Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Dairy Res ; 87(2): 196-203, 2020 May.
Article in English | MEDLINE | ID: mdl-32308161

ABSTRACT

Subclinical (SCK) and clinical (CK) ketosis are metabolic disorders responsible for big losses in dairy production. Although Fourier-transform mid-infrared spectrometry (FTIR) to predict ketosis in cows exposed to great metabolic stress was studied extensively, little is known about its suitability in predicting hyperketonemia using individual samples, e.g. in small dairy herds or when only few animals are at risk of ketosis. The objective of the present research was to determine the applicability of milk metabolites predicted by FTIR spectrometry in the individual screening for ketosis. In experiment 1, blood and milk samples were taken every two weeks after calving from Holstein (n = 80), Brown Swiss (n = 72) and Swiss Fleckvieh (n = 58) cows. In experiment 2, cows diagnosed with CK (n = 474) and 420 samples with blood ß-hydroxybutyrate [BHB] <1.0 mmol/l were used to investigate if CK could be detected by FTIR-predicted BHB and acetone from a preceding milk control. In experiment 3, correlations between data from an in farm automatic milk analyser and FTIR-predicted BHB and acetone from the monthly milk controls were evaluated. Hyperketonemia occurred in majority during the first eight weeks of lactation. Correlations between blood BHB and FTIR-predicted BHB and acetone were low (r = 0.37 and 0.12, respectively, P < 0.0001), as well as the percentage of true positive values (11.9 and 16.6%, respectively). No association of FTIR predicted ketone bodies with the interval of milk sampling relative to CK diagnosis was found. Data obtained from the automatic milk analyser were moderately correlated with the same day FTIR-predicted BHB analysis (r = 0.61). In conclusion, the low correlations with blood BHB and the small number of true positive samples discourage the use of milk mid-infrared spectrometry analyses as the only method to predict hyperketonemia at the individual cow level.


Subject(s)
3-Hydroxybutyric Acid/analysis , Acetone/analysis , Cattle Diseases/diagnosis , Ketosis/veterinary , Milk/chemistry , Stress, Physiological/physiology , 3-Hydroxybutyric Acid/blood , Animals , Cattle , Female , Ketosis/diagnosis , Lactation , Spectroscopy, Fourier Transform Infrared/veterinary
2.
J Gen Virol ; 100(2): 246-258, 2019 02.
Article in English | MEDLINE | ID: mdl-30526737

ABSTRACT

Protecting against persistent viruses is an unsolved challenge. The clearest example for a gamma-herpesvirus is resistance to super-infection by Murid herpesvirus-4 (MuHV-4). Most experimental infections have delivered MuHV-4 into the lungs. A more likely natural entry site is the olfactory epithelium. Its protection remains unexplored. Here, prior exposure to olfactory MuHV-4 gave good protection against super-infection. The protection was upstream of B cell infection, which occurs in lymph nodes, and showed redundancy between antibody and T cells. Adding antibody to virions that blocked heparan binding strongly reduced olfactory host entry - unlike in the lungs, opsonized virions did not reach IgG Fc receptor+ myeloid cells. However, the nasal antibody response to primary infection was too low to reduce host entry. Instead, the antibody acted downstream, reducing viral replication in the olfactory epithelium. This depended on IgG Fc receptor engagement rather than virion neutralization. Thus antibody can protect against natural γ-herpesvirus infection before it reaches B cells and independently of neutralization.


Subject(s)
Antibodies, Viral/immunology , Herpesviridae Infections/immunology , Herpesviridae/immunology , Olfactory Mucosa/immunology , Olfactory Mucosa/virology , Animals , Mice , Virus Attachment , Virus Internalization , Virus Replication
3.
PLoS One ; 12(1): e0170908, 2017.
Article in English | MEDLINE | ID: mdl-28125695

ABSTRACT

As their names imply, parvoviruses of the genus Dependovirus rely for their efficient replication on the concurrent presence of a helpervirus, such as herpesvirus, adenovirus, or papilloma virus. Adeno-associated virus 2 (AAV2) is such an example, which in turn can efficiently inhibit the replication of each helpervirus by distinct mechanisms. In a previous study we have shown that expression of the AAV2 rep gene is not compatible with efficient replication of herpes simplex virus 1 (HSV-1). In particular, the combined DNA-binding and ATPase/helicase activities of the Rep68/78 proteins have been shown to exert opposite effects on the replication of AAV2 and HSV-1. While essential for AAV2 DNA replication these protein activities account for the Rep-mediated inhibition of HSV-1 replication. Here, we describe a novel Rep mutant (Rep-D371Y), which displayed an unexpected phenotype. Rep-D371Y did not block HSV-1 replication, but still supported efficient AAV2 replication, at least when a double-stranded AAV2 genome template was used. We also found that the capacity of Rep-D371Y to induce apoptosis and a Rep-specific DNA damage response was significantly reduced compared to wild-type Rep. These findings suggest that AAV2 Rep-helicase subdomains exert diverging activities, which contribute to distinct steps of the AAV2 life cycle. More important, the novel AAV2 mutant Rep-D371Y may allow deciphering yet unsolved activities of the AAV2 Rep proteins such as DNA second-strand synthesis, genomic integration or packaging, which all involve the Rep-helicase activity.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , Dependovirus/genetics , Herpesvirus 1, Human/genetics , Viral Proteins/genetics , Virus Replication , Animals , Chlorocebus aethiops , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Dependovirus/metabolism , Herpesvirus 1, Human/metabolism , Vero Cells , Viral Proteins/metabolism
4.
J Virol ; 89(21): 11150-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292324

ABSTRACT

Adeno-associated virus type 2 is known to inhibit replication of herpes simplex virus 1 (HSV-1). This activity has been linked to the helicase- and DNA-binding domains of the Rep68/Rep78 proteins. Here, we show that Rep68 can bind to consensus Rep-binding sites on the HSV-1 genome and that the Rep helicase activity can inhibit replication of any DNA if binding is facilitated. Therefore, we hypothesize that inhibition of HSV-1 replication involves direct binding of Rep68/Rep78 to the HSV-1 genome.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Genome, Viral/genetics , Herpesvirus 1, Human/genetics , Viral Proteins/metabolism , Binding Sites/genetics , Blotting, Western , Dependovirus/metabolism , Herpesvirus 1, Human/metabolism , Humans
5.
J Virol ; 87(19): 10828-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903840

ABSTRACT

Glycoprotein B (gB) is a conserved herpesvirus virion component implicated in membrane fusion. As with many-but not all-herpesviruses, the gB of murid herpesvirus 4 (MuHV-4) is cleaved into disulfide-linked subunits, apparently by furin. Preventing gB cleavage for some herpesviruses causes minor infection deficits in vitro, but what the cleavage contributes to host colonization has been unclear. To address this, we mutated the furin cleavage site (R-R-K-R) of the MuHV-4 gB. Abolishing gB cleavage did not affect its expression levels, glycosylation, or antigenic conformation. In vitro, mutant viruses entered fibroblasts and epithelial cells normally but had a significant entry deficit in myeloid cells such as macrophages and bone marrow-derived dendritic cells. The deficit in myeloid cells was not due to reduced virion binding or endocytosis, suggesting that gB cleavage promotes infection at a postendocytic entry step, presumably viral membrane fusion. In vivo, viruses lacking gB cleavage showed reduced lytic spread in the lungs. Alveolar epithelial cell infection was normal, but alveolar macrophage infection was significantly reduced. Normal long-term latency in lymphoid tissue was established nonetheless.


Subject(s)
Glycoproteins/metabolism , Lung/virology , Myeloid Cells/virology , Rhadinovirus/physiology , Viral Envelope Proteins/metabolism , Animals , Antibodies, Neutralizing/immunology , Base Sequence , Blotting, Western , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/virology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Flow Cytometry , Fluorescent Antibody Technique , Furin/metabolism , Glycoproteins/genetics , Lung/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Viral Envelope Proteins/genetics , Virion , Virus Replication
6.
J Gen Virol ; 93(Pt 6): 1316-1327, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22377583

ABSTRACT

Herpesviruses consistently transmit from immunocompetent carriers, implying that their neutralization is hard to achieve. Murid herpesvirus-4 (MuHV-4) exploits host IgG Fc receptors to bypass blocks to cell binding, and pH-dependent protein conformation changes to unveil its fusion machinery only after endocytosis. Nevertheless, neutralization remains possible by targeting the virion glycoprotein H (gH)-gL heterodimer, and the neutralizing antibody responses of MuHV-4 carriers are improved by boosting with recombinant gH-gL. We analysed here how gH-gL-directed neutralization works. The MuHV-4 gH-gL binds to heparan sulfate. However, most gH-gL-specific neutralizing antibodies did not block this interaction; neither did they act directly on fusion. Instead, they blocked virion endocytosis and transport to the late endosomes, where membrane fusion normally occurs. The poor endocytosis of gH-gL-neutralized virions was recapitulated precisely by virions genetically lacking gL. Therefore, driving virion uptake appears to be an important function of gH-gL that provides a major target for antibody-mediated neutralization.


Subject(s)
Endocytosis , Herpesviridae Infections/physiopathology , Herpesviridae Infections/veterinary , Rhadinovirus/immunology , Rodent Diseases/immunology , Virion/immunology , Animals , Antibodies, Neutralizing/immunology , Cell Line , Cricetinae , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Mice , Neutralization Tests , Rhadinovirus/genetics , Rhadinovirus/physiology , Rodent Diseases/physiopathology , Rodent Diseases/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virion/genetics , Virion/physiology
7.
Cold Spring Harb Protoc ; 2012(3): 352-6, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22383640

ABSTRACT

Herpes simplex virus type 1 (HSV-1)-based amplicon vectors conserve most properties of the parental virus: broad host range, the ability to transduce dividing and nondiving cells, and a large transgene capacity. This permits incorporation of genomic sequences as well as cDNA, large transcriptional regulatory sequences for cell-specific expression, multiple transgene cassettes, or genetic elements from other viruses. Hybrid vectors use elements from HSV-1 that allow replication and packaging of large-vector DNA into highly infectious particles, and elements from other viruses that confer genetic stability to vector DNA in the transduced cell. For example, adeno-associated virus (AAV) has the unique ability to integrate its genome into a specific site on human chromosome 19. The viral rep gene and the inverted terminal repeats (ITRs) that flank the AAV genome are sufficient for this process. However, AAV-based vectors have a very small transgene capacity and do not conventionally contain the rep gene to support site-specific genomic integration. HSV/AAV hybrid vectors contain both HSV-1 replication and packaging functions and the AAV rep gene and a transgene cassette flanked by the AAV ITRs. This combines the large transgene capacity of HSV-1 with the capability of site-specific genomic transgene integration and long-term transgene expression of AAV. This protocol describes the preparation of HSV/AAV hybrid vectors using a replication-competent/conditional, packaging-defective HSV-1 genome cloned as a bacterial artificial chromosome (BAC) to provide helper functions for vector replication and packaging. The advantages and limitations of such vectors compared to standard HSV-1 amplicon vectors are also discussed.


Subject(s)
Dependovirus/physiology , Genetic Vectors , Herpesvirus 1, Human/physiology , Molecular Biology/methods , Virus Assembly , Animals , Chlorocebus aethiops , Dependovirus/genetics , Herpesvirus 1, Human/genetics , Host Specificity , Recombination, Genetic , Transduction, Genetic , Vero Cells , Virology/methods
8.
PLoS One ; 7(1): e29726, 2012.
Article in English | MEDLINE | ID: mdl-22276127

ABSTRACT

Refined vaccines and adjuvants are urgently needed to advance immunization against global infectious challenges such as HIV, hepatitis C, tuberculosis and malaria. Large-scale screening efforts are ongoing to identify adjuvants with improved efficacy profiles. Reactogenicity often represents a major hurdle to the clinical use of new substances. Yet, irrespective of its importance, this parameter has remained difficult to screen for, owing to a lack of sensitive small animal models with a capacity for high throughput testing. Here we report that continuous telemetric measurements of heart rate, heart rate variability, body core temperature and locomotor activity in laboratory mice readily unmasked systemic side-effects of vaccination, which went undetected by conventional observational assessment and clinical scoring. Even minor aberrations in homeostasis were readily detected, ranging from sympathetic activation over transient pyrogenic effects to reduced physical activity and apathy. Results in real-time combined with the potential of scalability and partial automation in the industrial context suggest multiparameter telemetry in laboratory mice as a first-line screen for vaccine reactogenicity. This may accelerate vaccine discovery in general and may further the success of vaccines in combating infectious disease and cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Telemetry/methods , Vaccination , Animals , Female , Mice
9.
PLoS One ; 7(1): e30152, 2012.
Article in English | MEDLINE | ID: mdl-22253913

ABSTRACT

Herpesvirus entry is a complicated process involving multiple virion glycoproteins and culminating in membrane fusion. Glycoprotein conformation changes are likely to play key roles. Studies of recombinant glycoproteins have revealed some structural features of the virion fusion machinery. However, how the virion glycoproteins change during infection remains unclear. Here using conformation-specific monoclonal antibodies we show in situ that each component of the Murid Herpesvirus-4 (MuHV-4) entry machinery--gB, gH/gL and gp150--changes in antigenicity before tegument protein release begins. Further changes then occurred upon actual membrane fusion. Thus virions revealed their final fusogenic form only in late endosomes. The substantial antigenic differences between this form and that of extracellular virions suggested that antibodies have only a limited opportunity to block virion membrane fusion.


Subject(s)
Antigens, Viral/immunology , Membrane Fusion/immunology , Rhadinovirus/immunology , Viral Fusion Proteins/immunology , Ammonium Chloride/pharmacology , Animals , Cell Line , Endosomes/drug effects , Endosomes/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Humans , Hydrogen-Ion Concentration/drug effects , Kinetics , Macrolides/pharmacology , Membrane Fusion/drug effects , Neutralization Tests , Rhadinovirus/drug effects , Virion/immunology , Virus Internalization/drug effects
10.
J Virol ; 86(1): 143-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013059

ABSTRACT

Adeno-associated virus type 2 (AAV2) is a human parvovirus that relies on a helper virus for efficient replication. Herpes simplex virus 1 (HSV-1) supplies helper functions and changes the environment of the cell to promote AAV2 replication. In this study, we examined the accumulation of cellular replication and repair proteins at viral replication compartments (RCs) and the influence of replicating AAV2 on HSV-1-induced DNA damage responses (DDR). We observed that the ATM kinase was activated in cells coinfected with AAV2 and HSV-1. We also found that phosphorylated ATR kinase and its cofactor ATR-interacting protein were recruited into AAV2 RCs, but ATR signaling was not activated. DNA-PKcs, another main kinase in the DDR, was degraded during HSV-1 infection in an ICP0-dependent manner, and this degradation was markedly delayed during AAV2 coinfection. Furthermore, we detected phosphorylation of DNA-PKcs during AAV2 but not HSV-1 replication. The AAV2-mediated delay in DNA-PKcs degradation affected signaling through downstream substrates. Overall, our results demonstrate that coinfection with HSV-1 and AAV2 provokes a cellular DDR which is distinct from that induced by HSV-1 alone.


Subject(s)
Coinfection/genetics , DNA Damage , Dependovirus/physiology , Herpes Simplex/genetics , Herpesvirus 1, Human/physiology , Parvoviridae Infections/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Coinfection/enzymology , Coinfection/virology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Herpes Simplex/enzymology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions , Humans , Parvoviridae Infections/enzymology , Parvoviridae Infections/virology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Virus Replication
11.
J Gen Virol ; 92(Pt 9): 2020-2033, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21593277

ABSTRACT

Glycoprotein B (gB) is a conserved, essential component of gammaherpes virions and so potentially vulnerable to neutralization. However, few good gB-specific neutralizing antibodies have been identified. Here, we show that murid herpesvirus 4 is strongly neutralized by mAbs that recognize an epitope close to one of the gB fusion loops. Antibody binding did not stop gB interacting with its cellular ligands or initiating its fusion-associated conformation change, but did stop gB resolving stably to its post-fusion form, and so blocked membrane fusion to leave virions stranded in late endosomes. The conservation of gB makes this mechanism a possible general route to gammaherpesvirus neutralization.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Glycoproteins/immunology , Rhadinovirus/immunology , Viral Structural Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Line , Epitopes/immunology , Mice , Mice, Inbred BALB C , Neutralization Tests
12.
J Virol ; 84(8): 3808-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20106923

ABSTRACT

Adeno-associated virus (AAV) has previously been shown to inhibit the replication of its helper virus herpes simplex virus type 1 (HSV-1), and the inhibitory activity has been attributed to the expression of the AAV Rep proteins. In the present study, we assessed the Rep activities required for inhibition of HSV-1 replication using a panel of wild-type and mutant Rep proteins lacking defined domains and activities. We found that the inhibition of HSV-1 replication required Rep DNA-binding and ATPase/helicase activities but not endonuclease activity. The Rep activities required for inhibition of HSV-1 replication precisely coincided with the activities that were responsible for induction of cellular DNA damage and apoptosis, suggesting that these three processes are closely linked. Notably, the presence of Rep induced the hyperphosphorylation of a DNA damage marker, replication protein A (RPA), which has been reported not to be normally hyperphosphorylated during HSV-1 infection and to be sequestered away from HSV-1 replication compartments during infection. Finally, we demonstrate that the execution of apoptosis is not required for inhibition of HSV-1 replication and that the hyperphosphorylation of RPA per se is not inhibitory for HSV-1 replication, suggesting that these two processes are not directly responsible for the inhibition of HSV-1 replication by Rep.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Dependovirus/physiology , Herpesvirus 1, Human/physiology , Trans-Activators/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Apoptosis , Chlorocebus aethiops , DNA Damage , DNA, Viral/metabolism , Dependovirus/growth & development , Herpesvirus 1, Human/growth & development , Phosphorylation , Sequence Deletion , Vero Cells
13.
J Immunol ; 182(12): 7569-79, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494280

ABSTRACT

Regulation of cytotoxic effector molecule expression in human CTLs after viral or bacterial activation is poorly understood. By using human autologous dendritic cells (DCs) to prime T lymphocytes, we found perforin only highly up-regulated in virus- (HSV-1, vaccinia virus) but not in intracellular bacteria- (Listeria innocua, Listeria monocytogenes, Mycobacterium tuberculosis, Chlamydophila pneumoniae) activated CTLs. In contrast, larger quantities of IFN-gamma and TNF-alpha were produced in Listeria-stimulated cultures. Granzyme B and granulysin were similarly up-regulated by all tested viruses and intracellular bacteria. DCs infected with HSV-1 showed enhanced surface expression of the costimulatory molecule CD252 (CD134L) compared with Listeria-infected DC and induced enhanced secretion of IL-2. Adding blocking CD134 or neutralizing IL-2 Abs during T cell activation reduced the HSV-dependent up-regulation of perforin. These data indicate a distinct CTL effector function in response to intracellular pathogens triggered via differing endogenous IL-2 production upon costimulation through CD252.


Subject(s)
Cytotoxicity, Immunologic/immunology , Interleukin-2/immunology , Lymphocyte Activation/immunology , OX40 Ligand/immunology , T-Lymphocytes/immunology , Cells, Cultured , Chlamydophila/immunology , Dendritic Cells/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Interleukin-2/metabolism , Listeria/immunology , Listeria/pathogenicity , Mycobacterium tuberculosis/immunology , Perforin/immunology , T-Lymphocytes/metabolism , Transcription, Genetic/genetics , Up-Regulation/immunology , Vaccinia virus/immunology
14.
J Gen Virol ; 90(Pt 5): 1202-1214, 2009 May.
Article in English | MEDLINE | ID: mdl-19264603

ABSTRACT

Antibodies readily neutralize acute, epidemic viruses, but are less effective against more indolent pathogens such as herpesviruses. Murid herpesvirus 4 (MuHV-4) provides an accessible model for tracking the fate of antibody-exposed gammaherpesvirus virions. Glycoprotein L (gL) plays a central role in MuHV-4 entry: it allows gH to bind heparan sulfate and regulates fusion-associated conformation changes in gH and gB. However, gL is non-essential: heparan sulfate binding can also occur via gp70, and the gB-gH complex alone seems to be sufficient for membrane fusion. Here, we investigated how gL affects the susceptibility of MuHV-4 to neutralization. Immune sera neutralized gL(-) virions more readily than gL(+) virions, chiefly because heparan sulfate binding now depended on gp70 and was therefore easier to block. However, there were also post-binding effects. First, the downstream, gL-independent conformation of gH became a neutralization target; gL normally prevents this by holding gH in an antigenically distinct heterodimer until after endocytosis. Second, gL(-) virions were more vulnerable to gB-directed neutralization. This covered multiple epitopes and thus seemed to reflect a general opening up of the gH-gB entry complex, which gL again normally restricts to late endosomes. gL therefore limits MuHV-4 neutralization by providing redundancy in cell binding and by keeping key elements of the virion fusion machinery hidden until after endocytosis.


Subject(s)
Antibodies, Viral/immunology , Glycoproteins/immunology , Rhadinovirus/immunology , Rhadinovirus/metabolism , Viral Envelope Proteins/immunology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Epithelial Cells , Female , Fibroblasts , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding
15.
Vet Microbiol ; 137(3-4): 235-42, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19249164

ABSTRACT

Sheep-associated malignant catarrhal fever (MCF), caused by Ovine herpesvirus 2 (OvHV-2), is a usually fatal disease of various ruminants and swine. A system for propagation of OvHV-2 in vitro has not yet been identified, although persistently infected cells have been derived from diseased animals and used to establish an animal model in rabbits. OvHV-2 structural proteins have not been detected in diseased animals and the pathogenesis of OvHV-2 infection is poorly understood. Recently, the genomic sequence of OvHV-2 has been determined, which allowed to predict the amino acid sequences of putative OvHV-2 structural proteins. Based on those predictions, we have generated antisera against two putative structural proteins (ORF43 and ORF63) of OvHV-2 in order to detect sites of active virus replication in experimentally OvHV-2-infected rabbits with signs of MCF. Although histological lesions typical of MCF were detected in multiple tissues, those sera detected viral capsid and tegument antigens exclusively in the appendix but not in other tissues of rabbits with MCF. More specifically, those viral proteins were detected in epithelial cells as well as in M-cells. However, in situ hybridization revealed that ORF63 mRNA was present in epithelial cells of infected rabbits but not in M-cells. Our data suggest that active OvHV-2 replication takes place in certain tissues of animals with MCF and that M-cells may play a role in the pathogenesis of MCF.


Subject(s)
Appendix/cytology , Appendix/virology , Epithelial Cells/virology , Herpesviridae/physiology , Malignant Catarrh/virology , Rabbits , Animals , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology
16.
J Virol ; 82(10): 4974-90, 2008 May.
Article in English | MEDLINE | ID: mdl-18337577

ABSTRACT

We have constructed a recombinant herpes simplex virus type 1 (HSV-1) that simultaneously encodes selected structural proteins from all three virion compartments-capsid, tegument, and envelope-fused with autofluorescent proteins. This triple-fluorescent recombinant, rHSV-RYC, was replication competent, albeit with delayed kinetics, incorporated the fusion proteins into all three virion compartments, and was comparable to wild-type HSV-1 at the ultrastructural level. The VP26 capsid fusion protein (monomeric red fluorescent protein [mRFP]-VP26) was first observed throughout the nucleus and later accumulated in viral replication compartments. In the course of infection, mRFP-VP26 formed small foci in the periphery of the replication compartments that expanded and coalesced over time into much larger foci. The envelope glycoprotein H (gH) fusion protein (enhanced yellow fluorescent protein [EYFP]-gH) was first observed accumulating in a vesicular pattern in the cytoplasm and was then incorporated primarily into the nuclear membrane. The VP16 tegument fusion protein (VP16-enhanced cyan fluorescent protein [ECFP]) was first observed in a diffuse nuclear pattern and then accumulated in viral replication compartments. In addition, it also formed small foci in the periphery of the replication compartments which, however, did not colocalize with the small mRFP-VP26 foci. Later, VP16-ECFP was redistributed out of the nucleus into the cytoplasm, where it accumulated in vesicular foci and in perinuclear clusters reminiscent of the Golgi apparatus. Late in infection, mRFP-VP26, EYFP-gH, and VP16-ECFP were found colocalizing in dots at the plasma membrane, possibly representing mature progeny virus. In summary, this study provides new insights into the dynamics of compartmentalization and interaction among capsid, tegument, and envelope proteins. Similar strategies can also be applied to assess other dynamic events in the virus life cycle, such as entry and trafficking.


Subject(s)
Herpesvirus 1, Human/growth & development , Viral Proteins/biosynthesis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cell Membrane/chemistry , Cell Nucleus/chemistry , Chlorocebus aethiops , Cricetinae , Cytoplasm/chemistry , Fluorescent Antibody Technique , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpesvirus 1, Human/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Envelope/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Viral Proteins/genetics , Red Fluorescent Protein
17.
J Virol ; 81(9): 4732-43, 2007 May.
Article in English | MEDLINE | ID: mdl-17314170

ABSTRACT

We performed live cell visualization assays to directly assess the interaction between competing adeno-associated virus (AAV) and herpes simplex virus type 1 (HSV-1) DNA replication. Our studies reveal the formation of separate AAV and HSV-1 replication compartments and the inhibition of HSV-1 replication compartment formation in the presence of AAV. AAV Rep is recruited into AAV replication compartments but not into those of HSV-1, while the single-stranded DNA-binding protein HSV-1 ICP8 is recruited into both AAV and HSV-1 replication compartments, although with differential staining patterns. Slot blot analysis of coinfected cells revealed a dose-dependent inhibition of HSV-1 DNA replication by wild-type AAV but not by rep-negative recombinant AAV. Consistent with this, Western blot analysis indicated that wild-type AAV affects the levels of the HSV-1 immediate-early protein ICP4 and the early protein ICP8 only modestly but strongly inhibits the accumulation of the late proteins VP16 and gC. Furthermore, we demonstrate that the presence of Rep in the absence of AAV DNA replication is sufficient for the inhibition of HSV-1. In particular, Rep68/78 proteins severely inhibit the formation of mature HSV-1 replication compartments and lead to the accumulation of ICP8 at sites of cellular DNA synthesis, a phenomenon previously observed in the presence of viral polymerase inhibitors. Taken together, our results suggest that AAV and HSV-1 replicate in separate compartments and that AAV Rep inhibits HSV-1 at the level of DNA replication.


Subject(s)
DNA Replication/physiology , Dependovirus/physiology , Herpesvirus 1, Human/physiology , Virus Replication , Animals , Blotting, Western , Chlorocebus aethiops , DNA Primers , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Vero Cells , Viral Proteins/metabolism
18.
Curr Gene Ther ; 6(3): 315-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16787183

ABSTRACT

Chimeric or hybrid herpes simplex virus type 1/adeno-associated virus amplicon vectors combine the large transgene capacity of HSV-1 with the potential for site-specific genomic integration and stable transgene expression of AAV. These chimeric vectors have been demonstrated to support transgene expression for significantly longer periods than standard HSV-1 amplicons. Moreover, HSV/AAV hybrid vectors can mediate integration at the AAVS1 pre-integration site on human chromosome 19 at a relatively high rate, although random integration has also been observed. One major remaining hurdle of HSV/AAV hybrid vectors is the low packaging efficiency and titers when AAV rep sequences are included in the amplicon vector. In the conditions prevalent during the replication/packaging of HSV/AAV hybrid amplicons into HSV-1 virions, in particular the presence of HSV-1 replication factors and AAV Rep protein, at least three different viral origins of DNA replication are active: the HSV-1 ori, the AAV inverted terminal repeats (ITRs), and the p5 promoter/ori driving expression of the AAV rep gene. A detailed understanding of the properties of these origins of DNA replication and the molecular mechanisms of interactions between them, may allow designing novel hybrid vectors that allow the efficient and precise integration of large transgenes in the human genome.


Subject(s)
Dependovirus/genetics , Genetic Engineering , Genetic Vectors , Simplexvirus/genetics
19.
J Virol ; 79(20): 13047-59, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16189007

ABSTRACT

Herpesvirus envelopment is assumed to follow an uneconomical pathway including primary envelopment at the inner nuclear membrane, de-envelopment at the outer nuclear membrane, and reenvelopment at the trans-Golgi network. In contrast to the hypothesis of de-envelopment by fusion of the primary envelope with the outer nuclear membrane, virions were demonstrated to be transported from the perinuclear space to rough endoplasmic reticulum (RER) cisternae. Here we show by high-resolution microscopy that herpes simplex virus 1 envelopment follows two diverse pathways. First, nuclear envelopment includes budding of capsids at the inner nuclear membrane into the perinuclear space whereby tegument and a thick electron dense envelope are acquired. The substance responsible for the dense envelope is speculated to enable intraluminal transportation of virions via RER into Golgi cisternae. Within Golgi cisternae, virions are packaged into transport vacuoles containing one or several virions. Second, for cytoplasmic envelopment, capsids gain direct access from the nucleus to the cytoplasm via impaired nuclear pores. Cytoplasmic capsids could bud at the outer nuclear membrane, at membranes of RER, Golgi cisternae, and large vacuoles, and at banana-shaped membranous entities that were found to continue into Golgi membranes. Envelopes originating by budding at the outer nuclear membrane and RER membrane also acquire a dense substance. Budding at Golgi stacks, designated wrapping, results in single virions within small vacuoles that contain electron-dense substances between envelope and vacuolar membranes.


Subject(s)
Capsid/physiology , Herpesvirus 1, Human/physiology , Animals , Biological Transport , Capsid/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/virology , Endoplasmic Reticulum, Rough/virology , Golgi Apparatus/virology , HeLa Cells , Herpesvirus 1, Human/metabolism , Humans , Immunohistochemistry , Microscopy, Confocal , Vero Cells , Virus Assembly
20.
J Virol ; 79(19): 12218-30, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160148

ABSTRACT

The adeno-associated virus (AAV) inverted terminal repeats (ITRs) contain the AAV Rep protein-binding site (RBS) and the terminal resolution site (TRS), which together act as a minimal origin of DNA replication. The AAV p5 promoter also contains an RBS, which is involved in Rep-mediated regulation of promoter activity, as well as a functional TRS, and origin activity of these signals has in fact been demonstrated previously in the presence of adenovirus helper functions. Here, we show that in the presence of herpes simplex virus type 1 (HSV-1) and AAV Rep protein, p5 promoter-bearing plasmids are efficiently amplified to form large head-to-tail concatemers, which are readily packaged in HSV-1 virions if an HSV-1 DNA-packaging/cleavage signal is provided in cis. We also demonstrate simultaneous and independent replication from the two alternative AAV replication origins, p5 and ITR, on the single-cell level using multicolor-fluorescence live imaging, a finding which raises the possibility that both origins may contribute to the AAV life cycle. Furthermore, we assess the differential affinities of Rep for the two different replication origins, p5 and ITR, both in vitro and in live cells and identify this as a potential mechanism to control the replicative and promoter activities of p5.


Subject(s)
Dependovirus/physiology , Replication Origin/physiology , Animals , Chlorocebus aethiops , DNA Packaging , DNA Replication , Genes, Reporter , HeLa Cells , Herpesvirus 1, Human/physiology , Humans , Microscopy, Confocal , Plasmids , Promoter Regions, Genetic , Terminal Repeat Sequences , Vero Cells , Viral Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...