Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 96(9): e0213721, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35404095

ABSTRACT

Vaccinia virus (VV), the prototypic poxvirus, encodes a repertoire of proteins responsible for the metabolism of its large dsDNA genome. Previous work has furthered our understanding of how poxviruses replicate and recombine their genomes, but little is known about whether the poxvirus genome undergoes DNA repair. Our studies here are aimed at understanding how VV responds to exogenous DNA damage introduced by UV irradiation. Irradiation of cells prior to infection decreased protein synthesis and led to an ∼12-fold reduction in viral yield. On top of these cell-specific insults, irradiation of VV infections at 4 h postinfection (hpi) introduced both cyclobutene pyrimidine dimer (CPD) and 6,4-photoproduct (6,4-PP) lesions into the viral genome led to a nearly complete halt to further DNA synthesis and to a further reduction in viral yield (∼35-fold). DNA lesions persisted throughout infection and were indeed present in the genomes encapsidated into nascent virions. Depletion of several cellular proteins that mediate nucleotide excision repair (XP-A, -F, and -G) did not render viral infections hypersensitive to UV. We next investigated whether viral proteins were involved in combatting DNA damage. Infections performed with a virus lacking the A50 DNA ligase were moderately hypersensitive to UV irradiation (∼3-fold). More strikingly, when the DNA polymerase inhibitor cytosine arabinoside (araC) was added to wild-type infections at the time of UV irradiation (4 hpi), an even greater hypersensitivity to UV irradiation was seen (∼11-fold). Virions produced under the latter condition contained elevated levels of CPD adducts, strongly suggesting that the viral polymerase contributes to the repair of UV lesions introduced into the viral genome. IMPORTANCE Poxviruses remain of significant interest because of their continuing clinical relevance, their utility for the development of vaccines and oncolytic therapies, and their illustration of fundamental principles of viral replication and virus/cell interactions. These viruses are unique in that they replicate exclusively in the cytoplasm of infected mammalian cells, providing novel challenges for DNA viruses. How poxviruses replicate, recombine, and possibly repair their genomes is still only partially understood. Using UV irradiation as a form of exogenous DNA damage, we have examined how vaccinia virus metabolizes its genome following insult. We show that even UV irradiation of cells prior to infection diminishes viral yield, while UV irradiation during infection damages the genome, causes a halt in DNA accumulation, and reduces the viral yield more severely. Furthermore, we show that viral proteins, but not the cellular machinery, contribute to a partial repair of the viral genome following UV irradiation.


Subject(s)
Genome, Viral , Ultraviolet Rays , Vaccinia virus , Virus Replication , Animals , DNA Damage , DNA Repair , Genome, Viral/radiation effects , Mammals , Vaccinia virus/genetics , Vaccinia virus/radiation effects , Viral Proteins/metabolism
2.
J Photochem Photobiol B ; 224: 112319, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34598020

ABSTRACT

The germicidal properties of short wavelength ultraviolet C (UVC) light are well established and used to inactivate many viruses and other microbes. However, much less is known about germicidal effects of terrestrial solar UV light, confined exclusively to wavelengths in the UVA and UVB regions. Here, we have explored the sensitivity of the human coronaviruses HCoV-NL63 and SARS-CoV-2 to solar-simulated full spectrum ultraviolet light (sUV) delivered at environmentally relevant doses. First, HCoV-NL63 coronavirus inactivation by sUV-exposure was confirmed employing (i) viral plaque assays, (ii) RT-qPCR detection of viral genome replication, and (iii) infection-induced stress response gene expression array analysis. Next, a detailed dose-response relationship of SARS-CoV-2 coronavirus inactivation by sUV was elucidated, suggesting a half maximal suppression of viral infectivity at low sUV doses. Likewise, extended sUV exposure of SARS-CoV-2 blocked cellular infection as revealed by plaque assay and stress response gene expression array analysis. Moreover, comparative (HCoV-NL63 versus SARS-CoV-2) single gene expression analysis by RT-qPCR confirmed that sUV exposure blocks coronavirus-induced redox, inflammatory, and proteotoxic stress responses. Based on our findings, we estimate that solar ground level full spectrum UV light impairs coronavirus infectivity at environmentally relevant doses. Given the urgency and global scale of the unfolding SARS-CoV-2 pandemic, these prototype data suggest feasibility of solar UV-induced viral inactivation, an observation deserving further molecular exploration in more relevant exposure models.


Subject(s)
Coronavirus Infections/prevention & control , Coronavirus NL63, Human/radiation effects , Respiratory Tract Infections/prevention & control , SARS-CoV-2/radiation effects , Sunlight , Ultraviolet Rays , Animals , Cell Line , Chlorocebus aethiops , Coronavirus NL63, Human/physiology , Epithelial Cells/virology , Genome, Viral/radiation effects , Humans , SARS-CoV-2/physiology , Transcriptome/radiation effects , Viral Plaque Assay , Virus Inactivation/radiation effects , Virus Replication/radiation effects
3.
BMC Infect Dis ; 20(1): 585, 2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32762666

ABSTRACT

BACKGROUND: The polymerase chain reaction (PCR) is commonly used to detect viral pathogens because of its high sensitivity and specificity. However, conventional PCR methods cannot determine virus infectivity. Virus infectivity is conventionally examined with methods such as the plaque assay, even though such assays require several days. Long-range reverse-transcription quantitative PCR (RT-qPCR) has previously been suggested for the rapid assessment of RNA virus infectivity where the loss of infectivity is attributable to genomic fragmentation. METHODS: IAV was irradiated with 253.7 nm ultraviolet (UV) rays to induce genomic strand breaks that were confirmed by a full-length RT-PCR assay. The IAV was then subjected to plaque assay, conventional RT-qPCR and long-range RT-qPCR to examine the relationship between infectious titer and copy number. A simple linear regression analysis was performed to examine the correlation between the results of these assays. RESULTS: A long-range RT-qPCR assay was developed and validated for influenza A virus (IAV). Although only a few minutes of UV irradiation was required to completely inactivate IAV, genomic RNA remained detectable by the conventional RT-qPCR and the full-length RT-PCR for NS of viral genome following inactivation. A long-range RT-qPCR assay was then designed using RT-priming at the 3' termini of each genomic segment and subsequent qPCR of the 5' regions. UV-mediated IAV inactivation was successfully analyzed by the long-range RT-qPCR assay especially when targeting PA of the viral genome. This was also supported by the regression analysis that the long-range RT-qPCR is highly correlated with plaque assay (Adjusted R2 = 0.931, P = 0.000066). CONCLUSIONS: This study suggests that IAV infectivity can be predicted without the infectivity assays. The rapid detection of pathogenic IAV has, therefore, been achieved with this sensing technology.


Subject(s)
Influenza A virus/genetics , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/methods , Virulence/genetics , Animals , Dogs , Genome, Viral/genetics , Genome, Viral/radiation effects , Influenza A virus/isolation & purification , Influenza A virus/radiation effects , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/virology , RNA Stability/radiation effects , RNA, Viral/genetics , RNA, Viral/radiation effects , Ultraviolet Rays , Virus Inactivation/radiation effects
4.
Food Environ Virol ; 12(1): 20-27, 2020 03.
Article in English | MEDLINE | ID: mdl-31664651

ABSTRACT

Quantitative PCR (qPCR) is a convenient tool for monitoring virus concentrations in water and wastewater treatment trains, though it only informs about virus presence, but not infectivity. This limitation can be overcome if the relationship between infectivity loss and genome decay induced by a given disinfectant is known. Here, we performed inactivation experiments using two human enteroviruses, Coxsackievirus B5 and Echovirus 11, with three disinfection methods: low-pressure ultraviolet light (UV254), free chlorine (FC), and ozone. We compared the inactivation rates as measured by culturing to the decay rates of the whole genome, to evaluate the extent of qPCR-measurable genome damage as a function of inactivation. To determine genome damage, we used an approach that estimates damage across the full viral genome from the measured decay of multiple amplicons distributed across the viral genome. Correlations between inactivation and genome decay were observed for all viruses and all disinfection treatments, but results showed that even among closely related viruses, disinfection methods can damage the viral genome to different extents and that genome damage does not necessarily translate to inactivation. For both viruses, UV254 treatment had the closest relationship between inactivation and genome decay and with ozone, the rate of genome decay exceeded the inactivation rate. Finally, for FC, the ratios between methods were vastly different between viruses. This work provides the basis to relate qPCR measurements to infectivity loss and enables the establishment of molecular monitoring tools for assessing enterovirus inactivation during disinfection treatments of water and wastewater.


Subject(s)
Chlorine/pharmacology , Disinfectants/pharmacology , Enterovirus B, Human/genetics , Genome, Viral/drug effects , Genome, Viral/radiation effects , Ozone/pharmacology , Enterovirus B, Human/drug effects , Enterovirus B, Human/radiation effects , Humans , Ultraviolet Rays , Virus Inactivation/drug effects , Virus Inactivation/radiation effects
5.
Am J Trop Med Hyg ; 100(5): 1275-1277, 2019 05.
Article in English | MEDLINE | ID: mdl-30860018

ABSTRACT

Gamma irradiation using a cobalt-60 source is a commonly used method for the inactivation of infectious specimens to be handled safely in subsequent laboratory procedures. Here, we determined irradiation doses to safely inactivate liquid proteinaceous specimens harboring different emerging/reemerging viral pathogens known to cause neglected tropical and other diseases of regional or global public health importance. By using a representative arenavirus, bunyavirus, coronavirus, filovirus, flavivirus, orthomyxovirus, and paramyxovirus, we found that these enveloped viruses differed in their susceptibility to irradiation treatment with adsorbed doses for inactivation of a target dose of 1 × 106 50% tissue culture infectious dose (TCID50)/mL ranging from 1 to 5 MRads. This finding seemed generally inversely correlated with genome size. Our data may help to guide other facilities in testing and verifying safe inactivation procedures.


Subject(s)
Gamma Rays , Virus Inactivation/radiation effects , Viruses/pathogenicity , Viruses/radiation effects , Flavivirus/radiation effects , Genome, Viral/radiation effects , Orthobunyavirus/radiation effects , Orthomyxoviridae/radiation effects , Virology/methods
6.
Chemosphere ; 214: 195-202, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30265926

ABSTRACT

We determined the potential interference of extracellular algal organic matter (EAOM) and intracellular algal organic matter (IAOM) extracted from Microcystis aeruginosa on MS2 bacteriophage inactivation under UV irradiation at two wavelengths (220 and 254 nm). UV irradiation at 220 nm doubled the inactivation rate of MS2 in water containing EAOM than in organic-free phosphate buffered solution. In contrast, EAOM did not change MS2 inactivation by exposure to UV 254 nm. The presence of IAOM did not significantly influence MS2 inactivation by exposure to either UV 254 or UV 220 nm. To achieve 3 log10 inactivation of MS2, UV254 nm required more than double the dose of UV220 nm (45 mJ/cm2 vs. 20 mJ/cm2). Linear correlations between the reduction in infectivity and the reduction in genome copies detected by reverse transcription quantitative polymerase chain reaction suggested that genomic damage is the main mechanism responsible for MS2 inactivation in water containing algal organic matter (AOM) by exposure to UV irradiation. These findings suggest that the presence of AOM did not negatively influence MS2 inactivation by either 220 or 254 nm irradiation, and that a lower UV dose of 220 nm irradiation can be used to achieve the same level of inactivation in water containing AOM.


Subject(s)
Levivirus/radiation effects , Microcystis/chemistry , Ultraviolet Rays , Virus Inactivation , Dose-Response Relationship, Radiation , Genome, Viral/radiation effects , Levivirus/drug effects , Water Purification/methods
7.
Food Environ Virol ; 6(4): 269-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25106777

ABSTRACT

The damage to a viral capsid after low-pressure (LP) and medium-pressure (MP) UV irradiation was assessed, using the quantitative or quantitative reverse transcription PCR coupled with ethidium monoazide treatment (EMA-PCR). After UV irradiation, adenovirus 5 (Ad5) and poliovirus 1 (PV1) were subjected to a plaque assay, PCR, and EMA-PCR to investigate the effect of UV irradiation on viral infectivity, genome damage, and capsid damage, respectively. The effectiveness of UV wavelengths in a viral genome and capsid damage of both PV1 and Ad5 was also further investigated using a band-pass filter. It was found that an MPUV lamp was more effective than an LPUV lamp in inactivating Ad5, whereas there was no difference in the case of PV1. The results of viral reduction determined by PCR and EMA-PCR indicated that MP UV irradiation damaged Ad5 capsid. The damage to PV1 and Ad5 capsid was also not observed after LP UV irradiation. The investigation of effects of UV wavelengths suggested that UV wavelengths at 230-245 nm have greater effects on adenovirus capsid in addition to viral genome than UV wavelengths beyond 245 nm.


Subject(s)
Adenoviruses, Human/radiation effects , Affinity Labels/pharmacology , Azides/pharmacology , Capsid/radiation effects , Disinfection/methods , Genome, Viral/radiation effects , Poliovirus/radiation effects , Adenoviruses, Human/growth & development , Adenoviruses, Human/metabolism , Adenoviruses, Human/pathogenicity , Animals , Capsid/metabolism , Cell Line , Chlorocebus aethiops , DNA, Viral/metabolism , DNA, Viral/radiation effects , Humans , Poliovirus/growth & development , Poliovirus/metabolism , Poliovirus/pathogenicity , Pressure , RNA, Viral/metabolism , RNA, Viral/radiation effects , Radiation Tolerance , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ultraviolet Rays , Viral Plaque Assay , Virus Inactivation/radiation effects
8.
Appl Environ Microbiol ; 79(11): 3455-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23542618

ABSTRACT

Viral disinfection kinetics have been studied in depth, but the molecular-level inactivation mechanisms are not understood. Consequently, it is difficult to predict the disinfection behavior of nonculturable viruses, even when related, culturable viruses are available. The objective of this work was to determine how small differences in the composition of the viral genome and proteins impact disinfection. To this end, we investigated the inactivation of three related bacteriophages (MS2, fr, and GA) by UV254, singlet oxygen ((1)O2), free chlorine (FC), and chlorine dioxide (ClO2). Genome damage was quantified by PCR, and protein damage was assessed by quantitative matrix-assisted laser desorption ionization (MALDI) mass spectrometry. ClO2 caused great variability in the inactivation kinetics between viruses and was the only treatment that did not induce genome damage. The inactivation kinetics were similar for all viruses when treated with disinfectants possessing a genome-damaging component (FC, (1)O2, and UV254). On the protein level, UV254 subtly damaged MS2 and fr capsid proteins, whereas GA's capsid remained intact. (1)O2 oxidized a methionine residue in MS2 but did not affect the other two viruses. In contrast, FC and ClO2 rapidly degraded the capsid proteins of all three viruses. Protein composition alone could not explain the observed degradation trends; instead, molecular dynamics simulations indicated that degradation is dictated by the solvent-accessible surface area of individual amino acids. Finally, despite the similarities of the three viruses investigated, their mode of inactivation by a single disinfectant varied. This explains why closely related viruses can exhibit drastically different inactivation kinetics.


Subject(s)
Disinfection/methods , Levivirus/growth & development , Virus Inactivation/drug effects , Virus Inactivation/radiation effects , Chlorine/pharmacology , Chlorine Compounds/pharmacology , Genome, Viral/drug effects , Genome, Viral/radiation effects , Kinetics , Levivirus/drug effects , Levivirus/genetics , Levivirus/radiation effects , Molecular Dynamics Simulation , Oxides/pharmacology , Singlet Oxygen/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Ultraviolet Rays
9.
Environ Sci Technol ; 46(18): 10022-30, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22913402

ABSTRACT

UV disinfection of viruses frequently leads to tailing after an initial exponential decay. Aggregation, light shielding, recombination, or resistant virus subpopulations have been proposed as explanations; however, none of these options has been conclusively demonstrated. This study investigates how aggregation affects virus inactivation by UV(254) in general, and the tailing phenomenon in particular. Bacteriophage MS2 was aggregated by lowering the solution pH before UV(254) disinfection. Aggregates were redispersed prior to enumeration to obtain the remaining fraction of individual infectious viruses. Results showed that initial inactivation kinetics were similar for viruses incorporated in aggregates (up to 1000 nm in radius) and dispersed viruses; however, aggregated viruses started to tail more readily than dispersed ones. Neither light shielding, nor the presence of resistant subpopulations could account for the tailing. Instead, tailing was consistent with recombination arising from the simultaneous infection of the host by several impaired viruses. We argue that UV(254) treatment of aggregates permanently fused a fraction of viruses, which increased the likelihood of multiple infection of a host cell and ultimately enabled the production of infective viruses via recombination.


Subject(s)
Disinfection/methods , Genome, Viral/radiation effects , Levivirus/genetics , Levivirus/radiation effects , Virus Inactivation/radiation effects , Water Purification/methods , Models, Biological , Recombination, Genetic/radiation effects , Ultraviolet Rays , Water Microbiology
10.
Cell Host Microbe ; 12(1): 117-124, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22817993

ABSTRACT

Although pathogen inactivation by γ-radiation is an attractive approach for whole-organism vaccine development, radiation doses required to ensure sterility also destroy immunogenic protein epitopes needed to mount protective immune responses. We demonstrate the use of a reconstituted manganous peptide complex from the radiation-resistant bacterium Deinococcus radiodurans to protect protein epitopes from radiation-induced damage and uncouple it from genome damage and organism killing. The Mn(2+) complex preserved antigenic structures in aqueous preparations of bacteriophage lambda, Venezuelan equine encephalitis virus, and Staphylococcus aureus during supralethal irradiation (25-40 kGy). An irradiated vaccine elicited both antibody and Th17 responses, and induced B and T cell-dependent protection against methicillin-resistant S. aureus (MRSA) in mice. Structural integrity of viruses and bacteria are shown to be preserved at radiation doses far above those which abolish infectivity. This approach could expedite vaccine production for emerging and established pathogens for which no protective vaccines exist.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Vaccines/radiation effects , Deinococcus/radiation effects , Epitopes/radiation effects , Peptides/chemistry , Animals , Bacteriophage lambda/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/radiation effects , Epitopes/immunology , Gamma Rays , Genome, Viral/radiation effects , Manganese/chemistry , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Peptides/radiation effects , Solutions , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/radiation effects , Staphylococcus aureus/immunology , Staphylococcus aureus/radiation effects , Th17 Cells/immunology , Viral Vaccines/immunology , Viral Vaccines/radiation effects
11.
Chembiochem ; 13(6): 837-45, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22416020

ABSTRACT

Much research has been dedicated to understanding the molecular basis of UV damage to biomolecules, yet many questions remain regarding the specific pathways involved. Here we describe a genome-mediated mechanism that causes site-specific virus protein cleavage upon UV irradiation. Bacteriophage MS2 was disinfected with 254 nm UV, and protein damage was characterized with ESI- and MALDI-based FT-ICR, Orbitrap, and TOF mass spectroscopy. Top-down mass spectrometry of the products identified the backbone cleavage site as Cys46-Ser47 in the virus capsid protein, a location of viral genome-protein interaction. The presence of viral RNA was essential to inducing backbone cleavage. The similar bacteriophage GA did not exhibit site-specific protein cleavage. Based on the major protein fragments identified by accurate mass analysis, a cleavage mechanism is proposed by radical formation. The mechanism involves initial oxidation of the Cys46 side chain followed by hydrogen atom abstraction from Ser47 C(α). Computational protein QM/MM studies confirmed the initial steps of the radical mechanism. Collectively, this study describes a rare incidence of genome-induced protein cleavage without the addition of sensitizers.


Subject(s)
Genome, Viral/radiation effects , Levivirus/metabolism , Levivirus/radiation effects , Viral Proteins/metabolism , Viral Proteins/radiation effects , Levivirus/genetics , Mass Spectrometry , Ultraviolet Rays , Viral Proteins/genetics
12.
Appl Environ Microbiol ; 72(12): 7671-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17041164

ABSTRACT

Several models (animal caliciviruses, poliovirus 1 [PV1], and F-specific RNA bacteriophages) are usually used to predict inactivation of nonculturable viruses. For the same UV fluence, viral inactivation observed in the literature varies from 0 to 5 logs according to the models and the methods (infectivity versus molecular biology). The lack of knowledge concerning the mechanisms of inactivation due to UV prevents us from selecting the best model. In this context, determining if viral genome degradation may explain the loss of infectivity under UV radiation becomes essential. Thus, four virus models (PV1 and three F-specific RNA phages: MS2, GA, and Qbeta) were exposed to UV radiation from 0 to 150 mJ.cm-2. PV1 is the least-resistant virus, while MS2 and GA phages are the most resistant, with phage Qbeta having an intermediate sensitivity; respectively, 6-log, 2.3-log, 2.5-log, and 4-log decreases for 50 mJ.cm-2. In parallel, analysis of RNA degradation demonstrated that this phenomenon depends on the fragment size for PV1 as well as for MS2. Long fragments (above 2,000 bases) for PV1 and MS2 fell rapidly to the background level (>1.3-log decrease) for 20 mJ.cm-2 and 60 mJ.cm-2, respectively. Nevertheless, the size of the viral RNA is not the only factor affecting UV-induced RNA degradation, since viral RNA was more rapidly degraded in PV1 than in the MS2 phage with a similar size. Finally, extrapolation of inactivation and UV-induced RNA degradation kinetics highlights that genome degradation could fully explain UV-induced viral inactivation.


Subject(s)
Allolevivirus/radiation effects , Genome, Viral/radiation effects , Levivirus/radiation effects , Poliovirus/radiation effects , Ultraviolet Rays , Virus Inactivation/radiation effects , Allolevivirus/pathogenicity , Animals , Cell Line , Dose-Response Relationship, Radiation , Humans , Levivirus/pathogenicity , Poliovirus/pathogenicity , Poliovirus Vaccine, Oral , RNA, Viral/radiation effects
13.
Blood ; 108(12): 3691-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16931631

ABSTRACT

Human cytomegalovirus (HCMV) establishes and maintains a latent infection in myeloid cells and can reactivate to cause serious disease in allograft recipients. To better understand the molecular events associated with the establishment of latency, we tracked the virus following infection of primary human myeloid progenitor cells at days 1, 2, 3, 5, and 11. At all time points, the viral genome was maintained in most cells at approximately 10 copies. Infectious virus was not detected, but virus could be reactivated by extended fibroblast coculture. In contrast to wild-type HCMV, the viral genome was rapidly lost from myeloid progenitors infected with ultraviolet (UV)-inactivated virus, suggesting viral gene expression was required for efficient establishment of latency. To identify viral genes associated with the establishment phase, RNA from each time point was interrogated using custom-made HCMV gene microarrays. Using this approach, we detected expression of viral RNAs at all time points. The pattern of expression differed from that which occurs during productive infection, and decreased over time. This study provides evidence that a molecular pathway into latency is associated with expression of a unique subset of viral transcripts. Viral genes expressed during the establishment phase may serve as targets for therapies to interrupt this process.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Gene Expression Regulation, Viral/physiology , Myeloid Progenitor Cells/virology , Virus Activation/physiology , Virus Latency/physiology , Cells, Cultured , Coculture Techniques , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/prevention & control , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Profiling/methods , Gene Expression Regulation, Viral/radiation effects , Genome, Viral/physiology , Genome, Viral/radiation effects , Humans , Myeloid Progenitor Cells/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Viral/metabolism , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Ultraviolet Rays , Ultraviolet Therapy/methods , Virus Activation/radiation effects , Virus Inactivation/radiation effects , Virus Latency/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...