Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Virology ; 468-470: 57-62, 2014 Nov.
Article En | MEDLINE | ID: mdl-25146733

Some viruses and most eukaryotic cells have microRNAs that regulate the expression of many genes. Although many viral miRNAs have been identified, only a few have been included in in vivo functional studies. Here we show that a Py-encoded miRNA downregulates the expression of the pro-apoptotic factor Smad2, resulting in the suppression of the apoptosis pathway. To study the Py miRNA in an in vivo context, a miRNA-deficient mutant virus was created on the background of the LID virus strain which establishes a rapid and lethal infection in newborn mice. Apoptosis analysis on kidney tissues indicates that the pro-apoptotic pathway is targeted in the infected host as well. Suppression of apoptosis through targeting of Smad2 by the Py miRNA is expected to synergize with anti-apoptotic effects previously attributed to the polyoma tumor antigens in support of virus replication in the natural host.


Apoptosis/physiology , Epithelial Cells/physiology , Epithelial Cells/virology , MicroRNAs/metabolism , Polyomavirus/classification , Smad2 Protein/metabolism , Animals , Cell Line , Lentivirus , Mice , MicroRNAs/genetics , RNA Interference , Smad2 Protein/genetics
2.
FASEB J ; 25(4): 1275-83, 2011 Apr.
Article En | MEDLINE | ID: mdl-21228219

The evolutionarily conserved SALL genes encode transcription factors with roles in embryonic development. The product of the SALL2 gene was first identified as a binding partner of the mouse polyoma virus large T antigen and later shown to possess tumor suppressor-like functions. Independent studies identified SALL2 as a factor regulating the quiescent state in human fibroblasts. Here, we investigate factors that regulate the expression of SALL2 and turnover of p150(Sal2) in growing vs. resting cells. The transcription factor AP4 increases along with SALL2 in quiescent cells and positively regulates SALL2 expression. TGFß effectively inhibits expression of SALL2 and its regulator AP4 when added to quiescent fibroblasts. TGFß repression of SALL2 and AP4 is independent of the induction of connective tissue growth factor (CTGF) by TGFß. p150(Sal2) disappears rapidly on restoration of serum. In both growing fibroblasts and established ovarian surface epithelial cells, p150(Sal2) undergoes polyubiquitination and proteosomal degradation. A CUL4/DDB1 E3 ligase containing RBBP7 as the p150(Sal2) receptor has been identified as mediating the destruction of p150(Sal2) as cells transition from a quiescent to an actively growing state.


Protein Processing, Post-Translational , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Connective Tissue Growth Factor/physiology , Cullin Proteins/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Female , Foreskin/metabolism , HEK293 Cells , HeLa Cells , Humans , Leupeptins/pharmacology , Male , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Serum , Transcription Factors/pharmacology , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
Virology ; 409(2): 204-10, 2011 Jan 20.
Article En | MEDLINE | ID: mdl-21056449

We have examined the effect of expression of the protein tyrosine phosphatase Shp2 on transformation by the mouse polyoma virus middle T antigen (PyMT). Gain-of-function mutations in Shp2 indicate that it may serve as an oncogene in several types of human leukemia. Paradoxically, however, some catalytically dominant-negative mutations of Shp2 have also been identified in leukemia and neuroblastomas. In this study, we show that Shp2 suppresses transformation induced by PyMT, the major polyoma viral oncoprotein known to act through binding and activation of pp60(c-src). Over-expression of a catalytically inactive Shp2 mutant in NIH3T3 cells significantly enhanced PyMT-induced transformation. Conversely, re-introduction of Shp2 into Shp2-deficient cells strongly inhibited PyMT-induced transformation and tumorigenesis. Short hairpin RNA (shRNA)-mediated Shp2 knockdown potentiated PyMT-induced transformation. Finally, we present evidence that the transformation-suppressive effects of Shp2 are mediated at least partially through the inhibition of signal transducers and activators of transcription 3.


Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Neoplastic , Fibroblasts/virology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line , Gene Knockdown Techniques , Mice , Mice, Inbred BALB C , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
4.
Virology ; 387(1): 157-67, 2009 Apr 25.
Article En | MEDLINE | ID: mdl-19272626

MicroRNAs are small regulatory RNAs that post-transcriptionally regulate gene expression and can be encoded by viral as well as cellular genomes. The functions of most viral miRNAs are unknown and few have been studied in an in vivo context. Here we show that the murine polyomavirus (PyV) encodes a precursor microRNA that is processed into two mature microRNAs, both of which are active at directing the cleavage of the early PyV mRNAs. Furthermore, we identify a deletion mutant of polyomavirus that is defective in encoding the microRNAs. This mutant replicates normally and transforms cultured cells with efficiencies comparable to wildtype PyV. The miRNA mutant is competent to establish a transient infection of mice following parenteral inoculation, and is cleared post infection at approximately the same rate as the wildtype virus. In addition, under these laboratory conditions, we observe no differences in anti-viral CD8 T cell responses. These results indicate that PyV miRNA expression is not essential for infection of cultured cells or experimentally inoculated mice, and raise the possibility that its role in natural infection might involve aspects of acquisition or spread that are not recapitulated by experimental inoculation.


MicroRNAs/metabolism , Polyomavirus Infections/metabolism , Polyomavirus/physiology , RNA, Viral/metabolism , Animals , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Line , Gene Expression Regulation, Viral , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Polyomavirus/genetics , Polyomavirus Infections/genetics , RNA, Messenger/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics
5.
J Virol ; 82(10): 4946-54, 2008 May.
Article En | MEDLINE | ID: mdl-18337582

Middle T antigen (MT) is the principal oncoprotein of murine polyomavirus. Experiments on the acute immediate effects of MT expression on cellular RNA levels showed that expression of osteopontin (OPN) was strongly induced by MT expression. Osteopontin is a protein known to be associated with cancer. It has a role in tumor progression and invasion. Protein analysis confirmed that MT induced the secretion of OPN into the extracellular medium. Expression of antisense OPN RNA had no effect on the growth of MT-transformed cells. However, it had a strong effect on the ability of MT transformants to migrate or to fill a wound. Analysis of MT mutants implicated both the SHC and phosphatidylinositol 3-kinase pathways in OPN induction. Reporter assays showed that MT regulated the OPN promoter through two of its PEA3 (polyoma enhancer activator 3) sites. As critical PEA3 sites are also part of the polyomavirus enhancer, the same signaling important for viral replication also contributes to virally induced metastatic potential.


Antigens, Polyomavirus Transforming/metabolism , Cell Movement , Gene Expression Profiling , Osteopontin/biosynthesis , Polyomavirus/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Artificial Gene Fusion , Cell Line , Cell Proliferation , Enhancer Elements, Genetic , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , RNA, Antisense/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
6.
J Virol ; 81(18): 10064-71, 2007 Sep.
Article En | MEDLINE | ID: mdl-17626093

Minichromosomes of wild-type polyomavirus were previously shown to be highly acetylated on histones H3 and H4 compared either to bulk cell chromatin or to viral chromatin of nontransforming hr-t mutants, which are defective in both the small T and middle T antigens. A series of site-directed virus mutants have been used along with antibodies to sites of histone modifications to further investigate the state of viral chromatin and its dependence on the T antigens. Small T but not middle T was important in hyperacetylation at major sites in H3 and H4. Mutants blocked in middle T signaling pathways but encoding normal small T showed a hyperacetylated pattern similar to that of wild-type virus. The hyperacetylation defect of hr-t mutant NG59 was partially complemented by growth of the mutant in cells expressing wild-type small T. In contrast to the hypoacetylated state of NG59, NG59 minichromosomes were hypermethylated at specific lysines in H3 and also showed a higher level of phosphorylation at H3ser10, a modification associated with the late G(2) and M phases of the cell cycle. Comparisons of virus growth kinetics and cell cycle progression in wild-type- and NG59-infected cells showed a correlation between the phase of the cell cycle at which virus assembly occurred and histone modifications in the progeny virus. Replication and assembly of wild-type virus were completed largely during S phase. Growth of NG59 was delayed by about 12 h with assembly occurring predominantly in G(2). These results suggest that small T affects modifications of viral chromatin by altering the temporal coordination of virus growth and the cell cycle.


Antigens, Polyomavirus Transforming/metabolism , Cell Cycle , Chromatin/metabolism , Polyomavirus/growth & development , Virus Assembly/physiology , Virus Replication/physiology , Acetylation , Amino Acid Substitution , Animals , Antigens, Polyomavirus Transforming/genetics , Chromatin/genetics , Chromatin/virology , Histones/genetics , Histones/metabolism , Kinetics , Mice , Mutagenesis, Site-Directed , Mutation, Missense , NIH 3T3 Cells , Polyomavirus/genetics , Signal Transduction/genetics
7.
J Virol ; 80(17): 8739-44, 2006 Sep.
Article En | MEDLINE | ID: mdl-16912321

The pathways by which viruses enter cells are diverse, but in all cases, infection necessitates the transfer of the viral genome across a cellular membrane. Polyomavirus (Py) particles, after binding to glycolipid and glycoprotein receptors at the cell surface, are delivered to the lumen of the endoplasmic reticulum (ER). The nature and extent of virus disassembly in the ER, how the viral genome is transported to the cytosol and subsequently to the nucleus, and whether any cellular proteins are involved are not known. Here, we identify an ER-resident protein, Derlin-2, a factor implicated in the removal of misfolded proteins from the ER for cytosolic degradation, as a component of the machinery required for mouse Py to establish an infection. Inhibition of Derlin-2 function by expression of either a dominant-negative form of Derlin-2 or a short hairpin RNA that reduces Derlin-2 levels blocks Py infection by 50 to 75%. The block imposed by Derlin-2 inhibition occurs after the virus reaches the ER and can be bypassed by the introduction of Py DNA into the cytosol. These findings suggest a mode of Py entry that involves cytosolic access via the quality control machinery in the ER.


Endoplasmic Reticulum/metabolism , Neoplasm Proteins/metabolism , Polyomavirus/pathogenicity , Animals , Cell Line, Tumor , Endoplasmic Reticulum/virology , HeLa Cells , Humans , Membrane Proteins , Mice , Rats
8.
J Immunol ; 176(2): 1148-53, 2006 Jan 15.
Article En | MEDLINE | ID: mdl-16394003

PERA/Ei (PE) mice are highly susceptible to tumor induction by polyoma virus, whereas C57BR/cdj (BR) mice are highly resistant. PE mice respond to viral infection with a type 2 (IL-10) and BR mice with a type 1 (IL-12) cytokine response, underlining the importance of a sustained T cell response for effective antitumor immunity. PE and BR mice showed comparable Ab responses to the virus, indicating that a Th1 response is fully compatible with strong humoral immunity. Tumor susceptibility is dominant, and a type 2 response prevails in F1 mice derived from these strains. In this study, we show that the different cytokine responses of virus-infected hosts are recapitulated in vitro by exposure of APCs from uninfected PE, BR, and F1 animals to the virus. Importantly, virus-like particles formed from recombinant VP1, the major viral capsid protein, elicited the same host-specific cytokine responses as infectious virus. Assembly of VP1 pentamers into capsid shells is required because unassembled VP1 pentamers were ineffective. Binding of virus-like particles to sialic acid is required because pretreatment of APCs with neuraminidase prevented the response. Expression of TLR2 and TLR4 differed among different subpopulations of APCs and also between resistant and susceptible mice. Evidence is presented indicating that these TLRs play a role in mediating the host-specific cytokine responses to the virus.


Antigen-Presenting Cells/immunology , Cytokines/biosynthesis , Polyomavirus/immunology , Animals , Antibodies, Viral/biosynthesis , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred Strains , Neoplasms, Experimental/etiology , Neoplasms, Experimental/immunology , Neutralization Tests , Species Specificity , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Up-Regulation , Virion/immunology
9.
J Virol ; 79(20): 13007-17, 2005 Oct.
Article En | MEDLINE | ID: mdl-16189003

Progression from G(1) to S is essential for polyomavirus DNA replication and depends on the interaction of large T with the retinoblastoma gene product pRb. This virus-induced replication pathway is accompanied by p53 activation resembling a DNA damage response (12). We sought to determine whether this pathway depends in part on activation of the ATM (ataxia telangiectasia mutated) kinase and whether the virus gains advantages from this pathway beyond that of entry into S. We show that polyomavirus infection activates the S- and G(2)-phase checkpoints in primary as well as established mouse cells. Infected cells undergo a prolonged S phase compared to uninfected serum-stimulated cells and show no evidence of a G(2)-->M transition before lytic death ensues. Infection is accompanied by increases in ATM activity in vitro and in the level of ATM-S1981-P in vivo. The incubation of infected cells with caffeine, a known ATM inhibitor, did not block entry into S but reduced the rate of viral compared to cellular DNA synthesis. Importantly, caffeine lowered the yields of viral DNA an average of 3- to 6-fold and those of infectious virus by as much as 10-fold. Virus yields were 10-fold lower in ATM (-/-) p53(-/-) than in ATM(+/+) p53(-/-) mouse embryo fibroblasts, indicating a p53-independent role of ATM in productive infection. Replacement of the normal SMC1 (structural maintenance of chromosomes, or cohesin) protein, a critical ATM substrate in the DNA repair pathway, with its phosphorylation mutant SMC1(S957AS966A) also lowered virus yields by roughly 90%. We suggest that polyomavirus activates and utilizes a component(s) of an ATM pathway of DNA repair to prolong S phase and aid its own replication.


Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Polyomavirus Infections/virology , Polyomavirus/growth & development , Protein Serine-Threonine Kinases/physiology , Signal Transduction , Tumor Suppressor Proteins/physiology , Tumor Virus Infections/virology , Animals , Ataxia Telangiectasia Mutated Proteins , BALB 3T3 Cells , Cell Cycle , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
10.
EMBO J ; 22(17): 4346-55, 2003 Sep 01.
Article En | MEDLINE | ID: mdl-12941687

Polyoma virus (Py) and simian virus 40 (SV40) travel from the plasma membrane to the endoplasmic reticulum (ER) from where they enter the cytosol and then the nucleus to initiate infection. Here we demonstrate that specific gangliosides can serve as plasma membrane receptors for these viruses, GD1a and GT1b for Py and GM1 for SV40. Binding and flotation assays were used to show that addition of these gangliosides to phospholipid vesicles allowed specific binding of the respective viruses. The crystal structure of polyoma VP1 with a sialic acid-containing oligosaccharide was used to derive a model of how the two terminal sugars (sialic acid-alpha2,3-galactose) in one branch of GD1a and GT1b are recognized by the virus. A rat cell line deficient in ganglioside synthesis is poorly infectible by polyoma and SV40, but addition of the appropriate gangliosides greatly facilitates virus uptake, transport to the ER and infection. Lipid binding sites for polyoma are shown to be present in rough ER membranes, suggesting that the virus travel with the ganglioside(s) from the plasma membranes to the ER.


Gangliosides/physiology , Polyomavirus/physiology , Receptors, Virus/physiology , Simian virus 40/physiology , Animals , Capsid Proteins/chemistry , Capsid Proteins/physiology , Cell Line , Cell Membrane/virology , Endoplasmic Reticulum/virology , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/physiology , Gangliosides/chemistry , Humans , In Vitro Techniques , Macromolecular Substances , Mice , Models, Molecular , Molecular Structure , Polyomavirus/pathogenicity , Rats , Receptors, Virus/chemistry , Simian virus 40/pathogenicity
11.
J Virol ; 77(4): 2615-22, 2003 Feb.
Article En | MEDLINE | ID: mdl-12552000

The murine polyomavirus (Py) enters mouse fibroblasts and kidney epithelial cells via an endocytic pathway that is caveola-independent (as well as clathrin-independent). In contrast, uptake of simian virus 40 into the same cells is dependent on caveola. Following the initial uptake of Py, both microtubules and microfilaments play roles in trafficking of the virus to the nucleus. Colcemid, which disrupts microtubules, inhibits the ability of Py to reach the nucleus and replicate. Paclitaxel, which stabilizes microtubules and prevents microtubule turnover, has no effect, indicating that intact but not dynamic microtubules are required for Py infectivity. Compounds that disrupt actin filaments enhance Py uptake while stabilization of actin filaments impedes Py infection. Virus particles are seen in association with actin in cells treated with microfilament-disrupting or filament-stabilizing agents at levels comparable to those in untreated cells, suggesting that a dynamic state of the microfilament system is important for Py infectivity.


Polyomavirus/physiology , Polyomavirus/pathogenicity , Simian virus 40/physiology , Simian virus 40/pathogenicity , 3T3 Cells/virology , Actins/metabolism , Animals , Caveolae/drug effects , Caveolae/virology , Cells, Cultured , Cytoskeleton/metabolism , Demecolcine/pharmacology , Epithelial Cells/virology , Kidney/cytology , Kidney/virology , Mice , Microtubules , Nystatin/pharmacology , Paclitaxel/pharmacology
12.
J Virol ; 76(19): 9657-63, 2002 Oct.
Article En | MEDLINE | ID: mdl-12208944

Mice of the PERA/Ei strain (PE mice) are highly susceptible to tumor induction by polyomavirus and transmit their susceptibility in a dominant manner in crosses with resistant C57BR/cdJ mice (BR mice). BR mice respond to polyomavirus infection with a type 1 cytokine response and develop effective cell-mediated immunity to the virus-induced tumors. By enumerating virus-specific CD8(+) T cells and measuring cytokine responses, we show that the susceptibility of PE mice is due to the absence of a type 1 cytokine response and a concomitant failure to sustain virus-specific cytotoxic T lymphocytes. (PE x BR)F(1) mice showed an initial type 1 response that became skewed toward type 2. Culture supernatants of splenocytes from infected PE mice stimulated in vitro contained high levels of interleukin-10 and no detectable gamma interferon, while those from BR mice showed the opposite pattern. Differences in the innate immune response to polyomavirus by antigen-presenting cells in PE mice and BR mice led to polarization of T-cell cytokine responses. Adherent cells from spleens of infected BR mice produced high levels of interleukin-12, while those from infected PE and F(1) mice produced predominantly interleukin-10. PE and F(1) mice infected by polyomavirus responded with increases in antigen-presenting cells expressing B7.2 costimulatory molecules, whereas BR mice responded with increased expression of B7.1. Administration of recombinant interleukin-12 along with virus resulted in partial protection of PE mice and provided complete protection against tumor development in F(1) animals.


Immunity, Innate , Neoplasms, Experimental/immunology , Polyomavirus Infections/immunology , Tumor Virus Infections/immunology , Animals , Antigens, CD/analysis , B7-1 Antigen/analysis , B7-2 Antigen , Cytokines/biosynthesis , Disease Susceptibility , Interleukin-12/pharmacology , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocytes/immunology
13.
J Virol ; 76(18): 9526-32, 2002 Sep.
Article En | MEDLINE | ID: mdl-12186934

Lytic infection by polyomavirus leads to elevated levels of p53 and induction of p53 target genes p21Cip1/WAF1 (p21) and BAX. This is seen both in polyomavirus-infected primary mouse cell cultures and in kidney tissue of infected mice. Stabilization of p53 and induction of a p53 response are accompanied by phosphorylation of p53 on serine 18, mimicking a DNA damage response. Stabilization of p53 does not depend on p19Arf interaction with mdm2. Cells infected by a mutant virus defective in binding pRb and in inducing G(1)-to-S progression show a greatly diminished p53 response. However, cells infected by wild-type virus and blocked from entering S phase by addition of mimosine still show a p53 response. These results suggest a role of E2F target genes in inducing a p53 response. Polyomavirus large T antigen coprecipitates with p53 phosphorylated on serine 18 and also with p21Cip1/WAF1. Implications of these and other findings on possible mechanisms of induction and override of p53 functions during productive infection by polyomavirus are discussed.


Polyomavirus Infections/virology , Polyomavirus/pathogenicity , Tumor Suppressor Protein p53/metabolism , Tumor Virus Infections/virology , Animals , Animals, Newborn , Antigens, Polyomavirus Transforming/metabolism , Cell Line , DNA, Viral/metabolism , Kidney/virology , Mice , Phosphorylation , Tumor Suppressor Protein p53/genetics , Virulence
...