Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Cancer Gene Ther ; 19(7): 468-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539062

ABSTRACT

Rodent autonomous parvoviruses (PVs) are endowed with oncotropic properties and represent virotherapeutics with inherent oncolytic features. This work aimed to evaluate the capacity of Minute Virus of Mice (MVMp) to act as an adjuvant stimulating a mouse glioblastoma-specific immune response. MVMp was shown to induce cell death through apoptosis in glioma GL261 cells. Antigen-presenting cells (APCs) provide the initial cue for innate and adaptive immune responses, and thus MVMp-infected GL261 cells were tested for their ability to activate dendritic cells (DCs) and microglia (MG), two distinct cell types that are able to act as APCs. MG and discrete DC subsets were activated after co-culture with MVMp-infected glioma GL261 cells, as evidenced by upregulation of specific activation markers (CD80, CD86) and release of proinflammatory cytokines (tumor necrosis factor-α and interleukin-6). The in vivo analysis of immunodeficient and immunocompetent mice revealed a clear difference in their susceptibility to MVMp-mediated tumor suppression. Immunocompetent mice were fully protected from tumor outgrowth of GL261 cells infected ex vivo with MVMp. In contrast, immunodeficient animals were less competent for MVMp-dependent tumor inhibition, with only 20% of the recipients being protected, arguing for an additional immune component to allow full tumor suppression. In keeping with this conclusion, immunocompetent mice engrafted with MVMp-infected glioma cells developed a level of anti-tumor immunity with isolated splenocytes producing elevated levels of interferon-γ. In rechallenge experiments using uninfected GL261 cells, we could show complete protection against the tumor, arguing for the induction of a T-cell-mediated, tumor-specific, long-term memory response. These findings indicate that the anticancer effect of PVs can be traced back not only for their direct oncolytic effect, but also to their ability to break tumor tolerance.


Subject(s)
Glioma/immunology , Minute Virus of Mice/immunology , Oncolytic Viruses/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Glioma/pathology , Glioma/prevention & control , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Neoplasm Transplantation , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Cancer Gene Ther ; 18(4): 240-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21102423

ABSTRACT

Preclinical studies using various cell culture and animal systems highlight the potential of recombinant rodent parvoviruses (recPVs) for cancer therapy. Production of these viruses is, however, not efficient and this hampers the clinical applications of these agents. In this study, we show that the adenovirus genes E2a, E4(orf6) and VA RNA increase the production of recPVs by more than 10-fold and reduce the time of production from 3 to 2 days in HEK293T cells. The helper effects of these genes can be observed with different recPVs, regardless of the nature and size of the inserted transgene. Furthermore, we generated a recombinant Adenovirus 5 carrying the parvovirus VP transcription unit. This helper, named Ad-VP, allows recPVs to be efficiently produced through a protocol based only on cell infection, making possible to use cell lines, such as NB324K, which are good producers of parvoviruses but are hardly transfectable. Hence, we could further improve viral titers and reduce time and costs of production. This Ad-VP helper-based protocol could be scaled up to a bioreactor format for the generation of the large amounts of recPVs needed for future clinical applications.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Helper Viruses/genetics , Parvovirus/genetics , Adenoviridae/metabolism , Cells, Cultured , Genetic Vectors/metabolism , Humans , Parvovirus/metabolism , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly/genetics
3.
Acta Virol ; 53(1): 57-60, 2009.
Article in English | MEDLINE | ID: mdl-19301953

ABSTRACT

Standard therapies such as surgery and chemotherapy offer only minimal improvement in pancreatic cancer. However, the viruses killing cancer cells and substances like some antibiotics and phytoalexins with anticancer potential may represent a candidate non-conventional mean of cancer treatment in the future. In this study, the effect of infection with oncolytic H-1 parvovirus (H-1PV) combined with antibiotic norfloxacin (NFX) or phytoalexin resveratrol on the survival of cell lines Panc-1 and BxPC3 derived from human pancreatic carcinoma was tested. Whereas H-1PV with NFX exerted a synergistic effect, H-1PV with resveratrol resulted in an additive effect only. All the effects were partial, but they were more pronounced in Panc-1 compared to BxPC3 cells.


Subject(s)
Antineoplastic Agents/pharmacology , H-1 parvovirus/growth & development , Oncolytic Viruses/growth & development , Cell Line, Tumor , Cell Survival/drug effects , Drug Interactions , Humans , Norfloxacin/pharmacology , Resveratrol , Stilbenes/pharmacology
4.
Cancer Gene Ther ; 16(2): 149-60, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18670452

ABSTRACT

Interferon-gamma-inducible protein 10 is a potent chemoattractant for natural killer cells and activated T lymphocytes. It also displays angiostatic properties and some antitumor activity. Tumor necrosis factor-alpha (TNF-alpha) is a powerful immunomodulating cytokine with demonstrated tumoricidal activity in various tumor models and the ability to induce strong immune responses. This prompted us to evaluate the antitumor effects of recombinant parvoviruses designed to deliver IP-10 or TNF-alpha into a glioblastoma. When Gl261 murine glioma cells were infected in vitro with an IP-10- or TNF-alpha-transducing parvoviral vector and were subcutaneously implanted in mice, tumor growth was significantly delayed. Complete tumor regression was observed when the glioma cells were coinfected with both the vectors, demonstrating synergistic antitumor activity. In an established in vivo glioma model, however, repeated simultaneous peritumoral injection of the IP-10- and TNF-alpha-delivering parvoviruses failed to improve the therapeutic effect as compared with the use of a single cytokine-delivering vector. In this tumor model, cytokine-mediated immunostimulation, rather than inhibition of vascularization, is likely responsible for the therapeutic efficacy.


Subject(s)
Chemokine CXCL10/metabolism , Chemokine CXCL10/therapeutic use , Glioblastoma/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Chemokine CXCL10/administration & dosage , Chemokine CXCL10/immunology , Dendritic Cells/cytology , Dendritic Cells/virology , Drug Synergism , Female , Genetic Vectors , Glioblastoma/blood supply , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/virology , H-1 parvovirus/physiology , Humans , Immunocompetence , Mice , Mice, Inbred C57BL , Minute Virus of Mice/physiology , Necrosis/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics
5.
Gene Ther ; 15(10): 704-10, 2008 May.
Article in English | MEDLINE | ID: mdl-18356816

ABSTRACT

Autologous or allogenic tumour cells have long been used in the fight against cancer as vaccines to awaken the patient's immune system. On the other hand, oncolytic viruses have emerged in recent years as powerful therapeutic tools for selectively killing tumour cells. Yet despite recent improvements in virus production, administration and targeting, the latter strategy remains limited by poor access of oncolytic viruses to primary and metastatic tumour cells. The present review focuses on how to overcome these limitations on oncolytic virus delivery, at least in part, through the use of tumour-derived or in vitro transformed carrier cells. On the basis of existing evidence, novel strategies are proposed for using such cell vehicles, alone or in combination, both as virus factories and as anticancer vaccines.


Subject(s)
Cancer Vaccines/administration & dosage , Genetic Therapy/methods , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Cell Line, Transformed , Humans , Neoplasm Metastasis/therapy , Neoplasms/virology
6.
Cancer Gene Ther ; 15(4): 252-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18202715

ABSTRACT

Autonomous parvoviruses possess an intrinsic oncotropism based on viral genetic elements controlling gene expression and genome replication. We constructed a hybrid vector consisting of the H1 parvovirus-derived expression cassette comprising the p4 promoter, the ns1 gene and the p38 promoter flanked by the adeno-associated viruses 2 (AAV2) inverted terminal repeats and packaged into AAV2 capsids. Gene transduction using this vector could be stimulated by coinfection with adenovirus, by irradiation or treatment with genotoxic agents, similar to standard AAV2 vectors. However, the latter were in most cases less efficient in gene transduction than the hybrid vector. With the new vector, tumor cell-selective increase in transgene expression was observed in pairs of transformed and non-transformed cells, leading to selective killing of the transformed cells after expression of a prodrug-converting enzyme. Preferential gene expression in tumor versus normal liver tissue was also observed in vivo in a syngeneic rat model. Comparative transduction of a panel of different tumor cell lines with the H1 and the H1/AAV hybrid vector showed a preference of each vector for distinct cell types, probably reflecting the dependence of the viral tropism on capsid determinants.


Subject(s)
Genetic Vectors , Parvovirus/genetics , Transgenes , Animals , Blotting, Western , Cell Line, Transformed , HeLa Cells , Humans , Rats
7.
Article in English | MEDLINE | ID: mdl-16316394

ABSTRACT

Summary Rodent parvoviruses are promising candidates for oncolytic virotherapy of cancer in humans because of their oncotropism (preferential killing of transformed cells) in the absence of pathogenicity. Here, we give an overview concerning the possible application of parvovirus H-1 for cancer therapy, with specific emphasis on malignant brain tumours in humans.


Subject(s)
Antineoplastic Agents/therapeutic use , Parvovirus , Animals , Humans , Oncolytic Virotherapy , Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Rats , Rodent Diseases/virology
8.
J Virol ; 77(12): 6683-91, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12767988

ABSTRACT

The wnt signaling pathway is constitutively activated in colon tumors by mutations in the adenomatous polyposis coli and beta-catenin genes. We have modified the minute virus of mice (MVM) P4 promoter to make it responsive to wnt signaling by inserting binding sites for the heterodimeric beta-catenin/Tcf transcription factor. In luciferase assays we can see up to 20-fold selectivity of Tcf mutant P4 promoters for cells with activated wnt signaling. Hybrid MVM/H-1 viruses containing Tcf mutant promoters were tested for NS1 expression, viral DNA replication, virus replication, and cytopathic effect on colon, lung, kidney, and cervical cancer cell lines. Activation of the wnt pathway by expression of Delta N-beta-catenin increased NS1 expression and viral burst size in 293T and H1299 lung cancer cells, showing that the Tcf mutant P4 promoter can respond to wnt signals in the context of the virus. Compared to the parental virus, the burst size of the Tcf mutant viruses was reduced at least 1,000-fold in H1299, 293T, NB324K, and HeLa cells, which have inactive wnt signaling pathways. The burst size and cytopathic effect of the Tcf viruses was near wild-type levels in SW480 and Isreco1 colon cancer cell lines, which have high Tcf activity. The high specificity of these viruses should permit the development of H-1 virus-based vectors which combine high safety and greater efficacy in cancer therapy.


Subject(s)
Colonic Neoplasms , Gene Targeting/methods , Genetic Vectors , Parvovirus/physiology , Virus Replication , Zebrafish Proteins , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/virology , Animals , Binding Sites , Colonic Neoplasms/genetics , Colonic Neoplasms/virology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Mice , Minute Virus of Mice/genetics , Minute Virus of Mice/physiology , Mutation , Parvovirus/genetics , Parvovirus/pathogenicity , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Wnt Proteins , beta Catenin
9.
Gene Ther ; 9(5): 358-62, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11938456

ABSTRACT

Some of the therapeutic genes to be delivered by means of recombinant adenoviruses code for toxic compounds. Expression of these sequences can be deleterious to the complementation cells used for vector production, making it often difficult to generate high-titer stocks of toxin-transducing recombinant adenoviruses. In this work, we present a novel strategy for the transient post-transcriptional down-regulation of toxic transgene expression during the vector production phase, through the administration of phosphorothioate-modified antisense oligodeoxyribonucleotides. This method was successfully applied to the production of hybrid adenoviruses that contain the gene encoding the cytotoxic parvoviral protein NS1. The generation of recombinant adenoviruses in 293T cells was found to be fully suppressed as a result of adding of the NS gene to the vector genome. Yet, the production of NS-harboring hybrid adenoviruses could be rescued by treating the producer cells with antisense oligonucleotides specific for the translation initiation region of the NS transcript. This rescue correlated with a striking reduction of NS RNA and protein levels in the complementation cells. These data provide proof of principle of the suitability of the antisense oligonucleotides strategy for overcoming the interference of harmful transgenes with the production of adenoviral and other vectors.


Subject(s)
Gene Expression Regulation , Oligonucleotides, Antisense/genetics , T-Lymphocytes/metabolism , Viral Nonstructural Proteins/genetics , Adenoviridae/genetics , Cell Line , Genetic Engineering , Genetic Vectors , Humans , Parvovirus/genetics , Transfection , Transgenes
10.
J Gene Med ; 3(4): 326-37, 2001.
Article in English | MEDLINE | ID: mdl-11529662

ABSTRACT

BACKGROUND: The oncosuppressive properties of some autonomous parvoviruses such as H-1 virus, together with their low pathogenicity, make them attractive vectors for tumor-directed gene therapy. Indeed, it was recently shown that these viruses became endowed with an enhanced oncosuppressive activity after they had been engineered to deliver a recognized therapeutic transgene. This prompted us to use a parvoviral vector to analyse the antineoplastic capacity of MCP-3 (monocyte chemotactic protein-3), a CC chemokine which has a broad spectrum of target cells, and can thus be considered to be a promising candidate for cancer treatment. METHODS: We explored the use of a parvovirus H-1-based vector encoding human MCP-3 for its antitumor potential on human cervical carcinoma cells. HeLa cells were infected in vitro with the recombinant virus hH1/MCP-3 at a low multiplicity [1 replication unit (RU)/cell] and we investigated the effect of parvovirus-mediated MCP-3 transduction on tumor formation and growth upon implantation of HeLa cells in nude mice. RESULTS: Infection of HeLa cells with hH1/MCP-3 led to secretion of high levels of MCP-3 and to significant retardation of tumor growth in recipient mice, as compared with HeLa cells that were either buffer-treated or infected with a MCP-3-free vector. Tumors from hH1/MCP-3-infected HeLa cells were heavily infiltrated with activated macrophages and showed increased numbers of dendritic cells. In addition, activated natural killer (NK) cells were also recruited into MCP-3-transduced tumors. CONCLUSION: These observations indicate that parvovirus H-1-transduced MCP-3 is able to exert a significant antitumor activity which is mediated, at least in part, through macrophages and NK cells, under conditions in which activated T cells are lacking.


Subject(s)
Cytokines , Monocyte Chemoattractant Proteins/genetics , Parvovirus/genetics , Uterine Cervical Neoplasms/therapy , Animals , Chemokine CCL7 , Female , HeLa Cells , Humans , Mice , Mice, Nude , Monocyte Chemoattractant Proteins/pharmacokinetics , Monocyte Chemoattractant Proteins/therapeutic use , Plasmids , Recombinant Proteins/analysis , Transcription, Genetic , Transduction, Genetic , Transplantation, Heterologous
11.
J Virol ; 75(13): 5730-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390575

ABSTRACT

Minute virus of mice nonstructural protein NS1 is a multifunctional protein that is involved in many processes necessary for virus propagation. To perform its distinct activities in timely coordinated manner, NS1 was suggested to be regulated by posttranslational modifications, in particular phosphorylation. In fact, NS1 replicative functions are dependent on protein kinase C (PKC) phosphorylation, most likely due to alteration of the biochemical profile of the viral product as determined by comparing native NS1 with its dephosphorylated counterpart. Through the characterization of NS1 mutants at individual PKC consensus phosphorylation sites for their biochemical activities and nickase function, we were able to identify two target atypical PKC phosphorylation sites, T435 and S473, serving as regulatory elements for the initiation of viral DNA replication. Furthermore, by dissociating the energy-dependent helicase activity from the ATPase-independent trans esterification reaction using partially single-stranded substrates, we could demonstrate that atypical PKC regulation of NS1 nickase activity occurs at the level of origin unwinding prior to trans esterification.


Subject(s)
DNA Replication , Minute Virus of Mice/physiology , Protein Kinase C/metabolism , Viral Nonstructural Proteins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA/metabolism , Mice , Phosphorylation , Virus Replication
12.
Cancer Gene Ther ; 8(3): 158-67, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11332986

ABSTRACT

Autonomous parvoviruses preferentially replicate in and kill in vitro-transformed cells and reduce the incidence of spontaneous and implanted tumors in animals. Because of these natural oncotropic and oncolytic properties, parvoviruses deserve to be considered as potential antitumor vectors. Here, we assessed whether parvovirus H1 is able to kill human hepatoma cells by induction of apoptosis but spares primary human liver cells, and whether the former cells can efficiently be transduced by H1 virus-based vectors. Cell death, infectivity, and transgene transduction were investigated in Hep3B, HepG2, and Huh7 cells and in primary human hepatocytes with natural and recombinant H1 virus. All hepatoma cells were susceptible to H1 virus-induced cytolyis. Cell death correlated with H1 virus DNA replication, nonstructural protein expression, and with morphological features of apoptosis. H1 virus-induced apoptosis was more pronounced in p53-deleted Hep3B and p53-mutated Huh7 cells than in HepG2 cells which express wild-type p53. In Hep3B cells, apoptosis was partially inhibited by DEVD-CHO, a caspase-3 inhibitor. In contrast, H1 virus-infected primary hepatocytes were neither positive for nonstructural protein expression nor susceptible to H1 virus-induced killing. Infection with a recombinant parvovirus vector carrying the luciferase gene under control of parvovirus promoter P38 led to higher transgene activities in hepatoma cells than in the hepatocytes. Taken together, H1 virus kills human hepatoma cells at low virus multiplicity but not primary hepatocytes. Thus, recombinant H1 viruses carrying antitumor transgenes may be considered as potential therapeutic options for the treatment of hepatocellular carcinomas.


Subject(s)
Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Gene Transfer Techniques , Hepatocytes/pathology , Liver Neoplasms/genetics , Parvoviridae Infections/pathology , Parvovirus/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , DNA, Viral/biosynthesis , Genetic Vectors , Hepatocytes/cytology , Humans , Liver Neoplasms/pathology , Parvovirus/physiology , Transduction, Genetic , Tumor Cells, Cultured , Virus Replication
13.
J Virol ; 75(9): 4394-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11287588

ABSTRACT

Autonomous parvovirus minute virus of mice (MVM) DNA replication is strictly dependent on cellular factors expressed during the S phase of the cell cycle. Here we report that MVM DNA replication proceeds in specific nuclear structures termed autonomous parvovirus-associated replication bodies, where components of the basic cellular replication machinery accumulate. The presence of DNA polymerases alpha and delta in these bodies suggests that MVM utilizes partially preformed cellular replication complexes for its replication. The recruitment of cyclin A points to a role for this cell cycle factor in MVM DNA replication beyond its involvement in activating the conversion of virion single-stranded DNA to the duplex replicative form.


Subject(s)
CDC2-CDC28 Kinases , Cyclin A/metabolism , DNA Polymerase III/metabolism , DNA Polymerase I/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Minute Virus of Mice/physiology , Proliferating Cell Nuclear Antigen/metabolism , Virus Replication , Animals , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Humans , Mice , Minute Virus of Mice/metabolism , Protein Serine-Threonine Kinases/metabolism , Replication Protein A , Viral Nonstructural Proteins/metabolism
14.
Cancer Gene Ther ; 8(12): 958-65, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781658

ABSTRACT

The oncotropic and oncolytic behaviors of certain autonomous rodent parvoviruses make them promising vectors for anticancer gene therapies. However, these parvoviruses are often not potent enough to kill all tumor cells equally well. With the aim of enhancing the intrinsic antitumor effect and the range of natural parvoviruses, a recombinant H1 parvovirus vector was constructed that produces the Apoptin protein, a tumor cell-specific, p53-independent, Bcl-2-insensitive apoptotic effector. We compared the apoptotic activity exerted by a recombinant hH1/Apoptin virus with that of a Green Fluorescent Protein (GFP)-transducing recombinant virus, hH1/GFP, in three human tumor cell lines differing in their susceptibility to wild-type parvovirus H1-induced killing. We found that in cells that were rather resistant to the basal cytotoxic effect of wild-type H1 or the GFP recombinant virus, a parvovirus that expressed Apoptin caused a pronounced, additional cytotoxic effect. In contrast to its enhanced cytotoxicity toward tumor cells, hH1/Apoptin virus was not more toxic to normal human fibroblasts than was the wild-type H1 virus. Taken together, these data indicate that enhancing the oncotropic behavior of wild-type H1 parvoviruses with the tumor-specific apoptotic potency of Apoptin should lead to an effective replicative parvoviral vector.


Subject(s)
Apoptosis , Capsid Proteins , Capsid/genetics , Genetic Therapy , Genetic Vectors , Parvovirus/genetics , Apoptosis/genetics , Capsid/pharmacology , HeLa Cells , Humans , Reassortant Viruses/genetics , Virus Replication/genetics
15.
Virology ; 278(1): 151-67, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11112491

ABSTRACT

Minute virus of mice NS1, an 83-kDa mainly nuclear phosphoprotein, is the only viral nonstructural protein required in all cell types and it is involved in multiple processes necessary for virus propagation. The diversity of functions assigned to NS1, together with the variation of its complex phosphorylation pattern during infection, suggested that the various activities of NS1 could be regulated by distinct phosphorylation events. So far, it has been demonstrated that NS1 replicative functions, in particular, DNA-unwinding activities, are regulated by protein kinase C (PKC), as exemplified by the modulation of NS1 helicase activity by PKClambda phosphorylation. In order to determine further impact of phosphorylation on NS1 functions, including the induction of cytopathic effects, a mutational approach was pursued in order to produce NS1 variants harboring amino acid substitutions at candidate PKC target residues. Besides the determination of two additional in vivo phosphorylation sites in NS1, this mutagenesis allowed the segregation of distinct NS1 functions from one another, generating NS1 variants with a distinct activity profile. Thus, we obtained NS1 mutants that were fully proficient for trans activation of the viral P38 promoter, while being impaired in their replicative functions. Moreover, the alterations of specific PKC phosphorylation sites gave rise to NS1 polypeptides that exerted reduced cytotoxicity, leading to sustained gene expression, while keeping functions necessary for progeny virus production, i.e., viral DNA replication and activation of the capsid gene promoter. These data suggested that in the course of a viral infection, NS1 may undergo a shift from productive to cytotoxic functions as a result of a phosphorylation-dependent regulation.


Subject(s)
Cytopathogenic Effect, Viral , Minute Virus of Mice/physiology , Viral Nonstructural Proteins/physiology , Virus Replication , Animals , Binding Sites , Cell Line , DNA, Viral/biosynthesis , Mice , Minute Virus of Mice/chemistry , Mutagenesis, Site-Directed , Phosphorylation , Protein Kinase C , Viral Nonstructural Proteins/biosynthesis , Viral Nonstructural Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 97(10): 5522-7, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10792046

ABSTRACT

Replication of the single-stranded linear DNA genome of parvovirus minute virus of mice (MVM) starts with complementary strand synthesis from the 3'-terminal snap-back telomere, which serves as a primer for the formation of double-stranded replicative form (RF) DNA. This DNA elongation reaction, designated conversion, is exclusively dependent on cellular factors. In cell extracts, we found that complementary strand synthesis was inhibited by the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) and rescued by the addition of proliferating cell nuclear antigen, arguing for the involvement of DNA polymerase (Pol) delta in the conversion reaction. In vivo time course analyses using synchronized MVM-infected A9 cells allowed initial detection of MVM RF DNA at the G(1)/S phase transition, coinciding with the onset of cyclin A expression and cyclin A-associated kinase activity. Under in vitro conditions, formation of RF DNA was efficiently supported by A9 S cell extracts, but only marginally by G(1) cell extracts. Addition of recombinant cyclin A stimulated DNA conversion in G(1) cell extracts, and correlated with a concomitant increase in cyclin A-associated kinase activity. Conversely, a specific antibody neutralizing cyclin A-dependent kinase activity, abolished the capacity of S cell extracts for DNA conversion. We found no evidence for the involvement of cyclin E in the regulation of the conversion reaction. We conclude that cyclin A is necessary for activation of complementary strand synthesis, which we propose as a model reaction to study the cell cycle regulation of the Pol delta-dependent elongation machinery.


Subject(s)
Cell Cycle/physiology , Cyclin A/metabolism , DNA Polymerase III/metabolism , DNA Replication , Minute Virus of Mice/genetics , Animals , Cell Line , Cyclin A/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Cytosol/metabolism , Enzyme Inhibitors/metabolism , Fibroblasts , G1 Phase , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Kinetics , Mice , Minute Virus of Mice/enzymology , Recombinant Fusion Proteins/metabolism , S Phase , Spodoptera , Transfection , Virion/enzymology , Virion/genetics
17.
J Virol ; 74(10): 4807-15, 2000 May.
Article in English | MEDLINE | ID: mdl-10775619

ABSTRACT

We have identified a nuclear structure that is induced after infection with the autonomous parvovirus H-1. Using fluorescence microscopy, we observed that the major nonstructural protein (NS1) of H-1 virus which is essential for viral DNA amplification colocalized with virus-specific DNA sequences and sites of ongoing viral DNA replication in distinct nuclear bodies which we designated H-1 parvovirus-associated replication bodies (H-1 PAR-bodies). In addition, two cellular proteins were shown to accumulate in H1 PAR-bodies: (i) the proliferating cell nuclear antigen (PCNA) which is essential for chromosomal and parvoviral replication and (ii) the NS1-interacting small glutamine-rich TPR-containing protein (SGT), suggesting a role for the latter in parvoviral replication and/or gene expression. Since many DNA viruses target preexisting nuclear structures, known as PML-bodies, for viral replication and gene expression, we have determined the localization of H-1 PAR- and PML-bodies by double-fluorescence labeling and confocal microscopy and found them to be spatially unrelated. Furthermore, H-1 PAR-bodies did not colocalize with other prominent nuclear structures such as nucleoli, coiled bodies, and speckled domains. Electron microscopy analysis revealed that NS1, as detected by indirect immunogold labeling, was localized in ring-shaped electron-dense nuclear structures corresponding in size and frequency to H-1 PAR-bodies. These structures were also clearly visible without immunogold labeling and could be detected only in infected cells. Our results suggest that H-1 virus does not target known nuclear bodies for DNA replication but rather induces the formation of a novel structure in the nucleus of infected cells.


Subject(s)
Cell Nucleus/ultrastructure , Cell Nucleus/virology , Parvovirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Carrier Proteins , Cell Line , Cell Nucleus/metabolism , DNA Replication , Humans , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Microscopy, Electron , Molecular Chaperones , Parvoviridae Infections/virology , Parvovirus/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proteins/metabolism , Rats
18.
Hum Gene Ther ; 11(4): 597-609, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10724038

ABSTRACT

The possible use of recombinant autonomous parvoviruses as vectors to efficiently express therapeutic cytokines in human tumor cells was evaluated in vitro and in vivo. The parvovirus H1 was used to generate recombinant viruses (rH1) that carried transgenes encoding either human interleukin 2 (IL-2) or monocyte chemotactic protein 1 (MCP-1), in replacement of part of the capsid genes. Such rH11 viruses have been shown to retain in vitro the intrinsic oncotropic properties of the parental virus. On infection with the recombinant viruses at an input multiplicity of 1 replication unit (RU) per cell, HeLa cultures were induced to release 4-10 microg of cytokine per 10(6) cells over a period of 5 days. The expression of the rH1-transduced human cytokine/chemokine could also be detected in tumor material recovered from nude mice that had been subcutaneously engrafted with in vitro-infected HeLa cells. The formation of tumors from HeLa xenografts was reduced by 90% compared with wild-type or mock-infected cells as a result of cells preinfected with IL2-expressing virus at an input multiplicity as low as 1 RU per cell. Tumors arising from HeLa cells infected with transgene-free or MCP1-expressing vectors or with wild-type H1 virus were not rejected at this virus dose. Tumors infected with rH1/IL-2 virus displayed markers indicative of their infiltration with NK cells in which the cytocidal program was activated, whereas little NK activity was detected in wild-type virus or mock-infected tumors. Altogether, these data show that the IL-2 expressing H1 vector was a more potent antineoplastic agent than the parental virus, and point to the possible application of recombinant autonomous parvoviruses toward therapy of some human tumors.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , Genetic Vectors , Neoplasms, Experimental/prevention & control , Parvovirus/genetics , Transduction, Genetic , Animals , Base Sequence , Cell Line , Cytotoxicity, Immunologic/genetics , DNA Primers , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental/immunology , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transgenes , Viral Nonstructural Proteins/genetics
19.
Comp Med ; 50(6): 613-21, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11200567

ABSTRACT

To determine whether the multimammate mouse (Mastomys coucha) could be used to evaluate rodent parvovirus-based vectors, neonates were subcutaneously inoculated with minute virus of mice (prototype strain, MVMp) or rat parvovirus H-1. The course of infection with both viruses was similar. Seroconversion occurred within two weeks after virus inoculation, as detected by use of hemagglutination-inhibition assays, and antibody titers remained high for the entire observation period of 12 months. Viral DNA and infective virions were detected in several organs of inoculated animals prior to seroconversion, as measured by use of Southern blotting and plaque assays, respectively. Infective particles subsequently became undetectable, whereas viral DNA imprints persisted in distinct organs for at least nine months. Clinical signs of parvovirus infection appeared around six weeks after virus inoculation, and consisted of hemorrhages, stunted growth, and transient hair color changes. Sudden death occurred in a significant fraction of animals infected with MVMp, but not H-1 virus, at the time of weaning. Altogether, MVMp, which is innocuous to its natural host, the mouse, and H-1 virus, which is poorly pathogenic to the rat, appear to be pathogenic for Mastomys coucha.


Subject(s)
Minute Virus of Mice , Parvoviridae Infections/physiopathology , Parvovirus/isolation & purification , Animals , Animals, Laboratory , Animals, Newborn , Antibodies, Viral/blood , DNA Replication , DNA, Viral/isolation & purification , Death, Sudden , Female , Hair Color , Hemagglutination Inhibition Tests , Hemorrhage , Male , Minute Virus of Mice/isolation & purification , Minute Virus of Mice/physiology , Muridae , Parvoviridae Infections/pathology , Parvoviridae Infections/virology , Parvovirus/classification , Parvovirus/physiology , Rats , Virion/isolation & purification
20.
Virus Res ; 65(2): 161-74, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10581389

ABSTRACT

The autonomous parvovirus H-1 exerts tumor-suppressive effects in living organisms and has been shown to specifically interfere with the survival of transformed cells in culture. The mechanism(s) by which H-1 virus induces death of transformed cells is not yet well understood. It has recently been reported that H-1 virus induces apoptotic cell death in the human monocytic U937 cell line, as assessed by biochemical and morphological changes of infected cells (Rayet, B., Lopez-Guerrero, J.-A., Rommelaere, J., Dinsart, C., 1998. Induction of programmed cell death by parvovirus H-1 in U937 cells: connection with the TNFalpha signalling pathway. J. Virol. 72, 8893-8903). Here we show that parvovirus H-1 infection induced early biochemical changes pointing to apoptotic events also in the transformed human keratinocyte cell line, HeLa, and the transformed rat fibroblast cell line, P1. Morphologic changes, however, and in particular the early breakdown of plasma membrane integrity, suggested that apoptosis did not go to completion, leading to necrotic cell death as the major result of parvovirus infection of HeLa and P1 cells. Parvovirus infection of these, and to a significantly lesser extent of U937 cells, was accompanied by rapid depletion of intracellular NAD stores. Inhibition of NAD-consuming enzymes interfered with parvovirus-induced NAD depletion and increased the proportion of H-1 virus-infected cells displaying apoptotic features of cell death. In contrast, a similar prevention of NAD depletion through stimulation of NAD production had little influence on the cell death pathway, suggesting that NAD-consuming enzymes may promote necrosis in a direct way rather than through inducing the overall drop of intracellular NAD.


Subject(s)
Cell Death , NAD/metabolism , NAD/physiology , Parvovirus/genetics , Animals , Apoptosis , Cell Line , Cell Transformation, Viral , Fibroblasts , HeLa Cells , Humans , Keratinocytes/enzymology , Keratinocytes/pathology , Keratinocytes/virology , Necrosis , Parvoviridae Infections/metabolism , Parvoviridae Infections/pathology , Rats , U937 Cells/enzymology , U937 Cells/pathology , U937 Cells/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...