remains poorly understood. This pore is conserved across polyomaviruses and suggests either that these viruses have limited structural plasticity in this region or that this pore is important in infection or assembly. Using a structure-guided mutational approach, we showed that modulation of this pore severely inhibits JCPyV infection. These mutants do not appear deficient in assembly or early steps in infectious entry and are instead reduced in their ability to expose a minor capsid protein in the host cell endoplasmic reticulum. Our work demonstrates that the 5-fold pore is an important structural feature for JCPyV.


Capsid Proteins/metabolism , Capsid/physiology , JC Virus/physiology , Protein Multimerization , Virus Assembly , Virus Internalization , Amino Acid Substitution , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Crystallography, X-Ray , Humans , JC Virus/chemistry , JC Virus/genetics , JC Virus/growth & development , Macromolecular Substances/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation
7.
J Neurovirol ; 21(6): 601-13, 2015 Dec.
Article En | MEDLINE | ID: mdl-25078361

The human JC polyomavirus (JCPyV) causes a lifelong persistent infection in the reno-urinary tract in the majority of the adult population worldwide. In healthy individuals, infection is asymptomatic, while in immunocompromised individuals, the virus can spread to the central nervous system and cause a fatal demyelinating disease known as progressive multifocal leukoencephalopathy (PML). There are currently very few treatment options for this rapidly progressing and devastating disease. Understanding the basic biology of JCPyV-host cell interactions is critical for the development of therapeutic strategies to prevent or treat PML. Research in our laboratory has focused on gaining a detailed mechanistic understanding of the initial steps in the JCPyV life cycle in order to define how JCPyV selectively targets cells in the kidney and brain. JCPyV requires sialic acids to attach to host cells and initiate infection, and JCPyV demonstrates specificity for the oligosaccharide lactoseries tetrasaccharide c (LSTc) with an α2,6-linked sialic acid. Following viral attachment, JCPyV entry is facilitated by the 5-hydroxytryptamine (5-HT)2 family of serotonin receptors via clathrin-dependent endocytosis. JCPyV then undergoes retrograde transport to the endoplasmic reticulum (ER) where viral disassembly begins. A novel retrograde transport inhibitor termed Retro-2(cycl) prevents trafficking of JCPyV to the ER and inhibits both initial virus infection and infectious spread in cell culture. Understanding the molecular mechanisms by which JCPyV establishes infection will open up new avenues for the prevention or treatment of virus-induced disease.


Host-Parasite Interactions/physiology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Virus Attachment , Virus Internalization , Humans
8.
Bioorg Med Chem ; 22(17): 4836-47, 2014 Sep 01.
Article En | MEDLINE | ID: mdl-25087050

Human polyoma- and papillomaviruses are non-enveloped DNA viruses that cause severe pathologies and mortalities. Under circumstances of immunosuppression, JC polyomavirus causes a fatal demyelinating disease called progressive multifocal leukoencephalopathy (PML) and the BK polyomavirus is the etiological agent of polyomavirus-induced nephropathy and hemorrhagic cystitis. Human papillomavirus type 16, another non-enveloped DNA virus, is associated with the development of cancers in tissues like the uterine cervix and oropharynx. Currently, there are no approved drugs or vaccines to treat or prevent polyomavirus infections. We recently discovered that the small molecule Retro-2(cycl), an inhibitor of host retrograde trafficking, blocked infection by several human and monkey polyomaviruses. Here, we report diversity-oriented syntheses of Retro-2(cycl) and evaluation of the resulting analogs using an assay of human cell infections by JC polyomavirus. We defined structure-activity relationships and also discovered analogs with significantly improved potency as suppressors of human polyoma- and papillomavirus infection in vitro. Our findings represent an advance in the development of drug candidates that can broadly protect humans from non-enveloped DNA viruses and toxins that exploit retrograde trafficking as a means for cell entry.


Axonal Transport/drug effects , JC Virus/drug effects , Neurons/drug effects , Neurons/virology , Papillomaviridae/drug effects , Quinazolines/pharmacology , Virus Internalization/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , JC Virus/physiology , Molecular Structure , Papillomaviridae/physiology , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship
9.
J Virol ; 88(2): 948-60, 2014 Jan.
Article En | MEDLINE | ID: mdl-24198413

Progressive multifocal leukoencephalopathy (PML) is a fatal disease with limited treatment options, both clinically and in the research pipeline. Potential therapies would target and neutralize its etiologic agent, JC polyomavirus (JCPyV). The innate immune response to JCPyV infection has not been studied, and little is known about the initial host response to polyomavirus infection. This study examined the ability of a human alpha defensin, HD5, to neutralize JCPyV infection in human fetal glial cells. We show that HD5, by binding to the virion, blocks infection. The JCPyV-HD5 complexes bind to and enter host cells but are reduced in their ability to reach the endoplasmic reticulum (ER), where virions are normally uncoated. Furthermore, HD5 binding to the virion stabilizes the capsid and prevents genome release. Our results show that HD5 neutralizes JCPyV infection at an early postentry step in the viral life cycle by stabilizing the viral capsid and disrupting JCPyV trafficking. This study provides a naturally occurring platform for developing antivirals to treat PML and also expands on the known capabilities of human defensins.


Capsid/metabolism , Endoplasmic Reticulum/virology , JC Virus/physiology , Polyomavirus Infections/metabolism , Tumor Virus Infections/metabolism , alpha-Defensins/metabolism , Capsid/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Endoplasmic Reticulum/metabolism , Humans , JC Virus/genetics , Polyomavirus Infections/genetics , Polyomavirus Infections/virology , Protein Binding , Protein Transport , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , alpha-Defensins/genetics
10.
mBio ; 4(6): e00729-13, 2013 Nov 12.
Article En | MEDLINE | ID: mdl-24222489

UNLABELLED: Polyomaviruses are ubiquitous pathogens that cause severe disease in immunocompromised individuals. JC polyomavirus (JCPyV) is the causative agent of the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML), whereas BK polyomavirus (BKPyV) causes polyomavirus-induced nephropathy and hemorrhagic cystitis. Vaccines or antiviral therapies targeting these viruses do not exist, and treatments focus on reducing the underlying causes of immunosuppression. We demonstrate that retro-2(cycl), an inhibitor of ricin and Shiga-like toxins (SLTs), inhibits infection by JCPyV, BKPyV, and simian virus 40. Retro-2(cycl) inhibits retrograde transport of polyomaviruses to the endoplasmic reticulum, a step necessary for productive infection. Retro-2(cycl) likely inhibits polyomaviruses in a way similar to its ricin and SLT inhibition, suggesting an overlap in the cellular host factors used by bacterial toxins and polyomaviruses. This work establishes retro-2(cycl) as a potential antiviral therapy that broadly inhibits polyomaviruses and possibly other pathogens that use retrograde trafficking. IMPORTANCE: The human polyomaviruses JC polyomavirus (JCPyV) and BK polyomavirus (BKPyV) cause rare but severe diseases in individuals with reduced immune function. During immunosuppression, JCPyV disseminates from the kidney to the central nervous system and destroys oligodendrocytes, resulting in the fatal disease progressive multifocal leukoencephalopathy. Kidney transplant recipients are at increased risk of BKPyV-induced nephropathy, which results in kidney necrosis and loss of the transplanted organ. There are currently no effective therapies for JCPyV and BKPyV. We show that a small molecule named retro-2(cycl) protects cells from infection with JCPyV and BKPyV by inhibiting intracellular viral transport. Retro-2(cycl) treatment reduces viral spreading in already established infections and may therefore be able to control infection in affected patients. Further optimization of retro-2(cycl) may result in the development of an effective antiviral therapy directed toward pathogens that use retrograde trafficking to infect their hosts.


Antiviral Agents/metabolism , BK Virus/drug effects , Benzamides/metabolism , JC Virus/drug effects , Simian virus 40/drug effects , Thiophenes/metabolism , Virus Internalization/drug effects , Animals , BK Virus/physiology , Cell Line , Haplorhini , Humans , JC Virus/physiology , Protein Transport/drug effects , Ricin/antagonists & inhibitors , Shiga Toxins/antagonists & inhibitors , Simian virus 40/physiology
11.
J Virol ; 87(24): 13490-8, 2013 Dec.
Article En | MEDLINE | ID: mdl-24089568

The human JC polyomavirus (JCPyV) causes the rapidly progressing demyelinating disease progressive multifocal leukoencephalopathy (PML). The disease occurs most often in individuals with AIDS but also occurs in individuals receiving immunomodulatory therapies for immune-related diseases such as multiple sclerosis. JCPyV infection of host cells requires the pentasaccharide lactoseries tetrasaccharide c (LSTc) and the serotonin receptor 5-hydroxytryptamine (5-HT) receptor 5-HT2AR. While LSTc is involved in the initial attachment of virus to cells via interactions with VP1, the mechanism by which 5-HT2AR contributes to infection is not clear. To further define the roles of serotonin receptors in infection, HEK293A cells, which are poorly permissive to JCPyV, were transfected with 14 different isoforms of serotonin receptor. Only 5-HT2 receptors were found to support infection by JCPyV. None of the other 11 isoforms of serotonin receptor supported JCPyV infection. Expression of 5-HT2 receptors did not increase binding of JCPyV to cells, but this was not unexpected, given that the cells uniformly expressed the major attachment receptor, LSTc. Infection of these cells remained sensitive to inhibition with soluble LSTc, confirming that LSTc recognition is required for JCPyV infection. Virus internalization into HEK293A cells was significantly and specifically enhanced when 5HT2 receptors were expressed. Taken together, these data confirm that the carbohydrate LSTc is the attachment receptor for JCPyV and that the type 2 serotonin receptors contribute to JCPyV infection by facilitating entry.


JC Virus/physiology , Leukoencephalopathy, Progressive Multifocal/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Virus Internalization , HEK293 Cells , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2B/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin/metabolism
12.
Proc Natl Acad Sci U S A ; 110(18): 7452-7, 2013 Apr 30.
Article En | MEDLINE | ID: mdl-23569269

Despite major advances in our understanding of many aspects of human papillomavirus (HPV) biology, HPV entry is poorly understood. To identify cellular genes required for HPV entry, we conducted a genome-wide screen for siRNAs that inhibited infection of HeLa cells by HPV16 pseudovirus. Many retrograde transport factors were required for efficient infection, including multiple subunits of the retromer, which initiates retrograde transport from the endosome to the trans-Golgi network (TGN). The retromer has not been previously implicated in virus entry. Furthermore, HPV16 capsid proteins arrive in the TGN/Golgi in a retromer-dependent fashion during entry, and incoming HPV proteins form a stable complex with retromer subunits. We propose that HPV16 directly engages the retromer at the early or late endosome and traffics to the TGN/Golgi via the retrograde pathway during cell entry. These results provide important insights into HPV entry, identify numerous potential antiviral targets, and suggest that the role of the retromer in infection by other viruses should be assessed.


Genome, Human/genetics , Papillomaviridae/physiology , RNA Interference , RNA, Small Interfering/metabolism , Vesicular Transport Proteins/metabolism , Virus Internalization , Golgi Apparatus/virology , HeLa Cells , Human papillomavirus 16/physiology , Humans , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Protein Binding , Protein Transport , Reproducibility of Results , Viral Proteins/metabolism
13.
Clin Microbiol Rev ; 25(3): 471-506, 2012 Jul.
Article En | MEDLINE | ID: mdl-22763635

Progressive multifocal leukoencephalopathy (PML) is a debilitating and frequently fatal central nervous system (CNS) demyelinating disease caused by JC virus (JCV), for which there is currently no effective treatment. Lytic infection of oligodendrocytes in the brain leads to their eventual destruction and progressive demyelination, resulting in multiple foci of lesions in the white matter of the brain. Before the mid-1980s, PML was a relatively rare disease, reported to occur primarily in those with underlying neoplastic conditions affecting immune function and, more rarely, in allograft recipients receiving immunosuppressive drugs. However, with the onset of the AIDS pandemic, the incidence of PML has increased dramatically. Approximately 3 to 5% of HIV-infected individuals will develop PML, which is classified as an AIDS-defining illness. In addition, the recent advent of humanized monoclonal antibody therapy for the treatment of autoimmune inflammatory diseases such as multiple sclerosis (MS) and Crohn's disease has also led to an increased risk of PML as a side effect of immunotherapy. Thus, the study of JCV and the elucidation of the underlying causes of PML are important and active areas of research that may lead to new insights into immune function and host antiviral defense, as well as to potential new therapies.


Brain/virology , Demyelinating Diseases/virology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/epidemiology , Virus Replication/drug effects , Antibodies, Monoclonal, Humanized/adverse effects , Brain/pathology , Coinfection/epidemiology , Coinfection/pathology , Coinfection/virology , Demyelinating Diseases/pathology , Gene Expression Regulation, Viral , Genome, Viral , HIV/pathogenicity , HIV Infections/pathology , HIV Infections/virology , Humans , Immune Reconstitution Inflammatory Syndrome/pathology , JC Virus/genetics , JC Virus/physiology , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Risk Factors
14.
Virology ; 428(1): 30-40, 2012 Jun 20.
Article En | MEDLINE | ID: mdl-22516137

JC polyomavirus (JCV) is an important human pathogen that causes the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML). In this study we further delineate the early events of JCV entry in human glial cells and demonstrate that a pentameric subunit of the viral capsid is able to recapitulate early events in viral trafficking. We show that JCV traffics to the endoplasmic reticulum (ER) by 6h post infection, and that VP1 pentamers arrive at the ER with similar kinetics. Further, this JCV localization to the ER is critical for infection, as treatment of cells with agents that prevent ER trafficking, ER function, or ER quality control reduce JCV infectivity. These pentamers represent a new tool to study polyomavirus entry, and will be particularly useful in studying recently identified polyomaviruses that are difficult to propagate.


Capsid Proteins/metabolism , JC Virus/physiology , Polyomavirus Infections/virology , Virus Internalization , Capsid Proteins/genetics , Endoplasmic Reticulum/virology , Humans , JC Virus/genetics , Neuroglia/virology
15.
Virology ; 411(1): 142-52, 2011 Mar 01.
Article En | MEDLINE | ID: mdl-21239031

Avian polyomavirus (APV) causes a fatal, multi-organ disease among several bird species. Using cryogenic electron microscopy and other biochemical techniques, we investigated the structure of APV and compared it to that of mammalian polyomaviruses, particularly JC polyomavirus and simian virus 40. The structure of the pentameric major capsid protein (VP1) is mostly conserved; however, APV VP1 has a unique, truncated C-terminus that eliminates an intercapsomere-connecting ß-hairpin observed in other polyomaviruses. We postulate that the terminal ß-hairpin locks other polyomavirus capsids in a stable conformation and that absence of the hairpin leads to the observed capsid size variation in APV. Plug-like density features were observed at the base of the VP1 pentamers, consistent with the known location of minor capsid proteins VP2 and VP3. However, the plug density is more prominent in APV and may include VP4, a minor capsid protein unique to bird polyomaviruses.


Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Capsid/ultrastructure , Macromolecular Substances/ultrastructure , Polyomavirus/ultrastructure , Animals , Cryoelectron Microscopy , Melopsittacus/virology , Polyomavirus/isolation & purification
16.
Cell Host Microbe ; 8(4): 309-19, 2010 Oct 21.
Article En | MEDLINE | ID: mdl-20951965

The human JC polyomavirus (JCV) causes a fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML), in immunocompromised individuals. Current treatment options for PML are inadequate. Sialylated oligosaccharides and the serotonin receptor are known to be necessary for JCV entry, but the molecular interactions underlying JCV attachment remain unknown. Using glycan array screening and viral infectivity assays, we identify a linear sialylated pentasaccharide with the sequence NeuNAc-α2,6-Gal-ß1,4-GlcNAc-ß1,3-Gal-ß1,4-Glc (LSTc) present on host glycoproteins and glycolipids as a specific JCV recognition motif. The crystal structure of the JCV capsid protein VP1 was solved alone and in complex with LSTc. It reveals extensive interactions with the terminal sialic acid of the LSTc motif and specific recognition of an extended conformation of LSTc. Mutations in the JCV oligosaccharide-binding sites abolish cell attachment, viral spread, and infectivity, further validating the importance of this interaction. Our findings provide a powerful platform for the development of antiviral compounds.


Amino Acid Motifs , JC Virus/metabolism , Oligosaccharides/chemistry , Protein Array Analysis , Receptors, Virus/chemistry , Capsid Proteins/chemistry , Cell Line , Glycoproteins/chemistry , Humans , JC Virus/genetics , Molecular Conformation , N-Acetylneuraminic Acid/metabolism , Protein Structure, Tertiary , Sialoglycoproteins/chemistry , Structure-Activity Relationship , Virus Attachment
17.
J Virol ; 83(11): 5556-66, 2009 Jun.
Article En | MEDLINE | ID: mdl-19321620

The structures of canine parvovirus (CPV) and feline parvovirus (FPV) complexed with antibody fragments from eight different neutralizing monoclonal antibodies were determined by cryo-electron microscopy (cryoEM) reconstruction to resolutions varying from 8.5 to 18 A. The crystal structure of one of the Fab molecules and the sequence of the variable domain for each of the Fab molecules have been determined. The structures of Fab fragments not determined crystallographically were predicted by homology modeling according to the amino acid sequence. Fitting of the Fab and virus structures into the cryoEM densities identified the footprints of each antibody on the viral surface. As anticipated from earlier analyses, the Fab binding sites are directed to two epitopes, A and B. The A site is on an exposed part of the surface near an icosahedral threefold axis, whereas the B site is about equidistant from the surrounding five-, three-, and twofold axes. One antibody directed to the A site binds CPV but not FPV. Two of the antibodies directed to the B site neutralize the virus as Fab fragments. The differences in antibody properties have been linked to the amino acids within the antibody footprints, the position of the binding site relative to the icosahedral symmetry elements, and the orientation of the Fab structure relative to the surface of the virus. Most of the exposed surface area was antigenic, although each of the antibodies had a common area of overlap that coincided with the positions of the previously mapped escape mutations.


Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Capsid/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Parvovirus/chemistry , Parvovirus/immunology , Amino Acid Sequence , Antibodies, Monoclonal/ultrastructure , Antibody Specificity , Antigens/chemistry , Antigens/immunology , Capsid/chemistry , Capsid/ultrastructure , Computational Biology , Conserved Sequence , Cryoelectron Microscopy , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Parvovirus/ultrastructure , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein
18.
J Virol ; 82(21): 10397-407, 2008 Nov.
Article En | MEDLINE | ID: mdl-18701590

Parvovirus capsids are assembled from multiple forms of a single protein and are quite stable structurally. However, in order to infect cells, conformational plasticity of the capsid is required and this likely involves the exposure of structures that are buried within the structural models. The presence of functional asymmetry in the otherwise icosahedral capsid has also been proposed. Here we examined the protein composition of canine parvovirus capsids and evaluated their structural variation and permeability by protease sensitivity, spectrofluorometry, and negative staining electron microscopy. Additional protein forms identified included an apparent smaller variant of the virus protein 1 (VP1) and a small proportion of a cleaved form of VP2. Only a small percentage of the proteins in intact capsids were cleaved by any of the proteases tested. The capsid susceptibility to proteolysis varied with temperature but new cleavages were not revealed. No global change in the capsid structure was observed by analysis of Trp fluorescence when capsids were heated between 40 degrees C and 60 degrees C. However, increased polarity of empty capsids was indicated by bis-ANS binding, something not seen for DNA-containing capsids. Removal of calcium with EGTA or exposure to pHs as low as 5.0 had little effect on the structure, but at pH 4.0 changes were revealed by proteinase K digestion. Exposure of viral DNA to the external environment started above 50 degrees C. Some negative stains showed increased permeability of empty capsids at higher temperatures, but no effects were seen after EGTA treatment.


Capsid Proteins/analysis , Capsid/chemistry , Parvovirus, Canine/chemistry , Peptides/analysis , Capsid/ultrastructure , Hydrogen-Ion Concentration , Microscopy, Electron, Transmission , Negative Staining , Parvovirus, Canine/ultrastructure , Peptide Hydrolases/metabolism , Spectrometry, Fluorescence , Temperature
19.
J Virol ; 81(18): 9851-8, 2007 Sep.
Article En | MEDLINE | ID: mdl-17626084

The structure of virus-like particles of the lymphotropic, immunosuppressive strain of minute virus of mice (MVMi) in complex with the neutralizing Fab fragment of the mouse monoclonal antibody (MAb) B7 was determined by cryo-electron microscopy to 7-A resolution. The Fab molecule recognizes a conformational epitope at the vertex of a three-fold protrusion on the viral surface, thereby simultaneously engaging three symmetry-related viral proteins in binding. The location of the epitope close to the three-fold axis is consistent with the previous analysis of MVMi mutants able to escape from the B7 antibody. The binding site close to the symmetry axes sterically forbids the binding of more than one Fab molecule per spike. MAb as well as the Fab molecules inhibits the binding of the minute virus of mice (MVM) to permissive cells but can also neutralize MVM postattachment. This finding suggests that the interaction of B7 with three symmetry-related viral subunits at each spike hinders structural transitions in the viral capsid essential during viral entry.


Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , Antigen-Antibody Complex/ultrastructure , Capsid/ultrastructure , Immunoglobulin Fab Fragments/chemistry , Minute Virus of Mice/ultrastructure , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Binding Sites, Antibody/immunology , Capsid/chemistry , Capsid/immunology , Cryoelectron Microscopy , Epitopes/chemistry , Epitopes/immunology , Immunoglobulin Fab Fragments/immunology , Mice , Minute Virus of Mice/chemistry , Minute Virus of Mice/immunology , Molecular Sequence Data , Viral Proteins/chemistry , Viral Proteins/immunology , Virus Attachment , Virus Internalization
20.
Proc Natl Acad Sci U S A ; 104(16): 6585-9, 2007 Apr 17.
Article En | MEDLINE | ID: mdl-17420467

Although many viruses are icosahedral when they initially bind to one or more receptor molecules on the cell surface, such an interaction is asymmetric, probably causing a breakdown in the symmetry and conformation of the original infecting virion in preparation for membrane penetration and release of the viral genome. Cryoelectron microscopy and biochemical analyses show that transferrin receptor, the cellular receptor for canine parvovirus, can bind to only one or a few of the 60 icosahedrally equivalent sites on the virion, indicating that either canine parvovirus has inherent asymmetry or binding of receptor induces asymmetry. The asymmetry of receptor binding to canine parvovirus is reminiscent of the special portal in tailed bacteriophages and some large, icosahedral viruses. Asymmetric interactions of icosahedral viruses with their hosts might be a more common phenomenon than previously thought and may have been obscured by averaging in previous crystallographic and electron microscopic structure determinations.


Capsid/chemistry , Capsid/metabolism , Parvovirus, Canine/chemistry , Parvovirus, Canine/metabolism , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Animals , Binding Sites , Cats , Cryoelectron Microscopy , Crystallography, X-Ray , Dogs , Feline Panleukopenia Virus/chemistry , Feline Panleukopenia Virus/metabolism , Feline Panleukopenia Virus/ultrastructure , Humans , Parvovirus, Canine/ultrastructure , Protein Binding , Receptors, Transferrin/genetics , Spodoptera , Virion/chemistry , Virion/metabolism
...