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










Publication year range
1.
Int J Mol Sci ; 24(10)2023 May 11.
Article in English | MEDLINE | ID: mdl-37239927

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a rare demyelinating disease caused by infection with JC Polyomavirus (JCPyV). Despite the identification of the disease and isolation of the causative pathogen over fifty years ago, no antiviral treatments or prophylactic vaccines exist. Disease onset is usually associated with immunosuppression, and current treatment guidelines are limited to restoring immune function. This review summarizes the drugs and small molecules that have been shown to inhibit JCPyV infection and spread. Paying attention to historical developments in the field, we discuss key steps of the virus lifecycle and antivirals known to inhibit each event. We review current obstacles in PML drug discovery, including the difficulties associated with compound penetrance into the central nervous system. We also summarize recent findings in our laboratory regarding the potent anti-JCPyV activity of a novel compound that antagonizes the virus-induced signaling events necessary to establish a productive infection. Understanding the current panel of antiviral compounds will help center the field for future drug discovery efforts.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , Polyomavirus Infections , Humans , Leukoencephalopathy, Progressive Multifocal/drug therapy , JC Virus/physiology , Signal Transduction
2.
mBio ; 14(2): e0358322, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36786589

ABSTRACT

JC polyomavirus (JCPyV) is a ubiquitous, double-stranded DNA virus that causes the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in immunocompromised patients. Current treatments for PML are limited to immune reconstitution, and no effective antivirals exist. In this report, we show that the oxindole GW-5074 (3-(3,5-dibromo-4-hydroxybenzylidene)-5-iodoindolin-2-one) reduces JCPyV infection in primary and immortalized cells. This compound potently inhibits virus spread, which suggests that it could control infection in PML patients. We demonstrate that GW-5074 inhibits endogenous ERK phosphorylation, and that JCPyV infection in GW-5074-treated cells cannot be rescued with ERK agonists, which indicates that the antiviral mechanism may involve its antagonistic effects on MAPK-ERK signaling. Importantly, GW-5074 exceeds thresholds of common pharmacological parameters that identify promising compounds for further development. This MAPK-ERK antagonist warrants further investigation as a potential treatment for PML. IMPORTANCE Human polyomaviruses, such as JCPyV and BKPyV, cause significant morbidity and mortality in immunocompromised or immunomodulated patients. There are no treatments for polyomavirus-induced diseases other than restoration of immune function. We discovered that the oxindole GW-5074 potently inhibits infection by both JCPyV and BKPyV. Further optimization of this compound could result in the development of antiviral therapies for polyomavirus-induced diseases.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , Polyomavirus Infections , Polyomavirus , Humans , Oxindoles/pharmacology , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/genetics , JC Virus/genetics , MAP Kinase Signaling System , Antiviral Agents
3.
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.

4.
Int J Mol Sci ; 22(18)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34575975

ABSTRACT

Several classes of immunomodulators are used for treating relapsing-remitting multiple sclerosis (RRMS). Most of these disease-modifying therapies, except teriflunomide, carry the risk of progressive multifocal leukoencephalopathy (PML), a severely debilitating, often fatal virus-induced demyelinating disease. Because teriflunomide has been shown to have antiviral activity against DNA viruses, we investigated whether treatment of cells with teriflunomide inhibits infection and spread of JC polyomavirus (JCPyV), the causative agent of PML. Treatment of choroid plexus epithelial cells and astrocytes with teriflunomide reduced JCPyV infection and spread. We also used droplet digital PCR to quantify JCPyV DNA associated with extracellular vesicles isolated from RRMS patients. We detected JCPyV DNA in all patients with confirmed PML diagnosis (n = 2), and in six natalizumab-treated (n = 12), two teriflunomide-treated (n = 7), and two nonimmunomodulated (n = 2) patients. Of the 21 patients, 12 (57%) had detectable JCPyV in either plasma or serum. CSF was uniformly negative for JCPyV. Isolation of extracellular vesicles did not increase the level of detection of JCPyV DNA versus bulk unprocessed biofluid. Overall, our study demonstrated an effect of teriflunomide inhibiting JCPyV infection and spread in glial and choroid plexus epithelial cells. Larger studies using patient samples are needed to correlate these in vitro findings with patient data.


Subject(s)
Crotonates/pharmacology , DNA Viruses/drug effects , Hydroxybutyrates/pharmacology , Leukoencephalopathy, Progressive Multifocal/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Neuroglia/drug effects , Nitriles/pharmacology , Toluidines/pharmacology , Astrocytes/drug effects , Astrocytes/virology , Cell Line , Choroid Plexus/drug effects , Choroid Plexus/virology , DNA Viruses/pathogenicity , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Epithelial Cells/drug effects , Epithelial Cells/virology , Extracellular Vesicles/drug effects , Extracellular Vesicles/virology , Humans , Immunologic Factors/adverse effects , Immunologic Factors/therapeutic use , JC Virus/drug effects , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/chemically induced , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/pathology , Multiple Sclerosis, Relapsing-Remitting/virology , Neuroglia/virology , Virus Diseases/drug therapy , Virus Diseases/genetics , Virus Diseases/virology
5.
Virology ; 548: 17-24, 2020 09.
Article in English | MEDLINE | ID: mdl-32838939

ABSTRACT

The demyelinating disease progressive multifocal leukoencephalopathy (PML) is caused by the human polyomavirus, JCPyV, under conditions of prolonged immunosuppression. Initial infection is asymptomatic, and the virus establishes lifelong persistence in the host. Following the loss of immune surveillance, the virus can traffic to the central nervous system and infect oligodendrocytes to cause demyelination and PML. The mechanisms involved in glial cell infection are not completely understood. In a screen for N-glycosylated proteins that influence JCPyV pathology, we identified Adipocyte Plasma Membrane Associated Protein (APMAP) as a host cell modulator of JCPyV infection. The removal of APMAP by small interfering siRNA as well as by CRISPR-Cas9 gene editing resulted in a significant decrease in JCPyV infection. Exogenous expression of APMAP in APMAP knockout cell lines rescued susceptibility to infection. These data suggest that virus infection of glial cells is dependent on APMAP.


Subject(s)
JC Virus/physiology , Neuroglia/metabolism , Polyomavirus Infections/metabolism , Cell Line , Host-Pathogen Interactions , Humans , JC Virus/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins , Neuroglia/virology , Oligodendroglia/metabolism , Oligodendroglia/virology , Polyomavirus Infections/genetics , Polyomavirus Infections/virology
6.
PLoS Pathog ; 16(3): e1008371, 2020 03.
Article in English | MEDLINE | ID: mdl-32130281

ABSTRACT

The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy (PML) in immunosuppressed and immunomodulated patients. Initial infection with JCPyV is common and the virus establishes a long-term persistent infection in the urogenital system of 50-70% of the human population worldwide. A major gap in the field is that we do not know how the virus traffics from the periphery to the brain to cause disease. Our recent discovery that human choroid plexus epithelial cells are fully susceptible to virus infection together with reports of JCPyV infection of choroid plexus in vivo has led us to hypothesize that the choroid plexus plays a fundamental role in this process. The choroid plexus is known to relay information between the blood and the brain by the release of extracellular vesicles. This is particularly important because human macroglia (oligodendrocytes and astrocytes), the major targets of virus infection in the central nervous system (CNS), do not express the known attachment receptors for the virus and do not bind virus in human tissue sections. In this report we show that JCPyV infected choroid plexus epithelial cells produce extracellular vesicles that contain JCPyV and readily transmit the infection to human glial cells. Transmission of the virus by extracellular vesicles is independent of the known virus attachment receptors and is not neutralized by antisera directed at the virus. We also show that extracellular vesicles containing virus are taken into target glial cells by both clathrin dependent endocytosis and macropinocytosis. Our data support the hypothesis that the choroid plexus plays a fundamental role in the dissemination of virus to brain parenchyma.


Subject(s)
Choroid Plexus/metabolism , Epithelial Cells/metabolism , Extracellular Vesicles/metabolism , JC Virus/metabolism , Leukoencephalopathy, Progressive Multifocal/metabolism , Neuroglia/metabolism , Receptors, Virus/metabolism , Cell Line, Transformed , Choroid Plexus/pathology , Choroid Plexus/virology , Epithelial Cells/pathology , Epithelial Cells/virology , Extracellular Vesicles/pathology , Extracellular Vesicles/virology , Humans , Leukoencephalopathy, Progressive Multifocal/pathology , Neuroglia/pathology , Neuroglia/virology
7.
Cell Rep ; 27(7): 1960-1966.e6, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091436

ABSTRACT

JC polyomavirus (JCPyV) is a ubiquitous human pathogen that causes progressive multifocal leukoencephalopathy (PML). The entry receptors for JCPyV belong to the 5-hydroxytryptamine 2 receptor (5-HT2R) family, but how individual members of the family function to facilitate infection is not known. We used proximity ligation assay (PLA) to determine that JCPyV interacts with each of the 5-HT2 receptors (5-HT2Rs) in a narrow window of time during entry. We used CRISPR-Cas9 to randomly introduce stop codons in the gene for each receptor and discovered that the second intracellular loop of each was necessary for infection. This loop contains a motif possibly involved in receptor internalization by ß-arrestin. Mutation of this motif and small interfering RNA (siRNA) knockdown of ß-arrestin recapitulated the results of our CRISPR-Cas9 screen, showing that this motif is critical. Our results have implications for the role these receptors play in virus infection and for their normal functioning as receptors for serotonin.


Subject(s)
JC Virus/genetics , Receptors, Serotonin, 5-HT2/genetics , Receptors, Serotonin, 5-HT2/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Virus Internalization , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , JC Virus/pathogenicity , beta-Arrestins/genetics , beta-Arrestins/metabolism
8.
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
9.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29437972

ABSTRACT

JC polyomavirus (JCPyV) establishes a lifelong persistence in roughly half the human population worldwide. The cells and tissues that harbor persistent virus in vivo are not known, but renal tubules and other urogenital epithelial cells are likely candidates as virus is shed in the urine of healthy individuals. In an immunosuppressed host, JCPyV can become reactivated and cause progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease of the central nervous system. Recent observations indicate that JCPyV may productively interact with cells in the choroid plexus and leptomeninges. To further study JCPyV infection in these cells, primary human choroid plexus epithelial cells and meningeal cells were challenged with virus, and their susceptibility to infection was compared to the human glial cell line, SVG-A. We found that JCPyV productively infects both choroid plexus epithelial cells and meningeal cells in vitro Competition with the soluble receptor fragment LSTc reduced virus infection in these cells. Treatment of cells with neuraminidase also inhibited both viral infection and binding. Treatment with the serotonin receptor antagonist, ritanserin, reduced infection in SVG-A and meningeal cells. We also compared the ability of wild-type and sialic acid-binding mutant pseudoviruses to transduce these cells. Wild-type pseudovirus readily transduced all three cell types, but pseudoviruses harboring mutations in the sialic acid-binding pocket of the virus failed to transduce the cells. These data establish a novel role for choroid plexus and meninges in harboring virus that likely contributes not only to meningoencephalopathies but also to PML.IMPORTANCE JCPyV infects greater than half the human population worldwide and causes central nervous system disease in patients with weakened immune systems. Several recent reports have found JCPyV in the choroid plexus and leptomeninges of patients with encephalitis. Due to their role in forming the blood-cerebrospinal fluid barrier, the choroid plexus and leptomeninges are also poised to play roles in virus invasion of brain parenchyma, where infection of macroglial cells leads to the development of progressive multifocal leukoencephalopathy, a severely debilitating and often fatal infection. In this paper we show for the first time that primary choroid plexus epithelial cells and meningeal cells are infected by JCPyV, lending support to the association of JCPyV with meningoencephalopathies. These data also suggest that JCPyV could use these cells as reservoirs for the subsequent invasion of brain parenchyma.


Subject(s)
Choroid Plexus , Epithelial Cells , JC Virus/metabolism , Leukoencephalopathy, Progressive Multifocal , Meninges , Ritanserin/pharmacology , Cell Line , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/virology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/metabolism , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Meninges/metabolism , Meninges/pathology , Meninges/virology
10.
Annu Rev Virol ; 4(1): 349-367, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28637388

ABSTRACT

In 1971, the first human polyomavirus was isolated from the brain of a patient who died from a rapidly progressing demyelinating disease known as progressive multifocal leukoencephalopathy. The virus was named JC virus after the initials of the patient. In that same year a second human polyomavirus was discovered in the urine of a kidney transplant patient and named BK virus. In the intervening years it became clear that both viruses were widespread in the human population but only rarely caused disease. The past decade has witnessed the discovery of eleven new human polyomaviruses, two of which cause unusual and rare cancers. We present an overview of the history of these viruses and the evolution of JC polyomavirus-induced progressive multifocal leukoencephalopathy over three different epochs. We review what is currently known about JC polyomavirus, what is suspected, and what remains to be done to understand the biology of how this mostly harmless endemic virus gives rise to lethal disease.


Subject(s)
JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Polyomavirus/isolation & purification , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/virology , Animals , Autoimmune Diseases/virology , BK Virus/isolation & purification , BK Virus/pathogenicity , Humans , JC Virus/isolation & purification , Leukoencephalopathy, Progressive Multifocal/etiology , Mice , Multiple Sclerosis/complications , Polyomavirus/classification , Polyomavirus/pathogenicity , Polyomavirus Infections/virology , Tumor Virus Infections/virology
11.
Am J Pathol ; 185(8): 2246-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26056932

ABSTRACT

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.


Subject(s)
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
12.
J Clin Invest ; 124(12): 5103-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401466

ABSTRACT

JC virus (JCV) causes progressive multifocal leukoencephalopathy (PML), a demyelinating disease in humans. The disease, once considered fatal, is now managed with immune reconstitution therapy; however, surviving patients remain severely debilitated. Until now, there has been no animal model to study JCV in the brain, and research into treatment has relied on cell culture systems. In this issue of the JCI, Kondo and colleagues developed a mouse model in which human glial cells are engrafted into neonatal mice that are both immunodeficient and deficient for myelin basic protein. When challenged intracerebrally with JCV, these mice exhibit some of the characteristics of PML. The establishment of this chimeric mouse model is a significant advance toward understanding the mechanism of JCV pathogenesis and the identification of drugs to treat or prevent the disease.


Subject(s)
Astrocytes/immunology , JC Virus/physiology , Leukoencephalopathy, Progressive Multifocal/immunology , Stem Cell Transplantation , Stem Cells/immunology , Transplantation Chimera/immunology , Virus Replication/immunology , Animals , Female , Humans , Male
13.
J Virol ; 87(24): 13490-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089568

ABSTRACT

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.


Subject(s)
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
14.
J Virol ; 84(19): 9677-84, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660194

ABSTRACT

JC virus (JCV) is a human polyomavirus and the causative agent of the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML). JCV infection of host cells is dependent on interactions with cell surface asparagine (N)-linked sialic acids and the serotonin 5-hydroxytryptamine(2A) receptor (5-HT(2A)R). The 5-HT(2A)R contains five potential N-linked glycosylation sites on the extracellular N terminus. Glycosylation of other serotonin receptors is essential for expression, ligand binding, and receptor function. Also, glycosylation of cellular receptors has been reported to be important for JCV infection. Therefore, we hypothesized that the 5-HT(2A)R N-linked glycosylation sites are required for JCV infection. Treatment of 5-HT(2A)R-expressing cells with tunicamycin, an inhibitor of N-linked glycosylation, reduced JCV infection. Individual mutation of each of the five N-linked glycosylation sites did not affect the capacity of 5-HT(2A)R to support JCV infection and did not alter the cell surface expression of the receptor. However, mutation of all five N-linked glycosylation sites simultaneously reduced the capacity of 5-HT(2A)R to support infection and altered the cell surface expression. Similarly, tunicamycin treatment reduced the cell surface expression of 5-HT(2A)R. Mutation of all five N-linked glycosylation sites or tunicamycin treatment of cells expressing wild-type 5-HT(2A)R resulted in an altered electrophoretic mobility profile of the receptor. Treatment of cells with PNGase F, to remove N-linked oligosaccharides from the cell surface, did not affect JCV infection in 5-HT(2A)R-expressing cells. These data affirm the importance of 5-HT(2A)R as a JCV receptor and demonstrate that the sialic acid component of the receptor is not directly linked to 5-HT(2A)R.


Subject(s)
JC Virus/pathogenicity , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Virus/metabolism , Base Sequence , Binding Sites/genetics , Cell Line , DNA Primers/genetics , Glycosylation/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , JC Virus/physiology , Leukoencephalopathy, Progressive Multifocal/etiology , Leukoencephalopathy, Progressive Multifocal/metabolism , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid/chemistry , Neuraminidase/pharmacology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Virus/chemistry , Receptors, Virus/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Tunicamycin/pharmacology
15.
Pediatr Res ; 68(1): 57-62, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20375852

ABSTRACT

We determined the time-specific effects of FasL overexpression on perinatal alveolar type II cell growth kinetics. To achieve temporal overexpression of respiratory epithelium-specific FasL expression, tetracycline inducible CCSP-rtTA/FasL-TetOp transgenic mice were given doxycycline (Dox) from gestational d 14 (E14) to E19 (antenatal treatment group), from postnatal d 1 (P1) to P7 (postnatal group), or from E14 to P7 (combined antenatal and postnatal group). Antenatal Dox administration induced an increase of pulmonary FasL mRNA levels in double transgenic animals up to >300-fold over single transgenic littermate controls, associated with massive fetal respiratory epithelial apoptosis and excessive postnatal lethality. Although animals from the combined antenatal/postnatal Dox treatment group continued to display evidence of increased apoptosis, there was a paradoxical increase in alveolar type II cell proliferation, resulting in a net increase in type II cell density, elevated pulmonary surfactant protein C levels and improved postnatal survival. Postnatal Dox administration was also associated with increased type II cell density, although FasL up-regulation was more variable. In conclusion, these results, and our previous studies, suggest that FasL signaling has dual timing-dependent proapoptotic and proproliferative effects on postcanalicular type II cell kinetics.


Subject(s)
Fas Ligand Protein/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/embryology , Pulmonary Alveoli/growth & development , Animals , Animals, Newborn , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Proliferation/drug effects , Doxycycline/pharmacology , Fas Ligand Protein/genetics , Mice , Mice, Transgenic , Pulmonary Alveoli/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Survival Rate
16.
Virus Res ; 149(1): 128-32, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20122973

ABSTRACT

Carcinoid syndrome is caused by the unregulated secretion of bioactive amines from neuroendocrine tumors arising primarily in the gastrointestinal tract and lungs. The incidence of carcinoid syndrome is 1-2/100,000 and the syndrome is thought to be increasing. Carcinoid tumors are relatively slow growing but can become highly metastatic. Currently, there is no effective therapy to inhibit cell proliferation or metastasis of neuroendocrine tumor (NET) disease. Polyomaviruses are a family of viruses that are able to transform cells and promote tumor formation. In this study, the polyomaviruses SV40, JCV, and BKV were used to assess the ability of polyomaviruses to productively infect a range of human carcinoid cell lines. Infection was assessed by the immunofluorescence detection of T antigen and V antigen. Viruses and cell lines that exhibited productive infections were subsequently assayed by FACS analysis for cell binding and dual promoter luciferase assay for early and late promoter activity. Most carcinoid cell lines were not susceptible to infection by polyomaviruses. However, BKV efficiently infected the pulmonary carcinoid H727 cell line but did not infect a control, non-carcinoid lung cell line (A549). BKV was found to bind to both the susceptible H727 cells and to the non-susceptible A549 cells but viral genes were only efficiently expressed in the H727 cell line. The data demonstrate that BKV can infect human pulmonary carcinoid cells. Infection does not seem to be solely mediated by the virus' ability to bind to cells, as the virus will also bind to non-carcinoid control cells. Both early and late gene expression are supported by the pulmonary carcinoid cells.


Subject(s)
BK Virus/pathogenicity , Carcinoid Tumor/virology , Lung Neoplasms/virology , Antigens, Viral, Tumor/biosynthesis , BK Virus/growth & development , Cell Line, Tumor , Flow Cytometry , Gene Expression Profiling , Genes, Reporter , Humans , JC Virus/growth & development , JC Virus/pathogenicity , Luciferases/biosynthesis , Luciferases/genetics , Simian virus 40/growth & development , Simian virus 40/pathogenicity , Viral Structural Proteins/biosynthesis , Virus Attachment , Virus Replication
17.
Am J Pathol ; 173(1): 42-56, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18535181

ABSTRACT

Premature infants are at risk for bronchopulmonary dysplasia, a complex condition characterized by impaired alveolar development and increased alveolar epithelial apoptosis. The functional involvement of pulmonary apoptosis in bronchopulmonary dysplasia- associated alveolar disruption remains undetermined. The aims of this study were to generate conditional lung-specific Fas-ligand (FasL) transgenic mice and to determine the effects of FasL-induced respiratory epithelial apoptosis on alveolar remodeling in postcanalicular lungs. Transgenic (TetOp)(7)-FasL responder mice, generated by pronuclear microinjection, were bred with Clara cell secretory protein (CCSP)-rtTA activator mice. Doxycycline (Dox) was administered from embryonal day 14 to postnatal day 7, and lungs were studied between embryonal day 19 and postnatal day 21. Dox administration induced marked respiratory epithelium-specific FasL mRNA and protein up-regulation in double-transgenic CCSP-rtTA(+)/(TetOp)(7)-FasL(+) mice compared with single-transgenic CCSP-rtTA(+) littermates. The Dox-induced FasL up-regulation was associated with dramatically increased apoptosis of alveolar type II cells and Clara cells, disrupted alveolar development, decreased vascular density, and increased postnatal lethality. These data demonstrate that FasL-induced alveolar epithelial apoptosis during postcanalicular lung remodeling is sufficient to disrupt alveolar development after birth. The availability of inducible lung-specific FasL transgenic mice will facilitate studies of the role of apoptosis in normal and disrupted alveologenesis and may lead to novel therapeutic approaches for perinatal and adult pulmonary diseases characterized by dysregulated apoptosis.


Subject(s)
Apoptosis/physiology , Epithelial Cells/pathology , Fas Ligand Protein/metabolism , Lung Diseases/pathology , Pulmonary Alveoli/growth & development , Animals , Base Sequence , Blotting, Western , Disease Models, Animal , Fas Ligand Protein/genetics , Female , Immunohistochemistry , In Situ Nick-End Labeling , Lung Diseases/physiopathology , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Molecular Sequence Data , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
18.
BMC Physiol ; 8: 4, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18312674

ABSTRACT

BACKGROUND: Myocardial hypoxic-ischemic injury is the cause of significant morbidity and mortality worldwide. The cardiomyocyte response to hypoxic-ischemic injury is known to include changes in cell cycle regulators. The cyclin-dependent kinase inhibitor p57Kip2 is involved in cell cycle control, differentiation, stress signaling and apoptosis. In contrast to other cyclin-dependent kinase inhibitors, p57Kip2 expression diminishes during postnatal life and is reactivated in the adult heart under conditions of cardiac stress. Overexpression of p57Kip2 has been previously shown to prevent apoptotic cell death in vitro by inhibiting stress-activated kinases. Therefore, we hypothesized that p57Kip2 has a protective role in cardiomyocytes under hypoxic conditions. To investigate this hypothesis, we created a transgenic mouse (R26loxpTA-p57k/+) that expresses p57Kip2 specifically in cardiac tissue under the ventricular cardiomyocyte promoter Mlc2v. RESULTS: Transgenic mice with cardiac specific overexpression of p57Kip2 are viable, fertile and normally active and their hearts are morphologically indistinguishable from the control hearts and have similar heart weight/body weight ratio. The baseline functional parameters, including left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP), LVdp/dtmax, heart rate (HR) and rate pressure product (RPR) were not significantly different between the different groups as assessed by the Langendorff perfused heart preparation. However, after subjecting the heart ex vivo to 30 minutes of ischemia-reperfusion injury, the p57Kip2 overexpressing hearts demonstrated preserved cardiac function compared to control mice with higher left ventricular developed pressure (63 +/- 15 vs 30 +/- 6 mmHg, p = 0.05), rate pressure product (22.8 +/- 4.86 vs 10.4 +/- 2.1 x 103bpm x mmHg, p < 0.05) and coronary flow (3.5 +/- 0.5 vs 2.38 +/- 0.24 ml/min, p <0.05). CONCLUSION: These data suggest that forced cardiac expression of p57Kip2 does not affect myocardial growth, differentiation and baseline function but attenuates injury from ischemia-reperfusion in the adult mouse heart.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Cardiotonic Agents/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/genetics , Mice , Mice, Transgenic
19.
Dev Biol ; 272(1): 191-202, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15242800

ABSTRACT

Sea urchin eggs secrete a serine protease activity, CGSP1, at fertilization that is essential for the block to polyspermy. Several targets of this proteolytic activity on the plasma membrane were identified here using a cell surface biotinylation approach. Amino acid microsequencing of one of these proteins led to the identification of a 4.75-kb cDNA clone from a Strongylocentrotus purpuratus ovary cDNA library that encodes a 160-kDa protein called p160. This protein contains five CUB domains and a putative transmembrane domain suggesting that p160 is an integral membrane protein with protein-protein interaction motifs facing the extracellular matrix of the egg. Whole-mount immunolocalization studies demonstrate that p160 is on the surface of the egg, enriched at the tips of microvilli. The protein is removed at fertilization in a protease-dependent manner, and functional assays suggest that p160 serves to link the plasma membrane to the vitelline layer until fertilization. Thus, p160 is a key candidate for a vitelline-layer linker protein, the selective proteolysis of which functions in the block to polyspermy in the sea urchin egg.


Subject(s)
Membrane Proteins/metabolism , Ovum/metabolism , Sea Urchins/physiology , Sperm-Ovum Interactions , Vitelline Membrane/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/metabolism , Embryo, Nonmammalian , Endopeptidases/metabolism , Female , Fertilization/physiology , Membrane Proteins/genetics , Molecular Sequence Data , Sea Urchins/embryology , Serine Endopeptidases/metabolism
20.
Mol Biol Cell ; 15(5): 2084-92, 2004 May.
Article in English | MEDLINE | ID: mdl-14978210

ABSTRACT

Cortical granules are specialized organelles whose contents interact with the extracellular matrix of the fertilized egg to form the block to polyspermy. In sea urchins, the granule contents form a fertilization envelope (FE), and this construction is critically dependent upon protease activity. An autocatalytic serine protease, cortical granule serine protease 1 (CGSP1), has been identified in the cortical granules of Strongylocentrotus purpuratus eggs, and here we examined the regulation of the protease activity and tested potential target substrates of CGSP1. We found that CGSP1 is stored in its full-length, enzymatically quiescent form in the granule, and is inactive at pH 6.5 or below. We determined the pH of the cortical granule by fluorescent indicators and micro-pH probe measurements and found the granules to be pH 5.5, a condition inhibitory to CGSP1 activity. Exposure of the protease to the pH of seawater (pH 8.0) at exocytosis immediately activates the protease. Activation of eggs at pH 6.5 or lower blocks activation of the protease and the resultant FE phenotypes are indistinguishable from a protease-null phenotype. We find that native cortical granule targets of the protease are beta-1,3 glucanase, ovoperoxidase, and the protease itself, but the structural proteins of the granule are not proteolyzed by CGSP1. Whole mount immunolocalization experiments demonstrate that inhibition of CGSP1 activity affects the localization of ovoperoxidase but does not alter targeting of structural proteins to the FE. The mistargeting of ovoperoxidase may lead to spurious peroxidative cross-linking activity and contribute to the lethality observed in protease-null cells. Thus, CGSP1 is proteolytically active only when secreted, due to the low pH of the cortical granules, and it has a small population of targets for cleavage within the cortical granules.


Subject(s)
Fertilization/physiology , Sea Urchins/embryology , Sea Urchins/enzymology , Secretory Vesicles/enzymology , Serine Endopeptidases/physiology , Zygote/enzymology , Animals , Benzamidines/pharmacology , Glucan 1,3-beta-Glucosidase/metabolism , Hyalin/metabolism , Hydrogen-Ion Concentration , Microscopy, Fluorescence , Peroxidases/metabolism , Seawater/chemistry , Serine Endopeptidases/isolation & purification , Serine Proteinase Inhibitors/pharmacology , Substrate Specificity , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...