Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 383
Filter
1.
Gene Ther ; 18(11): 1098-102, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21544094

ABSTRACT

Oncolytic viruses have been combined with standard cancer therapies to increase therapeutic efficacy. Given the sequential activation of herpes viral genes (herpes simplex virus-1, HSV-1) and the temporal cellular changes induced by ionizing radiation, we hypothesized an optimal temporal sequence existed in combining oncolytic HSV-1 with ionizing radiation. Murine U-87 glioma xenografts were injected with luciferase encoding HSV-1, and ionizing radiation (IR) was given at times before or after viral injection. HSV-1 replication and tumor-volume response were followed. Radiation given 6-9 h after HSV-1 injection resulted in maximal viral luciferase expression and infectious viral production in tumor xenografts. The greatest xenograft regression was also seen with radiation given 6 h after viral injection. We then tested if HSV-1 replication had a dose response to ionizing radiation. HSV-1 luciferase expression exhibited a dose response as xenografts were irradiated from 0 to 5 Gy. There was no difference in viral luciferase expression as IR dose increased from 5 Gy up to 20 Gy. These results suggest that the interaction of IR with the HSV-1 lytic cycle can be manipulated for therapeutic gain by delivering IR at a specific time within viral replicative cycle.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Herpesvirus 1, Human/growth & development , Oncolytic Virotherapy/methods , Radiation, Ionizing , Virus Replication/radiation effects , Animals , Combined Modality Therapy , Dose-Response Relationship, Radiation , Herpesvirus 1, Human/radiation effects , Mice , Mice, Nude , Oncolytic Viruses/growth & development , Oncolytic Viruses/radiation effects , Virus Replication/genetics
2.
Gene Ther ; 13(7): 621-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16292350

ABSTRACT

Earlier this laboratory constructed a herpes simplex virus 1 recombinant (R5111) that carries a IL13 ligand inserted into glycoprotein D and can enter cells via the IL13Ralpha2 receptor commonly expressed on the surface of malignant glioma cells. In this report, we describe the properties of two recombinant viruses carrying chiemric gD genes. In R5181 recombinant virus the chimeric gene consisted on the residues 20-155 of urokinase plaminogen activator (uPA) inserted between residues 24 and 25 of gD. In R5182 the insert consisted of a 23-residue sequence encoding the uPA binding domain for the urokinase plaminogen activator receptor (uPAR). These viruses were constructed for three reasons, to increase the number of viruses that specifically target receptors on the surface of malignant glioma cells, to determine whether viruses exhibiting novel ligands could enter cells via receptors anchored to the cell surface via glycosylphosphatidylinositol anchor as has been recently demonstrated for nectin1, and to determine whether receptors other than IL13Ralpha2 could be targeted by genetic engineering of the virus. We report that R5181 but not R5182 recombinant virus was able to enter cells expressing uPAR. The results indicate that HSV-1 recombinant viruses can be engineered to enter cells via a variety of unrelated nonviral receptors, including receptors that are anchored to the cells surface but without transmembrane domains.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioma/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy/methods , Amino Acid Sequence , Binding Sites , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Cell Line , Genetic Engineering , Genetic Vectors/metabolism , Glioma/metabolism , Glioma/virology , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Immunoblotting , Microscopy, Fluorescence , Molecular Sequence Data , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Urokinase-Type Plasminogen Activator/genetics , Vaccines, Synthetic/therapeutic use , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Vaccines/therapeutic use , Virion , Virus Replication
3.
Biochem Soc Trans ; 32(Pt 5): 697-701, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15493991

ABSTRACT

In HSV-1 (herpes simplex virus 1)-infected cells, the U(L)41 gene product carried with the virion has been shown to mediate the degradation of mRNA, leading to the shut-off of cellular protein synthesis. Analysis of the RNAs accumulating in cells infected with HSV-1 revealed the accumulation of RNAs encoding numerous cellular proteins both associated with and independent of activation of the NF-kappaB (nuclear factor kappaB) pathway. Studies on the activation of NF-kappaB and the expression and fate of selected cellular transcripts revealed the following. (i) In HSV-1-infected cells, NF-kappaB is activated by activated protein kinase R. Furthermore, the blockade of NF-kappaB translocation by suppression of protein kinase R activation does not render the cell more susceptible to apoptosis induced by viral gene expression. (ii) A number of mRNA up-regulated in infected cells [e.g. IkappaBalpha (inhibitory kappaBalpha), the immediate-early response protein IEX-1 and c-fos] are partially degraded and not translated. The degradation is U(L)41-dependent and results in deadenylation, endonucleolytic cleavage and 3'-5' degradation. The 5'-portion resulting from the endonucleolytic cleavage tends to linger in the infected cells. To date, the RNAs processed in this manner contained ARE (AU-rich elements) in their 3'-untranslated domains. RNAs lacking ARE were expressed and not degraded in this manner. (iii) Tristetraprolin and T-cell internal antigen-1, cellular proteins involved in the degradation of ARE-containing RNAs, are induced and activated in infected cells and tristetraprolin interacts physically with the U(L)41 protein.


Subject(s)
Fibroblasts/virology , Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , RNA/chemistry , Simplexvirus/metabolism , 3' Untranslated Regions , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Viral , Genes, Immediate-Early , Humans , I-kappa B Proteins/metabolism , Immediate-Early Proteins/metabolism , Models, Biological , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Poly(A)-Binding Proteins , Protein Transport , RNA/metabolism , RNA, Messenger/metabolism , T-Cell Intracellular Antigen-1 , Tristetraprolin , Up-Regulation
4.
Clin Microbiol Infect ; 8(9): 551-63, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12427216

ABSTRACT

Herpes simplex virus-1 (HSV-1) is a relatively large double-stranded DNA virus encoding at least 89 proteins with well characterized disease pathology. An understanding of the functions of viral proteins together with the ability to genetically engineer specific viral mutants has led to the development of attenuated HSV-1 for gene therapy. This review highlights the progress in creating attenuated genetically engineered HSV-1 mutants that are either replication competent (viral non-essential gene deleted) or replication defective (viral essential gene deleted). The choice between a replication-competent or -defective virus is based on the end-goal of the therapeutic intervention. Replication-competent HSV-1 mutants have primarily been employed as antitumor oncolytic viruses, with the lytic nature of the virus harnessed to destroy tumor cells selectively. In replacement gene therapy, replication-defective viruses have been utilized as delivery vectors. The advantages of HSV-1 vectors are that they infect quiescent and dividing cells efficiently and can encode for relatively large transgenes.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Neoplasms/therapy , Simplexvirus/genetics , Antineoplastic Agents/therapeutic use , Clinical Trials, Phase I as Topic , Combined Modality Therapy , Defective Viruses/genetics , Gene Transfer Techniques , Humans , Neoplasms/drug therapy , Neoplasms/radiotherapy , Simplexvirus/enzymology , Transcription, Genetic , Virus Replication
5.
Gene Ther ; 9(1): 75-80, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11850725

ABSTRACT

The herpes simplex virus (HSV) recombinant virus R7020 is an attenuated virus designed as a candidate for immunization against both HSV-1 and HSV-2 infections. It was extensively tested in an experimental animal system and in a healthy human adult population without significant untoward effects. We report on the use of R7020 with ionizing radiation as an oncolytic agent for hepatomas. Two hepatoma cell lines were studied, Hep3B and Huh7. R7020 replicated to higher titers in Hep3B cells than in Huh7 cells. Tissue culture studies correlated with hepatoma xenograft responses to R7020. R7020 was more effective in mediating Hep3B tumor xenograft regression compared with Huh7. Ionizing radiation combined with R7020 also showed differential results in antitumor efficacy between the two cell lines in tumor xenografts. Ionizing radiation enhanced the replication of R7020 in Hep3B xenografts. Moreover, the combination of ionizing radiation and virus caused a greater regression of xenograft volume than either R7020 or radiation alone. Ionizing radiation had no effect on the replication of R7020 virus in Huh7 xenografts. These results indicate that a regimen involving infection with an appropriate herpesvirus such as R7020 in combination with ionizing radiation can be highly effective in eradicating certain tumor xenografts.


Subject(s)
Genetic Therapy/methods , Herpes Simplex Virus Vaccines/administration & dosage , Herpesvirus 1, Human , Herpesvirus 2, Human , Liver Neoplasms, Experimental/therapy , Animals , Combined Modality Therapy , Humans , Liver Neoplasms, Experimental/radiotherapy , Mice , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured , Virus Replication/radiation effects
6.
J Virol ; 75(21): 10326-33, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581401

ABSTRACT

Earlier studies have shown that cdc2 kinase is activated during herpes simplex virus 1 infection and that its activity is enhanced late in infection even though the levels of cyclin A and B are decreased below levels of detection. Furthermore, activation of cdc2 requires the presence of infected cell protein no. 22 and the U(L)13 protein kinase, the same gene products required for optimal expression of a subset of late genes exemplified by U(S)11, U(L)38, and U(L)41. The possibility that the activation of cdc2 and expression of this subset may be connected emerged from the observation that dominant negative cdc2 specifically blocked the expression of U(S)11 protein in cells infected and expressing dominant negative cdc2. Here we report that in the course of searching for a putative cognate partner for cdc2 that may have replaced cyclins A and B, we noted that the DNA polymerase processivity factor encoded by the U(L)42 gene contains a degenerate cyclin box and has been reported to be structurally related to proliferating cell nuclear antigen, which also binds cdk2. Consistent with this finding, we report that (i) U(L)42 is able to physically interact with cdc2 at both the amino-terminal and carboxyl-terminal domains, (ii) the carboxyl-terminal domain of U(L)42 can be phosphorylated by cdc2, (iii) immunoprecipitates obtained with anti U(L)42 antibody contained a roscovitine-sensitive kinase activity, (iv) kinase activity associated with U(L)42 could be immunodepleted by antibody to cdc2, and (v) U(L)42 transfected into cells associates with a nocodazole-enhanced kinase. We conclude that U(L)42 can associate with cdc2 and that the kinase activity has the characteristic traits of cdc2 kinase.


Subject(s)
CDC2 Protein Kinase/metabolism , DNA-Directed DNA Polymerase , Exodeoxyribonucleases , Viral Proteins/metabolism , Amino Acid Sequence , Autoradiography , Cell Line , DNA/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Protein Kinases/metabolism , Viral Proteins/chemistry
7.
Proc Natl Acad Sci U S A ; 98(22): 12665-70, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11675498

ABSTRACT

U87 cells derived from human malignant gliomas and growtharrested human embryonic lung (HEL) fibroblasts were examined with respect to their response to ionizing radiation by profiling their RNAs. In the first series of experiments, cells grown in vitro were harvested and the RNAs were extracted 5 h after exposure to 1, 3, or 10 Gy. In the second series of experiments the U87 tumors were implanted in nude mice and subjected to the same doses of irradiation. The xenografts were harvested at 1, 5, or 24 h after irradiation and subjected to the same analyses. We observed and report on (i) cell-type common and cell-type specific responses, (ii) genes induced at low levels of irradiation but not at higher doses, (iii) temporal patterns of gene response in U87 xenografts that varied depending on radiation dose and temporal patterns of response that were similar at all doses tested, (iv) significantly higher up-regulation of cells in xenografts than in in vitro cultures, and (v) genes highly up-regulated by radiation. The responding genes could be grouped into nine functional clusters. The representation of the nine clusters was to some extent dependent on dose and time after irradiation. The results suggest that clinical outcome of ionizing radiation treatment may benefit significantly by taking into account both cell-type and radiation-dose specificity of cellular responses.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation/radiation effects , Neoplasms, Experimental/radiotherapy , Oligonucleotide Array Sequence Analysis , Animals , DNA-Activated Protein Kinase , Dose-Response Relationship, Radiation , Fibroblasts/radiation effects , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Nuclear Proteins , Phosphoprotein Phosphatases/genetics , Protein Serine-Threonine Kinases/genetics , RNA Splicing , Transplantation, Heterologous , Tumor Cells, Cultured
8.
J Virol ; 75(17): 7904-12, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11483735

ABSTRACT

The herpes simplex virus 1 (HSV-1) infected cell proteins 0 and 4 (ICP0 and ICP4) are multifunctional proteins extensively posttranscriptionally processed by both cellular and viral enzymes. We examined by two-dimensional separations the posttranslational forms of ICP0 and ICP4 in HEp-2 cells and in human embryonic lung (HEL) fibroblasts infected with wild-type virus, mutant R325, lacking the sequences encoding the U(S)1.5 protein and the overlapping carboxyl-terminal domain of ICP22, or R7914, in which the aspartic acid 199 of ICP0 was replaced by alanine. We report the following (i) Both ICP0 and ICP4 were sequentially posttranslationally modified at least until 12 h after infection. In HEL fibroblasts, the processing of ICP0 shifted from A+B forms at 4 h to D+G forms at 8 h and finally to G, E, and F forms at 12 h. The ICP4 progression was from the A' form noted at 2 h to B' and C' forms noted at 4 h to the additional D' and E' forms noted at 12 h. The progression tended to be toward more highly charged forms of the proteins. (ii) Although the overall patterns were similar, the mobility of proteins made in HEp-2 cells differed from those made in HEL fibroblasts. (iii) The processing of ICP0 forms E and F was blocked in HEL fibroblasts infected with R325 or with wild-type virus and treated with roscovitine, a specific inhibitor of cell cycle-dependent kinases cdc2, cdk2, and cdk5. R325-infected HEp-2 cells lacked the D' form of ICP4, and roscovitine blocked the appearance of the most highly charged E' form of ICP4. (iv) A characteristic of ICP0 is that it is translocated into the cytoplasm of HEL fibroblasts between 5 and 9 h after infection. Addition of MG132 to the cultures late in infection resulted in rapid relocation of cytoplasmic ICP0 back into the nucleus. Exposure of HEL fibroblasts to MG132 late in infection resulted in the disappearance of the highly charged ICP0 G isoform. The G form of ICP0 was also absent in cells infected with R7914 mutant. In cells infected with this mutant, ICP0 is not translocated to the cytoplasm. (v) Last, cdc2 was active in infected cells, and this activity was inhibited by roscovitine. In contrast, the activity of cdk2 exhibited by immunoprecipitated protein was reduced and resistant to roscovitine and may represent a contaminating kinase activity. We conclude from these results that the ICP0 G isoform is the cytoplasmic form, that it may be phosphorylated by cdc2, consistent with evidence published earlier (S. J., Advani, R. R. Weichselbaum, and B. Roizman, Proc. Natl. Acad. Sci. USA 96:10996-11001, 2000), and that the processing is reversed upon relocation of the G isoform from the cytoplasm into the nucleus. The processing of ICP4 is also affected by R325 and roscovitine. The latter result suggests that ICP4 may also be a substrate of cdc2 late in infection. Last, additional modifications are superimposed by cell-type-specific enzymes.


Subject(s)
CDC2-CDC28 Kinases , Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Protein Processing, Post-Translational , Subcellular Fractions/metabolism , CDC2 Protein Kinase/metabolism , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Humans , Immediate-Early Proteins/genetics , Leupeptins/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Purines/pharmacology , Roscovitine , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
9.
J Virol ; 75(17): 8105-16, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11483756

ABSTRACT

Following the lead of recent studies on the presence of RNA in virions of human cytomegalovirus, we investigated the presence and identity of RNAs from purified virions of herpes simple virus 1. To facilitate these studies, we designed primers for all known open reading frames (ORFs) and also constructed cDNA arrays containing probes designed to detect all known transcripts. In the first series of experiments, labeled DNA made by reverse transcription of poly(A)(+) RNA extracted from infected HEp-2 or rabbit skin cells hybridized to all but two of the probes in the cDNA array. A similar analysis of the RNA extracted from purified extracellular virions derived from infected HEp-2 cells hybridized to probes representing 24 of the ORFs. In the second series of analyses, we reverse transcribed and amplified by PCR RNAs from purified intracellular or extracellular virions derived from infected HEp-2 or Vero cell lines. The positive RNAs were retested by PCR with and without prior reverse transcription to ensure that the samples tested were free of contaminating DNA. The results were as follows. (i) Only a fraction of viral ORF transcripts were represented in virion RNA, and only nine RNAs (U(L)10, U(L)34/U(L)35, U(L)36, U(L)42, U(L)48, U(L)51, U(S)1/U(S)1.5, U(S)8.5, and U(S)10/U(S)11) were positive in all RT PCR assays. Of these, seven were positive by hybridization to cDNA arrays. (ii) RNA extracted from cells infected with a mutant virus lacking the U(S)8 to U(S)12 genes yielded results similar to those described above, indicating that U(S)11, a known RNA binding protein, does not play a role in packaging RNA in virions. (iii) Cellular RNAs detected in virions were representative of the abundant cellular RNAs. Last, RNA extracted from virions was translated in vitro and the translation products were reacted with antibody to alphaTIF (VIP16). The immune precipitate contained a labeled protein with the apparent molecular weight of alphaTIF, indicating that at least one mRNA packaged in virions was intact and capable of being translated. The basis for the apparent selectivity in the packaging of the viral RNAs packaged in virions is unknown.


Subject(s)
Herpesvirus 1, Human/genetics , RNA, Viral/isolation & purification , Virion/genetics , Virion/metabolism , Animals , Cell Line , DNA Primers , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Open Reading Frames/genetics , Protein Biosynthesis , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly
10.
Proc Natl Acad Sci U S A ; 98(18): 10410-5, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517326

ABSTRACT

Earlier studies have shown that the d120 mutant of herpes simplex virus 1, which lacks both copies of the alpha4 gene, induces apoptosis in all cell lines tested. In some cell lines d120-induced apoptosis, manifested by the release of cytochrome c, activation of caspase 3, and fragmentation of cellular DNA, is blocked by the overexpression of Bcl-2. In these cells viral protein kinase U(S)3 delivered in trans blocks apoptosis induced by the mutant virus at a premitochondrial stage. We report that the U(S)3 protein kinase targets the pro-apoptotic BAD member of the Bcl-2 family. Specifically, the U(S)3 protein kinase mediates a posttranslational modification of BAD and blocks its cleavage, which is reported to activate apoptosis. Thus, U(S)3 protein kinase is the sole viral protein required to block activation of caspase 3, prevent cleavage of poly(ADP-ribose) polymerase, and block fragmentation of cellular DNA induced by BAD.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Herpesvirus 1, Human/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Baculoviridae/genetics , Caspase 3 , Caspases/metabolism , Cells, Cultured , DNA Fragmentation , Herpesvirus 1, Human/genetics , Mutation , Peptide Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Rabbits , Viral Proteins/genetics , bcl-Associated Death Protein
11.
Proc Natl Acad Sci U S A ; 98(15): 8815-20, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11447293

ABSTRACT

The infected cell protein 0 (ICP0) of herpes simplex virus 1, a promiscuous transactivator shown to enhance the expression of genes introduced into cells by infection or transfection, interacts with numerous cellular proteins and has been linked to the disruption of ND10 and degradation of several proteins. ICP0 contains a RING finger domain characteristic of a class of E3 ubiquitin ligases. We report that: (i) in infected cells, ICP0 interacts dynamically with proteasomes and is bound to proteasomes in the presence of the proteasome inhibitor MG132. Also in infected cells, cdc34, a polyubiquitinated E2 ubiquitin-conjugating enzyme, exhibits increased ICP0-dependent dynamic interaction with proteasomes. (ii) In an in vitro substrate-independent ubiquitination system, the RING finger domain encoded by exon 2 of ICP0 binds cdc34, whereas the carboxyl-terminal domain of ICP0 functions as an E3 ligase independent of the RING finger domain. The results indicate that ICP0 can act as a unimolecular E3 ubiquitin ligase and that it promotes ubiquitin-protein ligation and binds the E2 cdc34. It differs from other unimolecular E3 ligases in that the domain containing the RING finger binds E2, whereas the ligase activity maps to a different domain of the protein. The results also suggest that ICP0 shuttles between nucleus and cytoplasm as a function of its dynamic interactions with proteasomes.


Subject(s)
Cysteine Endopeptidases/metabolism , Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Ligases/metabolism , Multienzyme Complexes/metabolism , Ubiquitin-Protein Ligase Complexes , Anaphase-Promoting Complex-Cyclosome , Cell Line , Enzyme Activation , Humans , Multienzyme Complexes/antagonists & inhibitors , Proteasome Endopeptidase Complex , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
12.
J Virol ; 75(13): 6166-72, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390618

ABSTRACT

An earlier report showed that viruses lacking the open reading frames encoding glycoproteins J and D but containing the glycoprotein D in their envelopes (gD-/+ stocks) and viruses lacking both the open reading frames and the glycoproteins in their envelopes (gD-/- stocks) induce apoptosis (G. Zhou, V. Galvan, G. Campadelli-Fiume, and B. Roizman, J. Virol. 74:11782-11791, 2000). Furthermore, apoptosis was blocked by delivery in trans of genes expressing glycoprotein D or J. Whereas gD-/- stocks attach but cannot initiate productive infection, gD-/+ stocks infect cells and produce gD-/- progeny virus. The difference in the infectivity of these two stocks suggested the possibility that the requirements for blocking apoptosis may be different. To test this hypothesis, we cloned into baculoviruses the entire wild-type glycoprotein D (Bac-gD-WT), the ectodomain only (Bac-gD-A), the ectodomain and the transmembrane domain (Bac-gD-B), the ectodomain and the cytoplasmic domain without the transmembrane domain (Bac-gD-C), or the transmembrane domain and the carboxyl-terminal cytoplasmic domain (Bac-gD-D). We report the following. Apoptosis induced by gD-/+ stocks was blocked by delivery in trans of recombinant baculovirus Bac-gD-WT, Bac-gD-A, Bac-gD-B, or Bac-gD-C but not of Bac-gD. Apoptosis induced by gD-/- stocks was blocked by Bac-gD-WT or by a mixture of Bac-gD-B and Bac-gD-D but not by any baculoviruses expressing truncated glycoprotein D alone or by the mixture of Bac-gD-A and Bac-gD-D. We conclude that the requirements to block apoptosis induced by the two virus stocks are different. The gD ectodomain is sufficient to block apoptosis induced by gD, whereas both the ectodomain and the cytoplasmic domain are required to block apoptosis induced by gD-/- stocks. The results indicate that in the case of gD-/- stocks, the transmembrane domain is required either to deliver the ectodomain to the appropriate intracellular compartment or to form multimeric constructs which virtually reconstitute gD through the interaction of transmembrane domains.


Subject(s)
Apoptosis , Genome, Viral , Herpesvirus 1, Human/genetics , Viral Envelope Proteins/physiology , Virion/genetics , Animals , Cell Line , Recombinant Proteins/biosynthesis , Spodoptera , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
13.
Gene Ther ; 8(10): 769-77, 2001 May.
Article in English | MEDLINE | ID: mdl-11420640

ABSTRACT

We have used interleukin (IL)-4 and -10-producing HSV-1 gamma(1)34.5 deletion viruses in gene therapy of a BALB/c model of experimental allergic encephalomyelitis (EAE), a T cell-mediated demyelinating disease of the central nervous system. It is known that in EAE of mice the Th2-type cytokines are down-regulated and the Th1-type cytokines up-regulated during the onset and relapse of the disease. Therefore, we tested two HSV-1 recombinants expressing the Th2-type cytokines IL-4 and IL-10. The recombinant viruses were injected intracranially (i.c.) in BALB/c mice 6 days after induction of EAE. As control groups we used mice without any infection, mice infected with backbone virus R3659 and mock-infected mice. Weights and symptoms of the mice were recorded daily and the tissue specimens were collected at specific time-points. The results indicate that the intracranial infection with IL-4-producing virus (1) precludes EAE symptoms, (2) protects the spinal cord from massive leukocyte infiltrations and (3) prevents demyelination and axonal loss. The IL-10-expressing virus R8308 did not have a similar favorable effect on the recovery of the mice as did the IL-4 virus R8306.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , Interleukin-10/genetics , Interleukin-4/genetics , Animals , Brain/virology , Female , Gene Expression , Immunohistochemistry , Mice , Mice, Inbred BALB C , Spinal Cord/virology , Statistics, Nonparametric
14.
Lancet ; 357(9267): 1513-8, 2001 May 12.
Article in English | MEDLINE | ID: mdl-11377626

ABSTRACT

Herpes simplex virus (HSV) is a member of the herpesviridae family. Recognised since ancient Greek times, the virus frequently infects human beings, causing a range of diseases from mild uncomplicated mucocutaneous infection to those that are life threatening. In the past 50 years, substantial advances in our knowledge of the molecular biology of HSV have led to insights into disease pathogenesis and management. This review provides a contemporary interpretation of the biological properties, function, epidemiology, and treatment of HSV diseases.


Subject(s)
Herpes Simplex/diagnosis , Antiviral Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Herpes Simplex/drug therapy , Herpes Simplex/virology , Humans , Recurrence , Simplexvirus/pathogenicity , Virus Replication
15.
J Virol ; 75(12): 5491-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356956

ABSTRACT

Earlier studies have shown that the d120 mutant of herpes simplex virus 1, which lacks both copies of the alpha4 gene, induces caspase-3-dependent apoptosis in HEp-2 cells. Apoptosis was also induced by the alpha4 rescuant but was blocked by the complementation of rescuant with a DNA fragment encoding the U(S)3 protein kinase (R. Leopardi and B. Roizman, Proc. Natl. Acad. Sci. USA 93:9583-9587, 1996, and R. Leopardi, C. Van Sant, and B. Roizman, Proc. Natl. Acad. Sci. USA 94:7891-7896, 1997). To investigate its role in the apoptotic cascade, the U(S)3 open reading frame was cloned into a baculovirus (Bac-U(S)3) under the control of the human cytomegalovirus immediate-early promoter. We report the following. (i) Bac-U(S)3 blocks processing of procaspase-3 to active caspase. Procaspase-3 levels remained unaltered if superinfected with Bac-U(S)3 at 3 h after d120 mutant infection, but significant amounts of procaspase-3 remained in cells superinfected with Bac-Us3 at 9 h postinfection with d120 mutant. (ii) The U(S)3 protein kinase blocks the proapoptotic cascade upstream of mitochondrial involvement inasmuch as Bac-U(S)3 blocks release of cytochrome c in cells infected with the d120 mutant. (iii) Concurrent infection of HEp-2 cells with Bac-U(S)3 and the d120 mutant did not alter the pattern of accumulation or processing of ICP0, -22, or -27, and therefore U(S)3 does not appear to block apoptosis by targeting these proteins.


Subject(s)
Apoptosis/drug effects , Herpesvirus 1, Human/pathogenicity , Immediate-Early Proteins/genetics , Mitochondria/physiology , Protein Serine-Threonine Kinases/pharmacology , Baculoviridae/genetics , Baculoviridae/metabolism , Caspase 3 , Caspases/metabolism , Cell Line , Enzyme Precursors/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/metabolism , Mutation , Peptide Hydrolases/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism
16.
J Virol ; 75(8): 3832-40, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264372

ABSTRACT

Earlier studies have shown that wild-type infected-cell protein 0 (ICP0), a key herpes simplex virus regulatory protein, translocates from the nucleus to the cytoplasm of human embryonic lung (HEL) fibroblasts within several hours after infection (Y. Kawaguchi, R. Bruni, and B. Roizman, J. Virol. 71:1019-1024, 1997). Translocation of ICP0 was also observed in cells infected with the d120 mutant, in which both copies of the gene encoding ICP4, the major regulatory protein, had been deleted (V. Galvan, R. Brandimarti, J. Munger, and B. Roizman, J. Virol. 74:1931-1938, 2000). Furthermore, a mutant (R7914) carrying the D199A substitution in ICP0 does not bind or stabilize cyclin D3 and is retained in the nucleus (C. Van Sant, P. Lopez, S. J. Advani, and B. Roizman, J. Virol. 75:1888-1898, 2001). Studies designed to elucidate the requirements for the translocation of ICP0 between cellular compartments revealed the following. (i) Translocation of ICP0 to the cytoplasm in productive infection maps to the D199 amino acid, inasmuch as wild-type ICP0 delivered in trans to cells infected with an ICP0 null mutant was translocated to the cytoplasm whereas the D199A-substituted mutant ICP0 was not. (ii) Translocation of wild-type ICP0 requires a function expressed late in infection, inasmuch as phosphonoacetate blocked the translocation of ICP0 in wild-type virus-infected cells but not in d120 mutant-infected cells. Moreover, whereas in d120 mutant-infected cells ICP0 was translocated rapidly from the cytoplasm to the nucleus at approximately 5 h after infection, the translocation of ICP0 in wild-type virus-infected cells extended from 5 to at least 9 h after infection. (iii) In wild-type virus-infected cells, the MG132 proteasomal inhibitor blocked the translocation of ICP0 to the cytoplasm early in infection, but when added late in infection, it caused ICP0 to be relocated back to the nucleus from the cytoplasm. (iv) MG132 blocked the translocation of ICP0 in d120 mutant-infected cells early in infection but had no effect on the ICP0 aggregated in vesicle-like structures late in infection. However, in d120 mutant-infected cells treated with MG132 at late times, proteasomes formed a shell-like structure around the aggregated ICP0. These structures were not seen in wild-type virus or R7914 mutant-infected cells. The results indicate the following. (i) In the absence of beta or gamma protein synthesis, ICP0 dynamically associates with proteasomes and is translocated to the cytoplasm. (ii) In cells productively infected beyond alpha gene expression, ICP0 is retained in the nucleus until after the onset of viral DNA synthesis and the synthesis of gamma2 proteins. (iii) Late in infection, ICP0 is actively sequestered in the cytoplasm by a process mediated by proteasomes, inasmuch as interference with proteasomal function causes rapid relocation of ICP0 to the nucleus.


Subject(s)
Cytoplasm/metabolism , Herpesvirus 1, Human , Immediate-Early Proteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Substitution/genetics , Cell Line , Cysteine Endopeptidases/metabolism , DNA, Viral/biosynthesis , Fibroblasts , Fluorescent Antibody Technique , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Immediate-Early Proteins/genetics , Leupeptins/pharmacology , Lung/embryology , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Mutation/genetics , Proteasome Endopeptidase Complex , Protein Binding , Time Factors , Ubiquitin-Protein Ligases
17.
Proc Natl Acad Sci U S A ; 98(4): 1877-82, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172044

ABSTRACT

The infected cell protein no. 0 (ICP0) of herpes simplex virus 1 (HSV-1) is a promiscuous transactivator shown to enhance the expression of gene introduced into cells by infection or transfection. At the molecular level, ICP0 is a 775-aa ring finger protein localized initially in the nucleus and late in infection in the cytoplasm and mediates the degradation of several proteins and stabilization of others. None of the known functions at the molecular level account for the apparent activity of ICP0 as a transactivator. Here we report that ICP0 functionally interacts with cellular transcription factor BMAL1, a member of the basic helix-loop-helix PER-ARNT-SIM (PAS) super family of transcriptional regulators. Specifically, sequences mapped to the exon II of ICP0 interacted with BMAL1 in the yeast two-hybrid system and in reciprocal pull-down experiments in vitro. Moreover, the enhancement of transcription of a luciferase reporter construct whose promoter contained multiple BMAL1-binding sites by ICP0 and BMAL1 was significantly greater than that observed by ICP0 or BMAL1 alone. Although the level of BMAL1 present in nuclei of infected cells remained unchanged between 3 and 8 h after infection, the level of cytoplasmic BMAL1 was reduced at 8 h after infection. The reduction of cytoplasmic BMAL1 was significantly greater in cells infected with the ICP0-null mutant than in the wild-type virus-infected cells, suggesting that ICP0 mediates partial stabilization of the protein. These results indicate that ICP0 interacts physically and functionally with at least one cellular transcription-regulatory factor.


Subject(s)
Helix-Loop-Helix Motifs , Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Transcription Factors/metabolism , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors , COS Cells , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , Glutathione Transferase/genetics , Herpesvirus 1, Human/physiology , Humans , Immediate-Early Proteins/genetics , Rabbits , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases , Vero Cells
18.
J Virol ; 75(4): 1888-98, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160688

ABSTRACT

Earlier reports from this laboratory have shown that the promiscuous transactivator infected-cell protein 0 (ICP0) binds and stabilizes cyclin D3, that the binding site maps to aspartic acid 199 (D199), and that replacement of D199 with alanine abolishes binding and reduces the capacity of the mutant virus to replicate in quiescent cells or to cause mortality in mice infected by a peripheral site. The objective of this report was to investigate the role of cyclin D3 in the biology of ICP0. We report the following results. (i) Wild-type ICP0 activates cyclin D-dependent kinase 4 (cdk4) and stabilizes cyclin D1 although ICP0 does not interact with this cyclin. (ii) The D199A mutant virus (R7914) does not activate cdk4 or stabilize cyclin D1, and neither the wild-type nor the mutant virus activates cdk2. (iii) Early in infection of human embryonic lung (HEL) fibroblasts both wild-type and D199A mutant ICP0s colocalize with PML, and in these cells the ND10 nuclear structures are dispersed. Whereas wild-type ICP0 is transported to the cytoplasm between 3 and 9 h. after infection, ICPO containing the D199A substitution remains quantitatively in the nucleus. (iv) To examine the interaction of ICP0 with cyclin D3, we used a previously described mutant carrying a wild-type ICP0 but expressing cyclin D3 (R7801) and in addition constructed a virus (R7916) that was identical except that it carried the D199A-substituted ICP0. Early in infection with R7801, ICP0 colocalized with cyclin D3 in structures similar to those containing PML. At 3 h after infection, ICP0 was translocated to the cytoplasm whereas cyclin D3 remained in the nucleus. The translocation of ICP0 to the cytoplasm was accelerated in cells expressing cyclin D3 compared with that of ICP0 expressed by wild-type virus. In contrast, ICP0 carrying the D199A substitution remained in the nucleus and did not colocalize with cyclin D3. These studies suggest the following conclusions. (i) ICP0 brings to the vicinity of ND10 cyclin D3 and, in consequence, an activated cdk4. The metabolic events occurring at or near that structure and involving cyclin D3 cause the translocation of ICP0 to the cytoplasm. (ii) In the absence of the cyclin D3 binding site in ICP0, cyclin D3 is not brought to ND10, cyclin D is not stabilized, and the function responsible for the translocation of ICP0 is not expressed, and in quiescent HEL fibroblasts the yields of virus are reduced.


Subject(s)
Cyclins/metabolism , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Cyclin D1/metabolism , Cyclin D3 , Cyclin-Dependent Kinases/metabolism , Fluorescent Antibody Technique , HeLa Cells , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Leupeptins/pharmacology , Recombinant Proteins/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases
19.
Herpes ; 8(1): 23-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11867013

ABSTRACT

Hippocrates was the first to describe lesions that could have been caused by herpes simplex virus (HSV), but the clinical conditions caused by this virus have only been described in more detail over the past 3 centuries. Most of the key findings relating to HSV infection and treatment have been made since the early 20th Century. These range from discovering some of the mechanisms behind virus latency and reactivation, to the development of the drug aciclovir, which was the first selective inhibitor of HSV replication. This review provides an evolutionary understanding of HSV, but HSV research is still in its golden age. New facts continue to emerge about HSV, and manipulation of the virus is providing much information. Genetic engineering of this virus is likely to have a most significant impact on future medical therapies, which could extend to specialties beyond virology.


Subject(s)
Herpesviridae Infections/history , Simplexvirus , Clinical Trials as Topic/history , History, 18th Century , History, 19th Century , History, 20th Century , History, Ancient , History, Medieval , Humans
20.
Gene Ther ; 8(24): 1840-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11821937

ABSTRACT

Neointimal hyperplasia resulting from vascular smooth muscle cell (SMC) proliferation and luminal migration is the major cause of autologous vein graft failure following vascular coronary or peripheral bypass surgery. Strategies to attenuate SMC proliferation by the delivery of oligonucleotides or genes controlling cell division rely on the use of high concentrations of vectors, and require pre-emptive disruption of the endothelial cell layer. We report a genetically engineered herpes simplex virus (HSV-1) mutant that, in an in vivo rabbit model system, infects all vascular layers without prior injury to the endothelium; expresses a reporter gene driven by a viral promoter with high efficiency for at least 4 weeks; exhibits no systemic toxicity; can be eliminated at will by administration of the antiviral drug acyclovir; and significantly reduces SMC proliferation and restenosis in vein grafts in immunocompetent hosts.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Graft Occlusion, Vascular/prevention & control , Herpesvirus 1, Human/genetics , Tunica Intima/pathology , Animals , Humans , Hyperplasia/prevention & control , Jugular Veins , Models, Animal , Muscle, Smooth, Vascular , Mutation , Organ Culture Techniques/methods , Rabbits , Recurrence , Saphenous Vein , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL