Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Curr Top Microbiol Immunol ; 325: 41-61, 2008.
Article in English | MEDLINE | ID: mdl-18637499

ABSTRACT

Bacterial artificial chromosomes (BACs) are DNA molecules assembled in vitro from defined constituents and are stably maintained as one large DNA fragment in Escherichia coli. Artificial chromosomes are useful for genome sequencing programs, for transduction of DNA segments into eukaryotic cells, and for functional characterization of genomic regions and entire viral genomes such as cytomegalovirus (CMV) genomes. CMV genomes in BACs are ready for the advanced tools of E. coli genetics. Homologous and site-specific recombination, or transposon-based approaches allow for the engineering of virtually any kind of genetic change.


Subject(s)
Cytomegalovirus/genetics , Genome, Viral , Chromosomes, Artificial, Bacterial , Escherichia coli/genetics , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Recombination, Genetic
2.
J Cell Mol Med ; 12(5B): 1974-89, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18194452

ABSTRACT

BACKGROUND: Diseases caused by gammaherpesviruses continue to be a challenge for human health and antiviral treatment. Most of the commonly used antiviral drugs are directed against viral gene products. However, the emergence of drug-resistant mutations ma limit the effectiveness of these drugs. Since viruses require a host cell to propagate, the search for host cell targets is an interesting alternative. METHODS: In this study, we infected three different cell types (fibroblasts, endothelial precursor cells and macrophages with a murine gammaherpesvirus and analysed the host cell response for changes either common to all or unique to a particular cell type using oligonucleotide microarrays. RESULTS: The analysis revealed a number of genes whose transcription was significantly up- or down-regulated in either one or two of the cell types tested. After infection, only two genes, Lman1 (also known as ERGIC53) an synaptobrevin-like 1 (sybl1) were significantly up-regulated in all three cell types, suggestive for a general role for the virus life cycl independent of the cell type. Both proteins have been implicated in cellular exocytosis and transport of glycoproteins through the secretory pathway. To test the significance of the observed up-regulation, the functionality of these proteins was modulated, and the effect on virus replication was monitored. Inhibition of either Lman1 or sybl1 resulted in a significant reduction in virus production. CONCLUSIONS: This suggests that proteins of the secretory pathway which appear to be rate limiting for virus production may represent new targets for intervention.


Subject(s)
Gammaherpesvirinae/physiology , Gene Expression Regulation, Viral , Herpesviridae Infections/pathology , Proteins/genetics , Secretory Pathway/physiology , Cell Line , Gammaherpesvirinae/genetics , Humans , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/metabolism , RNA, Viral/genetics , RNA, Viral/isolation & purification , RNA, Viral/metabolism , Secretory Pathway/genetics , Viral Proteins , Virus Physiological Phenomena , Virus Replication , Viruses/genetics
3.
J Gen Virol ; 86(Pt 4): 907-917, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784885

ABSTRACT

Several features make bovine herpesvirus 4 (BoHV-4) attractive as a backbone for use as a viral expression vector and/or as a model to study gammaherpesvirus biology. However, these developments have been impeded by the difficulty in manipulating its large genome using classical homologous recombination in eukaryotic cells. In the present study, the feasibility of exploiting bacterial artificial chromosome (BAC) cloning and prokaryotic recombination technology for production of BoHV-4 recombinants was explored. Firstly, the BoHV-4 genome was BAC cloned using two potential insertion sites. Both sites of insertion gave rise to BoHV-4 BAC clones stably maintained in bacteria and able to regenerate virions when transfected into permissive cells. Reconstituted virus replicated comparably to wild-type parental virus and the loxP-flanked BAC cassette was excised by growing them on permissive cells stably expressing Cre recombinase. Secondly, BoHV-4 recombinants expressing Ixodes ricinus anti-complement protein I or II (IRAC I/II) were produced using a two-step mutagenesis procedure in Escherichia coli. Both recombinants induced expression of high levels of functional IRAC molecules in the supernatant of infected cells. This study demonstrates that BAC cloning and prokaryotic recombination technology are powerful tools for the development of BoHV-4 as an expression vector and for further fundamental studies of this gammaherpesvirus.


Subject(s)
Chromosomes, Artificial, Bacterial , Cloning, Molecular , Genetic Vectors , Herpesvirus 4, Bovine/genetics , Herpesvirus 4, Bovine/metabolism , Animals , Cattle , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Herpesvirus 4, Bovine/physiology , Ixodes/immunology , Ixodes/metabolism , Recombination, Genetic , Virus Replication
4.
Curr Top Microbiol Immunol ; 269: 1-22, 2002.
Article in English | MEDLINE | ID: mdl-12224503

ABSTRACT

Co-evolution of herpesviruses with their hosts has resulted in multiple interactions between viral genes and cellular functions. Some interactions control genomic maintenance and replication in specific tissues, other affect the immune control at various stages. Few immunomodulatory functions of genes can be predicted by sequence homology. The majority of genes with immunomodulatory properties only become apparent in functional assays. This chapter reviews procedures which have been used for successful identification of immunomodulatory genes in the past and deals with recent methods which may be applicable for the identification of additional immunomodulatory functions unknown so far.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Genes, Viral , Animals , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Disease Models, Animal , Genes, MHC Class I , Genetic Techniques , Mice , Muromegalovirus/genetics , Muromegalovirus/immunology , Mutation , Viral Proteins/genetics , Viral Proteins/immunology
5.
Nat Immunol ; 3(8): 733-40, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101398

ABSTRACT

The murine gamma-herpesvirus-68 (MHV-68) K3 protein, like that of the Kaposi's sarcoma associated herpesvirus, down-regulates major histocompatibility complex (MHC) class I expression. However, how this contributes to viral replication in vivo is unclear. After intranasal MHV-68 infection, K3 was transcribed both during acute lytic infection in the lung and during latency establishment in lymphoid tissue. K3-deficient viruses were not cleared more rapidly from the lung, but the number of latently infected spleen cells was reduced and the frequency of virus-specific CD8(+) cytotoxic T lymphocytes (CTLs) was increased. CTL depletion reversed the viral latency deficit. Thus, a major function of K3 appears to be CTL evasion during viral latency expansion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Rhadinovirus/immunology , Viral Proteins/immunology , 3T3 Cells , Animals , Flow Cytometry , Gene Expression Regulation/immunology , Genes, MHC Class I/immunology , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Insertional , Polymerase Chain Reaction , RNA, Viral/analysis , RNA, Viral/genetics , Rhadinovirus/genetics , Rhadinovirus/growth & development , Spleen/virology , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic/immunology , Viral Proteins/biosynthesis , Viral Proteins/genetics
6.
J Exp Med ; 194(7): 967-78, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11581318

ABSTRACT

Both human cytomegaloviruses (HCMVs) and murine cytomegaloviruses (MCMVs) encode multiple genes that interfere with antigen presentation by major histocompatibility complex (MHC) class I, and thus protect infected targets from lysis by virus-specific cytotoxic T lymphocytes (CTLs). HCMV has been shown to encode four such genes and MCMV to encode two. MCMV m152 blocks the export of class I from a pre-Golgi compartment, and MCMV m6 directs class I to the lysosome for degradation. A third MCMV gene, m4, encodes a glycoprotein which is expressed at the cell surface in association with class I. Here we here show that m4 is a CTL-evasion gene which, unlike previously described immune-evasion genes, inhibited CTLs without blocking class I surface expression. m152 was necessary to block antigen presentation to both K(b)- and D(b)-restricted CTL clones, while m4 was necessary to block presentation only to K(b)-restricted clones. m152 caused complete retention of D(b), but only partial retention of K(b), in a pre-Golgi compartment. Thus, while m152 effectively inhibited D(b)-restricted CTLs, m4 was required to completely inhibit K(b)-restricted CTLs. We propose that cytomegaloviruses encode multiple immune-evasion genes in order to cope with the diversity of class I molecules in outbred host populations.


Subject(s)
Antigen Presentation , Genes, Viral , Muromegalovirus/genetics , Muromegalovirus/immunology , Viral Proteins/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Clone Cells/immunology , Cytotoxicity, Immunologic , Glycoproteins/genetics , Glycoproteins/immunology , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class I/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , T-Lymphocytes, Cytotoxic
7.
J Infect Dis ; 184(10): 1320-4, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11679922

ABSTRACT

Mouse infection with murine cytomegalovirus (MCMV) is an established model for studying human cytomegalovirus infection. In this study, the relationship was analyzed between MCMV activity in organs of infected mice and the presence of infectious virus (viremia), viral genomes (DNAemia), or secreted virus-encoded proteins in the blood. For the latter, 2 recombinant viruses were constructed that encode for the hepatitis B virus surface antigen and the secreted alkaline phosphatase, respectively, as secreted marker proteins. The secreted markers correlated better with the infection in organs than DNAemia and viremia. The marker protein assays can serve as practical and sensitive tools for longitudinal monitoring of MCMV infection in individual mice.


Subject(s)
Herpesviridae Infections/virology , Muromegalovirus/isolation & purification , Viral Proteins/blood , Alkaline Phosphatase/blood , Animals , DNA, Viral/blood , Disease Models, Animal , Hepatitis B Antigens/blood , Hepatitis B Surface Antigens/blood , Herpesviridae Infections/blood , Immunocompromised Host , Mice , Mice, Inbred BALB C , Muromegalovirus/genetics , Polymerase Chain Reaction , Recombination, Genetic , Time Factors , Viremia , Viscera/virology
8.
J Immunol ; 167(8): 4238-44, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591745

ABSTRACT

CTL recognize peptides that derive from viral protein Ags by proteolytic processing and are presented by MHC class I molecules. In this study we tested whether coexpression of viral Ags in the same cell leads to competition between them. To this end, two L(d)-restricted epitopes derived from HIV-1 envelope gp160 (ENV) and from CMV pp89 phosphoprotein were coexpressed. HIV ENV strain IIIB, but not MN variant, impaired recognition by specific CTL of CMV pp89 epitope 9pp89. Susceptibility to inhibition after ENV coexpression was inversely related to the amount of antigenic 9pp89 peptide processed from different antigenic constructs. In line with it, competition decreased the yield of naturally processed antigenic 9pp89 peptide bound to MHC class I molecules in coinfected cells. Also, point mutants of the presenting MHC class I molecule differed in their competition pattern. Collectively, the data imply that competition operates at the step of MHC-peptide complex assembly or stabilization. We conclude that, although not the rule, in certain combinations there is interference between different Ags expressed in the same cell and presented by the same MHC class I allele. These studies have implications for vaccine development and for understanding immunodominance.


Subject(s)
Antigen Presentation , HIV Envelope Protein gp160/immunology , HIV-1/immunology , Histocompatibility Antigens Class I/immunology , Immediate-Early Proteins/immunology , Animals , HIV Envelope Protein gp120/immunology , Histocompatibility Antigens Class I/genetics , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology , Point Mutation
9.
J Immunol ; 167(7): 3894-902, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564807

ABSTRACT

We have recently demonstrated that the murine CMV (MCMV) gene m4 is an immune evasion gene that protects MCMV-infected targets from some virus-specific CTL clones. m4 encodes m4/gp34, a 34-kDa glycoprotein that binds to major histocompatibility complex class I in the endoplasmic reticulum and forms a detergent-stable complex that is exported to the surface of the cell. To investigate how m4/gp34 promotes CTL evasion, we analyzed the assembly and export of m4/gp34-K(b) complexes. We found that 50-70% of K(b) exported over the course of MCMV infection was m4/gp34 associated. Because these complexes are present at the cell surface, it is possible that m4 mediates CTL evasion by interfering with contact between class I and receptors on the T cell. In addition, we found that K(b) retained by the MCMV immune evasion gene m152 formed a novel type of complex with Endo H-sensitive m4/gp34; these complexes are distinguished from the exported complexes by being stable in 1% digitonin and unstable in 1% Nonidet P-40. Because this association occurs in a pre-Golgi compartment, m4/gp34 might also interfere with Ag presentation by affecting some aspect of class I assembly, such as peptide loading. Although m4/gp34 requires beta(2)-microglobulin to bind class I, there was no significant binding of m4/gp34 to beta(2)-microglobulin in the absence of class I H chain, demonstrating that m4/gp34 forms Nonidet P-40-stable complexes specifically with folded conformations of class I. We conclude that m4/gp34 promotes immune evasion by a novel mechanism involving altered assembly and/or T cell recognition of class I molecules.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Golgi Apparatus/metabolism , H-2 Antigens/metabolism , Muromegalovirus/immunology , Viral Proteins , Animals , Cell Membrane/metabolism , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/virology , Macromolecular Substances , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Muromegalovirus/pathogenicity , Octoxynol , Polyethylene Glycols/chemistry , Protein Transport , beta 2-Microglobulin/metabolism
10.
J Virol ; 75(12): 5692-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356978

ABSTRACT

We studied the in vivo biological properties of viruses reconstituted from the genome of murine gammaherpesvirus 68 (MHV-68) cloned as an infectious bacterial artificial chromosome (BAC). Recombinant virus RgammaHV68A98.01, containing BAC vector sequences, is attenuated in vivo as determined by (i) viral titers in the lungs during the acute phase of infection, (ii) the extent of splenomegaly, and (iii) the number of latently infected spleen cells reactivating virus in an ex vivo reactivation assay. Since the BAC vector sequences were flanked by loxP sites, passaging the virus in fibroblasts expressing Cre recombinase resulted in the generation of recombinant virus RgammaHV68A98.02, with biological properties comparable to those of wild-type MHV-68. On the basis of these data we conclude (i) that excision of BAC vector sequences from cloned MHV-68 genomes is critical for reconstitution of the wild-type phenotypic properties of this virus and (ii) that the BAC-cloned MHV-68 genome is suitable for the construction of mutants and mutant libraries whose phenotypes can be reliably assessed in vivo.


Subject(s)
Chromosomes, Artificial, Bacterial , Gammaherpesvirinae/genetics , Gammaherpesvirinae/pathogenicity , Herpesviridae Infections/physiopathology , Animals , Cloning, Molecular , Disease Models, Animal , Female , Gammaherpesvirinae/physiology , Herpesviridae Infections/virology , Mice , Mice, Inbred C57BL , Mice, SCID , Virulence
11.
Science ; 291(5502): 303-5, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11209080

ABSTRACT

Human cytomegalovirus infects vascular tissues and has been associated with atherogenesis and coronary restenosis. Although established laboratory strains of human cytomegalovirus have lost the ability to grow on vascular endothelial cells, laboratory strains of murine cytomegalovirus retain this ability. With the use of a forward-genetic procedure involving random transposon mutagenesis and rapid phenotypic screening, we identified a murine cytomegalovirus gene governing endothelial cell tropism. This gene, M45, shares sequence homology to ribonucleotide reductase genes. Endothelial cells infected with M45-mutant viruses rapidly undergo apoptosis, suggesting that a viral strategy to evade destruction by cellular apoptosis is indispensable for viral growth in endothelial cells.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Genes, Viral , Muromegalovirus/genetics , Muromegalovirus/physiology , Ribonucleotide Reductases/genetics , Viral Proteins , 3T3 Cells , Animals , Apoptosis , Base Sequence , Cell Line , Cytopathogenic Effect, Viral , DNA Transposable Elements , Fibroblasts/virology , Frameshift Mutation , Gene Library , Mice , Molecular Sequence Data , Muromegalovirus/growth & development , Mutagenesis, Insertional , Open Reading Frames , Phenotype , Ribonucleotide Reductases/physiology , Virus Replication
12.
Curr Protoc Immunol ; Chapter 19: Unit 19.7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-18432758

ABSTRACT

This unit describes procedures for infecting newborn and adult mice with murine cytomegalovirus (mCMV). Methods are included for propagating mCMV in cell cultures and for preparing a more virulent form of mCMV from salivary glands of infected mice. A plaque-forming cell (PFC) assay is provided for measuring mCMV titers of infected tissues or virus stocks. In addition, a method is described for preparing the murine embryonic fibroblasts used for propagating mCMV and for the PFC assay.


Subject(s)
Cytomegalovirus Infections , Disease Models, Animal , Herpesviridae Infections , Muromegalovirus , Salivary Glands/virology , Viral Plaque Assay/methods , Animals , Cytomegalovirus Infections/virology , Fibroblasts/virology , Herpesviridae Infections/virology , Mice , Muromegalovirus/isolation & purification , Muromegalovirus/physiology , Viral Load , Virus Replication
14.
Trends Microbiol ; 8(9): 410-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10989308

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Antibody Formation , Apoptosis , Immunity, Cellular , Interferons , Major Histocompatibility Complex , Receptors, Cytokine
15.
Mol Med Today ; 6(9): 365-72, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954870

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Virus Diseases/virology , Animals , Antibody Formation , Apoptosis/immunology , Chemokines/immunology , Cytokines/immunology , Forecasting , Humans , Interferons/immunology , Killer Cells, Natural/immunology , Major Histocompatibility Complex/immunology , T-Lymphocytes, Cytotoxic/immunology
16.
Immunol Today ; 21(9): 447-55, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953097

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Animals , Apoptosis , Chemokines/physiology , Cytokines/physiology , Histocompatibility Antigens/physiology , Humans , Immune Tolerance , Interferons/physiology , T-Lymphocytes, Cytotoxic/immunology
17.
J Virol ; 74(17): 7720-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933677

ABSTRACT

We have cloned the human cytomegalovirus (HCMV) genome as an infectious bacterial artificial chromosome (BAC) in Escherichia coli. Here, we have subjected the HCMV BAC to random transposon (Tn) mutagenesis using a Tn1721-derived insertion sequence and have provided the conditions for excision of the BAC cassette. We report on a fast and efficient screening procedure for a Tn insertion library. Bacterial clones containing randomly mutated full-length HCMV genomes were transferred into 96-well microtiter plates. A PCR screening method based on two Tn primers and one primer specific for the desired genomic position of the Tn insertion was established. Within three consecutive rounds of PCR a Tn insertion of interest can be assigned to a specific bacterial clone. We applied this method to retrieve mutants of HCMV envelope glycoprotein genes. To determine the infectivities of the mutant HCMV genomes, the DNA of the identified BACs was transfected into permissive fibroblasts. In contrast to BACs with mutations in the genes coding for gB, gH, gL, and gM, which did not yield infectious virus, BACs with disruptions of open reading frame UL4 (gp48) or UL74 (gO) were viable, although gO-deficient viruses showed a severe growth deficit. Thus, gO (UL74), a component of the glycoprotein complex III, is dispensable for viral growth. We conclude that our approach of PCR screening for Tn insertions will greatly facilitate the functional analysis of herpesvirus genomes.


Subject(s)
Cytomegalovirus/genetics , Genome, Viral , Membrane Glycoproteins/genetics , Viral Envelope Proteins/genetics , DNA Transposable Elements , Escherichia coli/genetics , Fibroblasts/virology , Genetic Techniques , Humans , Mutagenesis, Insertional , Plasmids , Polymerase Chain Reaction
18.
J Virol ; 74(17): 7861-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933693

ABSTRACT

The mouse cytomegalovirus (MCMV) m152- and m06-encoded glycoproteins gp40 and gp48, respectively, independently downregulate major histocompatibility complex (MHC) class I surface expression during the course of productive MCMV infection in fibroblasts. As a result, presentation of an immediate-early protein pp89-derived nonapeptide to H-2L(d)-restricted CD8(+) cytotoxic T cells is completely prevented in fibroblasts. Here we demonstrate that MCMV-infected primary bone marrow macrophages and the macrophage cell line J774 constitutively present pp89 peptides during permissive MCMV infection to cytotoxic T lymphocytes (CTL). In contrast to fibroblasts, expression of the m152 and m06 genes in macrophages does not affect surface expression of MHC class I. Assessment of pp89 synthesis and quantification of extracted peptide revealed a significantly higher efficiency of macrophages than of fibroblasts to process pp89 into finally trimmed peptide. The yield of pp89 peptide determined in MCMV-infected tissues of bone marrow chimeras confirmed that bone marrow-derived cells represent a prime source of pp89 processing in parenchymal organs. The finding that macrophages resist the viral control of MHC I-dependent antigen presentation reconciles the paradox of efficient induction of CMV-specific CD8(+) CTL in vivo despite extensive potential of CMVs to subvert MHC class I.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/virology , Cytomegalovirus/metabolism , Histocompatibility Antigens Class I/metabolism , Macrophages/virology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytomegalovirus/immunology , Histocompatibility Antigens Class I/immunology , Immediate-Early Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Viral Envelope Proteins/metabolism , Viral Proteins/analysis
19.
J Virol ; 74(15): 6964-74, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10888635

ABSTRACT

Gammaherpesviruses cause important infections of humans, in particular in immunocompromised patients. Recently, murine gammaherpesvirus 68 (MHV-68) infection of mice has been developed as a small animal model of gammaherpesvirus pathogenesis. Efficient generation of mutants of MHV-68 would significantly contribute to the understanding of viral gene functions in virus-host interaction, thereby further enhancing the potential of this model. To this end, we cloned the MHV-68 genome as a bacterial artificial chromosome (BAC) in Escherichia coli. During propagation in E. coli, spontaneous recombination events within the internal and terminal repeats of the cloned MHV-68 genome, affecting the copy number of the repeats, were occasionally observed. The gene for the green fluorescent protein was incorporated into the cloned BAC for identification of infected cells. BAC vector sequences were flanked by loxP sites to allow the excision of these sequences using recombinase Cre and to allow the generation of recombinant viruses with wild-type genome properties. Infectious virus was reconstituted from the BAC-cloned MHV-68. Growth of the BAC-derived virus in cell culture was indistinguishable from that of wild-type MHV-68. To assess the feasibility of mutagenesis of the cloned MHV-68 genome, a mutant virus with a deletion of open reading frame 4 was generated. Genetically modified MHV-68 can now be analyzed in functionally modified mouse strains to assess the role of gammaherpesvirus genes in virus-host interaction and pathogenesis.


Subject(s)
Chromosomes, Bacterial/genetics , Cloning, Molecular , Gammaherpesvirinae/genetics , Gammaherpesvirinae/pathogenicity , Genome, Viral , Mutagenesis, Site-Directed , Animals , Electroporation/methods , Escherichia coli/genetics , Gammaherpesvirinae/physiology , Gene Deletion , Mice , Open Reading Frames , Plasmids/genetics , Recombination, Genetic , Viral Plaque Assay
20.
Trends Genet ; 16(6): 254-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10827452

ABSTRACT

The large, complex genomes of herpesviruses document the high degree of adaptation of these viruses to their hosts. Not surprisingly, the methods developed over the past 30 years to analyse herpesvirus genomes have paralleled those used to investigate the genetics of eukaryotic cells. The recent use of bacterial artificial chromosome (BAC) technology in herpesvirus genetics has made their genomes accessible to the tools of bacterial genetics. This has opened up new avenues for reverse and forward genetics of this virus family in basic research, and also for vector and vaccine development.


Subject(s)
Chromosomes, Bacterial , Genetic Techniques , Herpesviridae/genetics , Mutagenesis , Alleles , DNA Transposable Elements , Forecasting , Genome, Viral
SELECTION OF CITATIONS
SEARCH DETAIL
...