Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38562771

ABSTRACT

Legionella pneumophila grows within membrane-bound vacuoles in phylogenetically diverse hosts. Intracellular growth requires the function of the Icm/Dot type-IVb secretion system, which translocates more than 300 proteins into host cells. A screen was performed to identify L. pneumophila proteins that stimulate MAPK activation, using Icm/Dot translocated proteins ectopically expressed in mammalian cells. In parallel, a second screen was performed to identify L. pneumophila proteins expressed in yeast that cause growth inhibition in MAPK pathway-stimulatory high osmolarity medium. LegA7 was shared in both screens, a protein predicted to be a member of the bacterial cysteine protease family that has five carboxyl-terminal ankyrin repeats. Three conserved residues in the predicted catalytic triad of LegA7 were mutated. These mutations abolished the ability of LegA7 to inhibit yeast growth. To identify other residues important for LegA7 function, a generalizable selection strategy in yeast was devised to isolate mutants that have lost function and no longer cause growth inhibition on high osmolarity medium. Mutations were isolated in the two amino-terminal ankyrin repeats, as well as an inter-domain region located between the cysteine protease domain and the ankyrin repeats. These mutations were predicted by AlphaFold modeling to localize to the face opposite from the catalytic site, arguing that they interfere with the positive regulation of the catalytic activity. Based on our data, we present a model in which LegA7 harbors a cysteine protease domain with an inter-domain and two amino-terminal ankyrin repeat regions that modulate the function of the catalytic domain.

2.
J Extracell Biol ; 1(5)2022 May.
Article in English | MEDLINE | ID: mdl-36688929

ABSTRACT

JC polyomavirus (JCPyV) is a small, non-enveloped virus that persists in the kidney in about half the adult population. In severely immune-compromised individuals JCPyV causes the neurodegenerative disease progressive multifocal leukoencephalopathy (PML) in the brain. JCPyV has been shown to infect cells by both direct and indirect mechanisms, the latter involving extracellular vesicle (EV) mediated infection. While direct mechanisms of infection are well studied indirect EV mediated mechanisms are poorly understood. Using a combination of chemical and genetic approaches we show that several overlapping intracellular pathways are responsible for the biogenesis of virus containing EV. Here we show that targeting neutral sphingomyelinase 2 (nSMase2) with the drug cambinol decreased the spread of JCPyV over several viral life cycles. Genetic depletion of nSMase2 by either shRNA or CRISPR/Cas9 reduced EV-mediated infection. Individual knockdown of seven ESCRT-related proteins including HGS, ALIX, TSG101, VPS25, VPS20, CHMP4A, and VPS4A did not significantly reduce JCPyV associated EV (JCPyV(+) EV) infectivity, whereas knockdown of the tetraspanins CD9 and CD81 or trafficking and/or secretory autophagy-related proteins RAB8A, RAB27A, and GRASP65 all significantly reduced the spread of JCPyV and decreased EV-mediated infection. These findings point to a role for exosomes and secretory autophagosomes in the biogenesis of JCPyV associated EVs with specific roles for nSMase2, CD9, CD81, RAB8A, RAB27A, and GRASP65 proteins.

3.
mBio ; 10(2)2019 04 09.
Article in English | MEDLINE | ID: mdl-30967463

ABSTRACT

The endemic human JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy in immune-suppressed patients. The mechanisms of virus infection in vivo are not understood because the major target cells for virus in the brain do not express virus receptors and do not bind virus. We found that JCPyV associates with extracellular vesicles (EVs) and can infect target cells independently of virus receptors. Virus particles were found packaged inside extracellular vesicles and attached to the outer side of vesicles. Anti-JCPyV antisera reduced infection by purified virus but had no effect on infection by EV-associated virus. Treatment of cells with the receptor-destroying enzyme neuraminidase inhibited infection with purified virus but did not inhibit infection by EV-associated virus. Mutant pseudoviruses defective in sialic acid receptor binding could not transduce cells as purified pseudovirions but could do so when associated with EVs. This alternative mechanism of infection likely plays a critical role in the dissemination and spread of JCPyV both to and within the central nervous system.IMPORTANCE JC polyomavirus (JCPyV) is a ubiquitous human pathogen that causes progressive multifocal leukoencephalopathy (PML), a severe and often fatal neurodegenerative disease in immunocompromised or immunomodulated patients. The mechanisms responsible for initiating infection in susceptible cells are not completely known. The major attachment receptor for the virus, lactoseries tetrasaccharide c (LSTc), is paradoxically not expressed on oligodendrocytes or astrocytes in human brain, and virus does not bind to these cells. Because these are the major cell types targeted by the virus in the brain, we hypothesized that alternative mechanisms of infection must be responsible. Here we provide evidence that JCPyV is packaged in extracellular vesicles from infected cells. Infection of target cells by vesicle-associated virus is not dependent on LSTc and is not neutralized by antisera directed against the virus. This is the first demonstration of a polyomavirus using extracellular vesicles as a means of transmission.


Subject(s)
Extracellular Vesicles/virology , JC Virus/physiology , Virus Internalization , Cell Line , Humans
4.
PLoS Pathog ; 11(8): e1005112, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26302170

ABSTRACT

Trichodysplasia spinulosa-associated Polyomavirus (TSPyV) was isolated from a patient suffering from trichodysplasia spinulosa, a skin disease that can appear in severely immunocompromised patients. While TSPyV is one of the five members of the polyomavirus family that are directly linked to a human disease, details about molecular recognition events, the viral entry pathway, and intracellular trafficking events during TSPyV infection remain unknown. Here we have used a structure-function approach to shed light on the first steps of TSPyV infection. We established by cell binding and pseudovirus infection studies that TSPyV interacts with sialic acids during attachment and/or entry. Subsequently, we solved high-resolution X-ray structures of the major capsid protein VP1 of TSPyV in complex with three different glycans, the branched GM1 glycan, and the linear trisaccharides α2,3- and α2,6-sialyllactose. The terminal sialic acid of all three glycans is engaged in a unique binding site on TSPyV VP1, which is positioned about 18 Å from established sialic acid binding sites of other polyomaviruses. Structure-based mutagenesis of sialic acid-binding residues leads to reduction in cell attachment and pseudovirus infection, demonstrating the physiological relevance of the TSPyV VP1-glycan interaction. Furthermore, treatments of cells with inhibitors of N-, O-linked glycosylation, and glycosphingolipid synthesis suggest that glycolipids play an important role during TSPyV infection. Our findings elucidate the first molecular recognition events of cellular infection with TSPyV and demonstrate that receptor recognition by polyomaviruses is highly variable not only in interactions with sialic acid itself, but also in the location of the binding site.


Subject(s)
Capsid Proteins/metabolism , Polyomavirus Infections/metabolism , Polyomavirus/pathogenicity , Virus Internalization , Animals , Binding Sites , Capsid Proteins/chemistry , Cell Line , Flow Cytometry , Glycolipids/chemistry , Glycolipids/metabolism , Humans , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Polyomavirus/chemistry , Polyomavirus/metabolism , Protein Conformation , Sialic Acids/chemistry , Sialic Acids/metabolism , X-Ray Diffraction
5.
PLoS Pathog ; 10(7): e1004229, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25058342

ABSTRACT

Many pathogens, particularly those that require their host for survival, have devised mechanisms to subvert the host immune response in order to survive and replicate intracellularly. Legionella pneumophila, the causative agent of Legionnaires' disease, promotes intracellular growth by translocating proteins into its host cytosol through its type IV protein secretion machinery. At least 5 of the bacterial translocated effectors interfere with the function of host cell elongation factors, blocking translation and causing the induction of a unique host cell transcriptional profile. In addition, L. pneumophila also interferes with translation initiation, by preventing cap-dependent translation in host cells. We demonstrate here that protein translation inhibition by L. pneumophila leads to a frustrated host MAP kinase response, where genes involved in the pathway are transcribed but fail to be translated due to the bacterium-induced protein synthesis inhibition. Surprisingly, few pro-inflammatory cytokines, such as IL-1α and IL-1ß, bypass this inhibition and get synthesized in the presence of Legionella effectors. We show that the selective synthesis of these genes requires MyD88 signaling and takes place in both infected cells that harbor bacteria and neighboring bystander cells. Our findings offer a perspective of how host cells are able to cope with pathogen-encoded activities that disrupt normal cellular process and initiate a successful inflammatory response.


Subject(s)
Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Myeloid Differentiation Factor 88/immunology , Protein Biosynthesis/immunology , Animals , Humans , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Legionnaires' Disease/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Protein Biosynthesis/genetics , U937 Cells
6.
Virology ; 456-457: 87-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24889228

ABSTRACT

Autophagy is important for a variety for virus life cycles. We sought to determine the role of autophagy in human BK polyomavirus (BKPyV) infection. The addition excess amino acids during viral infection reduced BKPyV infection. Perturbing autophagy levels using inhibitors, 3-MA, bafilomycin A1, and spautin-1, also reduced infection, while rapamycin treatment of host cells increased infection. siRNA knockdown of autophagy genes, ATG7 and Beclin-1, corresponded to a decrease in BKPyV infection. BKPyV infection not only correlated with autophagosome formation, but also virus particles localized to autophagy-specific compartments early in infection. These data support a novel role for autophagy in the promotion of BKPyV infection.


Subject(s)
Autophagy , BK Virus/physiology , Host-Pathogen Interactions , Virus Replication , HeLa Cells , Humans
7.
J Immunol ; 190(12): 6329-39, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23686480

ABSTRACT

Legionella pneumophila is an intracellular bacterial pathogen that is the cause of a severe pneumonia in humans called Legionnaires' disease. A key feature of L. pneumophila pathogenesis is the rapid influx of neutrophils into the lungs, which occurs in response to signaling via the IL-1R. Two distinct cytokines, IL-1α and IL-1ß, can stimulate the type I IL-1R. IL-1ß is produced upon activation of cytosolic sensors called inflammasomes that detect L. pneumophila in vitro and in vivo. Surprisingly, we find no essential role for IL-1ß in neutrophil recruitment to the lungs in response to L. pneumophila. Instead, we show that IL-1α is a critical initiator of neutrophil recruitment to the lungs of L. pneumophila-infected mice. We find that neutrophil recruitment in response to virulent L. pneumophila requires the production of IL-1α specifically by hematopoietic cells. In contrast to IL-1ß, the innate signaling pathways that lead to the production of IL-1α in response to L. pneumophila remain poorly defined. In particular, although we confirm a role for inflammasomes for initiation of IL-1ß signaling in vivo, we find no essential role for inflammasomes in production of IL-1α. Instead, we propose that a novel host pathway, perhaps involving inhibition of host protein synthesis, is responsible for IL-1α production in response to virulent L. pneumophila. Our results establish IL-1α as a critical initiator of the inflammatory response to L. pneumophila in vivo and point to an important role for IL-1α in providing an alternative to inflammasome-mediated immune responses in vivo.


Subject(s)
Inflammation/immunology , Interleukin-1alpha/immunology , Legionnaires' Disease/immunology , Neutrophil Infiltration/immunology , Signal Transduction/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Legionella pneumophila , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
8.
J Virol ; 84(4): 1722-30, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19955309

ABSTRACT

The human polyomavirus BK virus (BKV) is a common virus for which 80 to 90% of the adult population is seropositive. BKV reactivation in immunosuppressed patients or renal transplant patients is the primary cause of polyomavirus-associated nephropathy (PVN). Using the Dunlop strain of BKV, we found that nuclear factor of activated T cells (NFAT) plays an important regulatory role in BKV infection. Luciferase reporter assays and chromatin immunoprecipitation assays demonstrated that NFAT4 bound to the viral promoter and regulated viral transcription and infection. The mutational analysis of the NFAT binding sites demonstrated complex functional interactions between NFAT, c-fos, c-jun, and the p65 subunit of NF-kappaB that together influence promoter activity and viral growth. These data indicate that NFAT is required for BKV infection and is involved in a complex regulatory network that both positively and negatively influences promoter activity and viral infection.


Subject(s)
BK Virus/genetics , BK Virus/immunology , NFATC Transcription Factors/metabolism , Animals , BK Virus/pathogenicity , Base Sequence , Binding Sites/genetics , Chlorocebus aethiops , DNA Primers/genetics , Genes, Viral , Humans , Mutation , Polyomavirus Infections/etiology , Promoter Regions, Genetic , Transcription, Genetic , Tumor Virus Infections/etiology , Vero Cells
9.
Cell Host Microbe ; 6(3): 253-67, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19748467

ABSTRACT

The amoeba Dictyostelium discoideum can support replication of Legionella pneumophila. Here we identify the dupA gene, encoding a putative tyrosine kinase/dual-specificity phosphatase, in a screen for D. discoideum mutants altered in allowing L. pneumophila intracellular replication. Inactivation of dupA resulted in depressed L. pneumophila growth and sustained hyperphosphorylation of the amoebal MAP kinase ERK1, consistent with loss of a phosphatase activity. Bacterial challenge of wild-type amoebae induced dupA expression and resulted in transiently increased ERK1 phosphorylation, suggesting that dupA and ERK1 are part of a response to bacteria. Indeed, over 500 of the genes misregulated in the dupA(-) mutant were regulated in response to L. pneumophila infection, including some thought to have immune-like functions. MAP kinase phosphatases are known to be highly upregulated in macrophages challenged with L. pneumophila. Thus, DupA may regulate a MAP kinase response to bacteria that is conserved from amoebae to mammals.


Subject(s)
Dictyostelium/enzymology , Dictyostelium/microbiology , Legionella pneumophila/growth & development , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , Animals , Dictyostelium/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Mitogen-Activated Protein Kinase 3/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics
10.
Virology ; 390(2): 279-88, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19523662

ABSTRACT

JC virus (JCV) is a human polyomavirus that infects the majority of the human population worldwide. It is responsible for the fatal demyelinating disease Progressive Multifocal Leukoencephalopathy. JCV binds to cells using the serotonin receptor 5-HT(2A)R and alpha(2-6)- or alpha(2-3)-linked sialic acid. It enters cells using clathrin-dependent endocytosis and traffics to the early endosome and possibly to the endoplasmic reticulum. Viral DNA is then delivered to the nucleus where transcription, replication, and assembly of progeny occur. We found that the early regulatory protein large T antigen accumulates in microdomains in the nucleus adjacent to ND-10 or PML domains. This observation prompted us to explore the role of these domains in JCV infection. We found that a reduction of nuclear PML enhanced virus infection and that an increase in nuclear PML reduced infection. Infection with JCV did not directly modulate nuclear levels of PML but our data indicate that a host response involving interferon beta is likely to restrict virus infection by increasing nuclear PML.


Subject(s)
Cell Nucleus/virology , Chromosomes/virology , JC Virus/growth & development , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line , Humans , Promyelocytic Leukemia Protein
11.
J Biol Chem ; 283(45): 31125-32, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18782756

ABSTRACT

BK virus (BKV) is a polyomavirus that establishes a lifelong persistence in most humans and is a major impediment to success of kidney grafts. The function of the innate immune system in BKV infection and pathology has not been investigated. Here we examine the role of antimicrobial defensins in BKV infection of Vero cells. Our data show that alpha-defensin human neutrophil protein 1 (HNP1) and human alpha-defensin 5 (HD5) inhibit BKV infection by targeting an early event in the viral lifecycle. HD5 treatment of BKV reduced viral attachment to cells, whereas cellular treatment with HD5 did not. Colocalization studies indicated that HD5 interacts directly with BKV. Ultrastructural analysis revealed HD5-induced aggregation of virions. HD5 also inhibited infection of cells by other related polyomaviruses. This is the first study to demonstrate polyomavirus sensitivity to defensins. We also show a novel mechanism whereby HD5 binds to BKV leading to aggregation of virion particles preventing normal virus binding to the cell surface and uptake into cells.


Subject(s)
Anti-Infective Agents/pharmacology , BK Virus/metabolism , Polyomavirus Infections/metabolism , Virus Internalization/drug effects , alpha-Defensins/pharmacology , Animals , Anti-Infective Agents/metabolism , BK Virus/ultrastructure , Chlorocebus aethiops , Humans , Polyomavirus Infections/pathology , Vero Cells , Virion/metabolism , Virion/ultrastructure , alpha-Defensins/metabolism
12.
J Virol ; 82(5): 2560-4, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094176

ABSTRACT

For the human polyomaviruses JC virus (JCV) and BK virus (BKV), the first step to a successful infection involves binding to sialic acid moieties located on the surfaces of host cells. By stripping and then reconstituting specific sialic acid linkages on host cells, we show that JCV uses both alpha(2,3)-linked and alpha(2,6)-linked sialic acids on N-linked glycoproteins to infect cells. For both JCV and BKV, the sialic acid linkages required for cell surface binding directly correlate with the linkages required for infection. In addition to sialic acid linkage data, these data suggest that the third sugar from the carbohydrate chain terminus is important for virus binding and infection.


Subject(s)
BK Virus/physiology , JC Virus/physiology , N-Acetylneuraminic Acid/metabolism , Cells, Cultured
13.
J Virol ; 81(21): 11798-808, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17699578

ABSTRACT

BK virus (BKV) is a ubiquitous pathogen that establishes a persistent infection in the urinary tract of 80% of the human population. Like other polyomaviruses, the major capsid protein of BKV, virion protein 1 (VP1), is critical for host cell receptor recognition and for proper virion assembly. BKV uses a carbohydrate complex containing alpha(2,3)-linked sialic acid attached to glycoprotein and glycolipid motifs as a cellular receptor. To determine the amino acids important for BKV binding to the sialic acid portion of the complex, we generated a series of 17 point mutations in VP1 and scored them for viral growth. The first set of mutants behaved identically to wild-type virus, suggesting that these amino acids were not critical for virus propagation. Another group of VP1 mutants rendered the virus nonviable. These mutations failed to protect viral DNA from DNase I digestion, indicating a role for these domains in capsid assembly and/or packaging of DNA. A third group of VP1 mutations packaged DNA similarly to the wild type but failed to propagate. The initial burst size of these mutations was similar to that of the wild type, indicating that there is no defect in the lytic release of the mutated virions. Binding experiments revealed that a subset of the BKV mutants were unable to attach to their host cells. These motifs are likely important for sialic acid recognition. We next mapped these mutations onto a model of BKV VP1 to provide atomic insight into the role of these sites in the binding of sialic acid to VP1.


Subject(s)
BK Virus/metabolism , Capsid Proteins/physiology , Amino Acid Sequence , Animals , Capsid/chemistry , Capsid Proteins/chemistry , Cell Survival , Chlorocebus aethiops , Glycoproteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid/chemistry , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Vero Cells
14.
Glycoconj J ; 23(1-2): 19-26, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16575519

ABSTRACT

JC virus (JCV) and BK virus (BKV) are human polyomaviruses that infect approximately 85% of the population worldwide [1,2]. JCV is the underlying cause of the fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML), a condition resulting from JCV induced lytic destruction of myelin producing oligodendrocytes in the brain [3]. BKV infection of kidneys in renal transplant recipients results in a gradual loss of graft function known as polyomavirus associated nephropathy (PVN) [4]. Following the identification of these viruses as the etiological agents of disease, there has been greater interest in understanding the basic biology of these human pathogens [5,6]. Recent advances in the field have shown that viral entry of both JCV and BKV is dependent on the ability to interact with sialic acid. This review focuses on what is known about the human polyomaviruses and the role that sialic acid plays in determining viral tropism.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Polyomavirus Infections/virology , Polyomavirus/physiology , Polyomavirus/pathogenicity , Humans , Receptors, Virus/chemistry , Receptors, Virus/metabolism
15.
Cancer Res ; 65(23): 10970-6, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322245

ABSTRACT

The alpha6beta4 integrin has been widely implicated in carcinoma function in vitro; however, in vivo data are scarce. To determine the importance of alpha6beta4 in tumor progression, a SUM-159 breast carcinoma cell line that is essentially devoid of alpha6beta4 expression was generated using an RNA interference strategy. Loss of alpha6beta4 expression inhibits colony formation in soft agar assays, suggesting a vital role for alpha6beta4 in survival signaling and anchorage-independent growth. Orthotopic injection of the beta4-deficient cell line into the mammary fat pad of immunocompromised mice yielded significantly fewer and smaller tumors than the control cell line, revealing a role for the alpha6beta4 integrin in tumor formation. Under conditions that mimicked the in vivo environment, decreased expression of the alpha6beta4 integrin led to enhanced apoptosis as determined by the percentage of Annexin V-FITC+, PI- cells and the presence of caspase-3 cleavage products. Recombinant vascular endothelial growth factor (VEGF) significantly inhibited the cell death observed in the beta4-deficient cell line, demonstrating the importance of VEGF expression in this survival pathway. Furthermore, loss of alpha6beta4 expression leads to enhanced apoptosis and reduced expression of VEGF in breast carcinoma cells in vivo. Importantly, the specificity of alpha6beta4 in both the in vitro and in vivo assays showed that reexpression of the beta4 subunit into the beta4-deficient cell line could rescue the functional phenotype. Taken together, these data implicate the alpha6beta4 integrin in tumor formation by regulating tumor cell survival in a VEGF-dependent manner.


Subject(s)
Breast Neoplasms/pathology , Integrin alpha6beta4/physiology , Apoptosis/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/physiology , Humans , Integrin alpha6beta4/biosynthesis , Integrin alpha6beta4/deficiency , Integrin alpha6beta4/genetics , Integrin beta4/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/biosynthesis
16.
J Virol ; 79(22): 14442-5, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16254379

ABSTRACT

BK virus (BKV) is a common human polyomavirus infecting >80% of the population worldwide. Infection with BKV is asymptomatic, but reactivation in renal transplant recipients can lead to polyomavirus-associated nephropathy. In this report, we show that enzymatic removal of alpha(2,3)-linked sialic acid from cells inhibited BKV infection. Reconstitution of asialo cells with alpha(2,3)-specific sialyltransferase restored susceptibility to infection. Inhibition of N-linked glycosylation with tunicamycin reduced infection, but inhibition of O-linked glycosylation did not. An O-linked-specific alpha(2,3)-sialyltransferase was unable to restore infection in asialo cells. Taken together, these data indicate that an N-linked glycoprotein containing alpha(2,3)-linked sialic acid is a critical component of the cellular receptor for BKV.


Subject(s)
Glycoproteins/physiology , N-Acetylneuraminic Acid/physiology , Receptors, Virus/physiology , Sialyltransferases/metabolism , Animals , Chlorocebus aethiops , Glycoproteins/chemistry , Neuraminidase , Tunicamycin/pharmacology , Vero Cells
17.
Cancer Res ; 64(23): 8694-701, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15574779

ABSTRACT

Although the RhoA and RhoC proteins comprise an important subset of the Rho GTPase family that have been implicated in invasive breast carcinomas, attributing specific functions to these individual members has been difficult. We have used a stable retroviral RNA interference approach to generate invasive breast carcinoma cells (SUM-159 cells) that lack either RhoA or RhoC expression. Analysis of these cells enabled us to deduce that RhoA impedes and RhoC stimulates invasion. Unexpectedly, this analysis also revealed a compensatory relationship between RhoA and RhoC at the level of both their expression and activation, and a reciprocal relationship between RhoA and Rac1 activation.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , rho GTP-Binding Proteins/physiology , rhoA GTP-Binding Protein/physiology , ADP Ribose Transferases/pharmacology , Breast Neoplasms/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Neoplasm Invasiveness , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , rho GTP-Binding Proteins/biosynthesis , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/genetics , rhoC GTP-Binding Protein
18.
Science ; 306(5700): 1380-3, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15550673

ABSTRACT

The human polyomavirus, JCV, causes the fatal demyelinating disease progressive multifocal leukoencephalopathy in immunocompromised patients. We found that the serotonergic receptor 5HT2AR could act as the cellular receptor for JCV on human glial cells. The 5HT2A receptor antagonists inhibited JCV infection, and monoclonal antibodies directed at 5HT2A receptors blocked infection of glial cells by JCV, but not by SV40. Transfection of 5HT2A receptor-negative HeLa cells with a 5HT2A receptor rescued virus infection, and this infection was blocked by antibody to the 5HT2A receptor. A tagged 5HT2A receptor colocalized with labeled JCV in an endosomal compartment following internalization. Serotonin receptor antagonists may thus be useful in the treatment of progressive multifocal leukoencephalopathy.


Subject(s)
JC Virus/physiology , Neuroglia/virology , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Virus/physiology , Antibodies, Monoclonal , Cell Line, Transformed , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Endosomes/metabolism , Endosomes/virology , Flow Cytometry , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Microscopy, Confocal , Neuroglia/physiology , Receptor, Serotonin, 5-HT2A/immunology , Receptors, Dopamine/immunology , Receptors, Dopamine/physiology , Serotonin/pharmacology , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Sialic Acids/physiology , Transfection
19.
Clin Exp Metastasis ; 20(6): 569-76, 2003.
Article in English | MEDLINE | ID: mdl-14598892

ABSTRACT

The application of small interfering RNA (siRNA) oligonucleotides to silence gene expression has profound implications for the intervention of human diseases including cancer. Using this technique, we explored the possibility that the alpha6beta4 integrin, a laminin adhesion receptor with a recognized role in the invasive phenotype of many carcinomas, represents a potential therapeutic target to inhibit the migration and invasion of carcinoma cells. We found that siRNA oligonucleotides targeted to either subunit of the alpha6beta4 integrin reduced cell surface expression of this integrin and resulted in decreased invasion of MDA-MB-231 breast carcinoma cells. Interestingly, reduced alpha6beta4 expression also promoted decreased migration on non-laminin substrata indicating that this integrin can function in a ligand-independent manner. In addition, the absence of beta4 expression in these cells augmented the formation of alpha6beta1 heterodimers and increased adhesion to laminin-1. Taken together, these results substantiate the importance of the alpha6beta4 integrin in invasion and migration that has been demonstrated previously by expression of the beta4 subunit in beta4-deficient cell lines and by function blocking antibodies. Furthermore, these data suggest that the utilization of siRNA oligonucleotides to reduce the expression of the alpha6beta4 integrin may be a useful approach to prevent carcinoma cell progression.


Subject(s)
Breast Neoplasms/pathology , Gene Silencing/drug effects , Integrin alpha6beta4/antagonists & inhibitors , Neoplasm Invasiveness/prevention & control , RNA, Small Interfering/pharmacology , Biotinylation , Breast Neoplasms/physiopathology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Dimerization , Female , Humans , Laminin/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...