1
|
Zhang X, Xu H, Sun R, Xiong G, Shi X. An insight into G-quadruplexes: Identification and potential therapeutic targets in livestock viruses. Eur J Med Chem 2024; 279:116848. [PMID: 39255642 DOI: 10.1016/j.ejmech.2024.116848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
G-quadruplexes (G4s) are non-canonical nucleic acids secondary structures that involve in the regulation of some key biological processes, such as replication, transcription, and translation. G4s have been extensively described in the genomes of human and related diseases. In recent years, G4s were identified in several livestock viruses, including those of the emerging epidemics, like Nipah virus (NiV). Since their discovery, G4s have been developed as the potential antiviral targets, and the employment of G4 ligands or interacting proteins has helped to expound the viral infectivity and pathogenesis through G4-mediated mechanisms, and highlight the potential as therapeutic approaches. However, the comprehensively studies of G4s in livestock viruses have not been summarized. This review delves into the reported literatures of G4s in livestock viruses, particular focus on the presence, biophysical identification, and possible function of G4s in viral genome, summarizing the G4 ligands, interacted proteins and aptamers on antiviral applications. The strengths and the challenges of G4 targeting in this field are also discussed. Therefore, this review will shed new light on the future development of highly potent and targeting antiviral therapy.
Collapse
Affiliation(s)
- Xianpeng Zhang
- Laboratory of Pesticide Toxicology and Pesticide Efficient Utilization, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China; Key Laboratory of Crop Physiology Ecology & Genetic Breeding, Jiangxi Agriculture University, Nanchang, Jiangxi Province, 330045, PR China
| | - Hongyu Xu
- College of Land Resources and Environment, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China
| | - Ranran Sun
- Laboratory of Pesticide Toxicology and Pesticide Efficient Utilization, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China
| | - Guihong Xiong
- Key Laboratory of Crop Physiology Ecology & Genetic Breeding, Jiangxi Agriculture University, Nanchang, Jiangxi Province, 330045, PR China
| | - Xugen Shi
- Laboratory of Pesticide Toxicology and Pesticide Efficient Utilization, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China; Key Laboratory of Crop Physiology Ecology & Genetic Breeding, Jiangxi Agriculture University, Nanchang, Jiangxi Province, 330045, PR China; Jiangxi Xiajiang Dry Direct-seeded Rice Science and Technology Backyard, Ji'an, Jiangxi Province, 331400, PR China.
| |
Collapse
|
2
|
A Comparison of Pseudorabies Virus Latency to Other A-Herpesvirinae Subfamily Members. Viruses 2022; 14:v14071386. [PMID: 35891367 PMCID: PMC9316381 DOI: 10.3390/v14071386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/12/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
Pseudorabies virus (PRV), the causative agent of Aujeszky’s disease, is one of the most important infectious pathogens threatening the global pig industry. Like other members of alphaherpesviruses, PRV establishes a lifelong latent infection and occasionally reactivates from latency after stress stimulus in infected pigs. Latent infected pigs can then serve as the source of recurrent infection, which is one of the difficulties for PRV eradication. Virus latency refers to the retention of viral complete genomes without production of infectious progeny virus; however, following stress stimulus, the virus can be reactivated into lytic infection, which is known as the latency-reactivation cycle. Recently, several research have indicated that alphaherpesvirus latency and reactivation is regulated by a complex interplay between virus, neurons, and the immune system. However, with those limited reports, the relevant advances in PRV latency are lagging behind. Therefore, in this review we focus on the regulatory mechanisms in PRV latency via summarizing the progress of PRV itself and that of other alphaherpesviruses, which will improve our understanding in the underlying mechanism of PRV latency and help design novel therapeutic strategies to control PRV latency.
Collapse
|
3
|
Deng J, Wu Z, Liu J, Ji Q, Ju C. The Role of Latency-Associated Transcripts in the Latent Infection of Pseudorabies Virus. Viruses 2022; 14:v14071379. [PMID: 35891360 PMCID: PMC9320458 DOI: 10.3390/v14071379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Pseudorabies virus (PRV) can cause neurological, respiratory, and reproductive diseases in pigs and establish lifelong latent infection in the peripheral nervous system (PNS). Latent infection is a typical feature of PRV, which brings great difficulties to the prevention, control, and eradication of pseudorabies. The integral mechanism of latent infection is still unclear. Latency-associated transcripts (LAT) gene is the only transcriptional region during latent infection of PRV which plays the key role in regulating viral latent infection and inhibiting apoptosis. Here, we review the characteristics of PRV latent infection and the transcriptional characteristics of the LAT gene. We also analyzed the function of non-coding RNA (ncRNA) produced by the LAT gene and its importance in latent infection. Furthermore, we provided possible strategies to solve the problem of latent infection of virulent PRV strains in the host. In short, the detailed mechanism of PRV latent infection needs to be further studied and elucidated.
Collapse
|
4
|
Hoffmann W, Lipińska AD, Bieńkowska-Szewczyk K. Functional Analysis of a Frontal miRNA Cluster Located in the Large Latency Transcript of Pseudorabies Virus. Viruses 2022; 14:v14061147. [PMID: 35746619 PMCID: PMC9227234 DOI: 10.3390/v14061147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 01/28/2023] Open
Abstract
MicroRNAs (miRNAs) have been identified as a class of crucial regulators of virus-host crosstalk, modulating such processes as viral replication, antiviral immune response, viral latency, and pathogenesis. Pseudorabies virus (PRV), a model for the study of alphaherpesvirus biology, codes for 11 distinct miRNAs mapped to the ~4.6 kb intron of Large Latency Transcript (LLT). Recent studies have revealed the role of clusters consisting of nine and eleven miRNA genes in the replication and virulence of PRV. The function of separate miRNA species in regulating PRV biology has not been thoroughly investigated. To analyze the regulatory potential of three PRV miRNAs located in the frontal cluster of the LLT intron, we generated a research model based on the constitutive expression of viral miRNAs in swine testis cells (ST_LLT [1–3] cell line). Using a cell culture system providing a stable production of individual miRNAs at high levels, we demonstrated that the LLT [1–3] miRNA cluster significantly downregulated IE180, EP0, and gE at the early stages of PRV infection. It was further determined that LLT [1–3] miRNAs could regulate the infection process, leading to a slight distortion in transmission and proliferation ability. Collectively, our findings indicate the potential of LLT [1–3] miRNAs to retard the host responses by reducing viral antigenic load and suppressing the expansion of progeny viruses at the early stages of infection.
Collapse
|
5
|
Shangguan A, Li J, Sun Y, Liu Z, Zhang S. Host-virus interactions in PK-15 cells infected with Pseudorabies virus Becker strain based on RNA-seq. Virus Res 2022; 318:198829. [DOI: 10.1016/j.virusres.2022.198829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
|
6
|
Li L, Wang R, Hu H, Chen X, Yin Z, Liang X, He C, Yin L, Ye G, Zou Y, Yue G, Tang H, Jia R, Song X. The antiviral activity of kaempferol against pseudorabies virus in mice. BMC Vet Res 2021; 17:247. [PMID: 34275451 PMCID: PMC8287772 DOI: 10.1186/s12917-021-02953-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 06/28/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Pseudorabies virus (PRV), a member of the Alphaherpesviruses, is one of the most important pathogens that harm the global pig industry. Accumulated evidence indicated that PRV could infect humans under certain circumstances, inducing severe clinical symptoms such as acute human encephalitis. Currently, there are no antiviral drugs to treat PRV infections, and vaccines available only for swine could not provide full protection. Thus, new control measures are urgently needed. RESULTS In the present study, kaempferol exhibited anti-PRV activity in mice through improving survival rate by 22.22 %, which was higher than acyclovir (Positive control) with the survival rate of 16.67 % at 6 days post infection (dpi); meanwhile, the survival rate was 0 % at 6 dpi in the infected-untreated group. Kaempferol could inhibit the virus replication in the brain, lung, kidney, heart and spleen, especially the viral gene copies were reduced by over 700-fold in the brain, which was further confirmed by immunohistochemical examination. The pathogenic changes induced by PRV infection in these organs were also alleviated. The transcription of the only immediate-early gene IE180 in the brain was significantly inhibited by kaempferol, leading to the decreased transcriptional levels of the early genes (EPO and TK). The expression of latency-associated transcript (LAT) was also inhibited in the brain, which suggested that kaempferol could inhibit PRV latency. Kaempferol-treatment could induce higher levels of IL-1β, IL-4, IL-6, TNF-α and IFN-γ in the serum at 3 dpi which were then declined to normal levels at 5 dpi. CONCLUSIONS These results suggested that kaempferol was expected to be a new alternative control measure for PRV infection.
Collapse
Affiliation(s)
- Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Rui Wang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Huaiyue Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Xu Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Guizhou Yue
- College of Science, Sichuan Agricultural University, 625014, Ya'an, China
| | - Huaqiao Tang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Renyong Jia
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, 611130, Chengdu, China.
| |
Collapse
|
7
|
Savoret J, Mesnard JM, Gross A, Chazal N. Antisense Transcripts and Antisense Protein: A New Perspective on Human Immunodeficiency Virus Type 1. Front Microbiol 2021; 11:625941. [PMID: 33510738 PMCID: PMC7835632 DOI: 10.3389/fmicb.2020.625941] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
It was first predicted in 1988 that there may be an Open Reading Frame (ORF) on the negative strand of the Human Immunodeficiency Virus type 1 (HIV-1) genome that could encode a protein named AntiSense Protein (ASP). In spite of some controversy, reports began to emerge some years later describing the detection of HIV-1 antisense transcripts, the presence of ASP in transfected and infected cells, and the existence of an immune response targeting ASP. Recently, it was established that the asp gene is exclusively conserved within the pandemic group M of HIV-1. In this review, we summarize the latest findings on HIV-1 antisense transcripts and ASP, and we discuss their potential functions in HIV-1 infection together with the role played by antisense transcripts and ASPs in some other viruses. Finally, we suggest pathways raised by the study of antisense transcripts and ASPs that may warrant exploration in the future.
Collapse
Affiliation(s)
- Juliette Savoret
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier, France
| | - Jean-Michel Mesnard
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier, France
| | - Antoine Gross
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier, France
| | - Nathalie Chazal
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, Montpellier, France
| |
Collapse
|
8
|
Lu JJ, Yuan WZ, Zhu YP, Hou SH, Wang XJ. Latent pseudorabies virus infection in medulla oblongata from quarantined pigs. Transbound Emerg Dis 2020; 68:543-551. [PMID: 32615031 DOI: 10.1111/tbed.13712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/09/2020] [Accepted: 06/27/2020] [Indexed: 12/15/2022]
Abstract
Pseudorabies virus (PRV) is a major pathogen in pig husbandry and is also a risk to human well-being. Pigs with latent PRV infection carry the virus lifelong, and it can be activated under conducive conditions. This poses a very important challenge to the control of the virus and may even prevent its elimination. To investigate latent infection with wild-type (wt) PRV, and also infection due to the use of live attenuated vaccines on farms, 80 pigs from two large-scale swine operations were traced. At 6 months old, the quarantined pigs were slaughtered and brain samples were collected. A PCR assay targeting the gB and gE genes was developed to detect PRV DNA fragments in medulla oblongata. Five of the samples (6.3%) were gB and gE gene fragment double-positive, 60 of the samples (75%) were gB single-positive, and 15 samples (18.7%) showed double-negative. A portion of latency-associated transcripts (LATs), EP0 mRNA, were found to be present in the gB gene fragment positive samples. Furthermore, the five double-positive samples were transmitted blindly, and apparent cytopathic effects were found in three of the five samples in the fourth generation. By means of Western blotting, PCR and sequencing, two of the isolated viruses were found to be related to vaccine strain Bartha-K61. Another was closely related to domestic epidemic strains HN1201 and LA and relatively unrelated to other Asian isolates. These results suggest that the live vaccines are latently present in brains, in a manner similar to wt PRV, and this poses potential safety issues in the pig husbandry industry. Wt PRV and live vaccine viruses were found to co-exist in pigs, demonstrating that the live vaccines were unable to confer complete sterilizing immunity, which may explain outbreaks of pseudorabies on vaccinated farms.
Collapse
Affiliation(s)
- Jin-Jin Lu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wan-Zhe Yuan
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yong-Ping Zhu
- Agricultural Bureau of Wuzhong District, Suzhou, China
| | - Shao-Hua Hou
- Beijing Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
9
|
Maturana CJ, Verpeut JL, Pisano TJ, Dhanerawala ZM, Esteves A, Enquist LW, Engel EA. Small Alphaherpesvirus Latency-Associated Promoters Drive Efficient and Long-Term Transgene Expression in the CNS. Mol Ther Methods Clin Dev 2020; 17:843-857. [PMID: 32368565 PMCID: PMC7191541 DOI: 10.1016/j.omtm.2020.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
Recombinant adeno-associated viruses (rAAVs) are used as gene therapy vectors to treat central nervous system (CNS) diseases. Despite their safety and broad tropism, important issues need to be corrected such as the limited payload capacity and the lack of small gene promoters providing long-term, pan-neuronal transgene expression in the CNS. Commonly used gene promoters are relatively large and can be repressed a few months after CNS transduction, risking the long-term performance of single-dose gene therapy applications. We used a whole-CNS screening approach based on systemic delivery of AAV-PHP.eB, iDisco+ tissue-clearing and light-sheet microscopy to identify three small latency-associated promoters (LAPs) from the herpesvirus pseudorabies virus (PRV). These promoters are LAP1 (404 bp), LAP2 (498 bp), and LAP1_2 (880 bp). They drive chronic transcription of the virus-encoded latency-associated transcript (LAT) during productive and latent phases of PRV infection. We observed stable, pan-neuronal transgene transcription and translation from AAV-LAPs in the CNS for 6 months post AAV transduction. In several CNS areas, the number of cells expressing the transgene was higher for LAP2 than the large conventional EF1α promoter (1,264 bp). Our data suggest that the LAPs are suitable candidates for viral vector-based CNS gene therapies requiring chronic transgene expression after one-time viral-vector administration.
Collapse
Affiliation(s)
- Carola J. Maturana
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Jessica L. Verpeut
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Thomas J. Pisano
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Zahra M. Dhanerawala
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Andrew Esteves
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lynn W. Enquist
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Esteban A. Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
10
|
Identification and characterization of G-quadruplex formation within the EP0 promoter of pseudorabies virus. Sci Rep 2018; 8:14029. [PMID: 30232344 PMCID: PMC6145870 DOI: 10.1038/s41598-018-32222-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
EP0 is an important early gene that modulates the life cycle of pseudorabies virus (PRV). A guanine-rich sequence overlapping with three Sp1 binding sites is located upstream of the transcription start site (TSS) in the EP0 promoter. Using native polyacrylamide gel electrophoresis (PAGE) and circular dichroism (CD), we verified that the G-rich region in the EP0 promoter forms an intramolecular parallel G-quadruplex (G4) in the presence of K+ ions. Further dimethyl sulphate (DMS) footprinting and Taq polymerase stop assays indicates the potential polymorphic folding of G4. In addition, a small chemical ligand, pyridostatin (PDS), promotes and stabilizes the formation of G4. Interestingly, based on the results of electrophoretic mobility shift assays (EMSA), the Sp1 protein bound to G4-bearing DNA with more affinity than DNA lacking the G4 structure. According to the luciferase reporter assay, G4 negatively regulates the EP0 promoter activity. These results demonstrate that Sp1 and G4 cooperate to regulate EP0 promoter activity.
Collapse
|
11
|
Wang X, Zhang MM, Yan K, Tang Q, Wu YQ, He WB, Chen HC, Liu ZF. The full-length microRNA cluster in the intron of large latency transcript is associated with the virulence of pseudorabies virus. Virology 2018; 520:59-66. [DOI: 10.1016/j.virol.2018.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/28/2022]
|
12
|
A spliced latency-associated VZV transcript maps antisense to the viral transactivator gene 61. Nat Commun 2018; 9:1167. [PMID: 29563516 PMCID: PMC5862956 DOI: 10.1038/s41467-018-03569-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/23/2018] [Indexed: 01/16/2023] Open
Abstract
Varicella-zoster virus (VZV), an alphaherpesvirus, establishes lifelong latent infection in the neurons of >90% humans worldwide, reactivating in one-third to cause shingles, debilitating pain and stroke. How VZV maintains latency remains unclear. Here, using ultra-deep virus-enriched RNA sequencing of latently infected human trigeminal ganglia (TG), we demonstrate the consistent expression of a spliced VZV mRNA, antisense to VZV open reading frame 61 (ORF61). The spliced VZV latency-associated transcript (VLT) is expressed in human TG neurons and encodes a protein with late kinetics in productively infected cells in vitro and in shingles skin lesions. Whereas multiple alternatively spliced VLT isoforms (VLTly) are expressed during lytic infection, a single unique VLT isoform, which specifically suppresses ORF61 gene expression in co-transfected cells, predominates in latently VZV-infected human TG. The discovery of VLT links VZV with the other better characterized human and animal neurotropic alphaherpesviruses and provides insights into VZV latency. Varicella-zoster virus (VZV) establishes lifelong infection in the majority of the population, but mechanisms underlying latency remain unclear. Here, the authors use ultra-deep RNA sequencing, enriched for viral RNAs, of latently infected human trigeminal ganglia and identify a spliced, latency-associated VZV mRNA.
Collapse
|
13
|
Csabai Z, Takács IF, Snyder M, Boldogkői Z, Tombácz D. Evaluation of the impact of ul54 gene-deletion on the global transcription and DNA replication of pseudorabies virus. Arch Virol 2017; 162:2679-2694. [PMID: 28577213 PMCID: PMC5927779 DOI: 10.1007/s00705-017-3420-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/23/2017] [Indexed: 01/28/2023]
Abstract
Pseudorabies virus (PRV) is an animal alphaherpesvirus with a wide host range. PRV has 67 protein-coding genes and several non-coding RNA molecules, which can be classified into three temporal groups, immediate early, early and late classes. The ul54 gene of PRV and its homolog icp27 of herpes simplex virus have a multitude of functions, including the regulation of viral DNA synthesis and the control of the gene expression. Therefore, abrogation of PRV ul54 function was expected to exert a significant effect on the global transcriptome and on DNA replication. Real-time PCR and real-time RT-PCR platforms were used to investigate these presumed effects. Our analyses revealed a drastic impact of the ul54 mutation on the genome-wide expression of PRV genes, especially on the transcription of the true late genes. A more than two hour delay was observed in the onset of DNA replication, and the amount of synthesized DNA molecules was significantly decreased in comparison to the wild-type virus. Furthermore, in this work, we were able to successfully demonstrate the utility of long-read SMRT sequencing for genotyping of mutant viruses.
Collapse
Affiliation(s)
- Zsolt Csabai
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, 6720, Hungary
| | - Irma F Takács
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, 6720, Hungary
| | - Michael Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Zsolt Boldogkői
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, 6720, Hungary.
| | - Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, 6720, Hungary.
| |
Collapse
|
14
|
Age-Dependent Differences in Pseudorabies Virus Neuropathogenesis and Associated Cytokine Expression. J Virol 2017; 91:JVI.02058-16. [PMID: 27852848 DOI: 10.1128/jvi.02058-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/03/2016] [Indexed: 11/20/2022] Open
Abstract
The severity of clinical symptoms induced by pseudorabies virus (PRV) infection of its natural host is inversely related to the age of the pig. During this study, 2- and 15-week-old pigs were inoculated with PRV strain NIA3. This resulted in important clinical disease, although the associated morbidity and mortality were lower in older pigs. Quantitative PCR analysis of viral DNA in different organs confirmed the general knowledge on PRV pathogenesis. Several new findings and potential explanations for the observed age-dependent differences in virulence, however, were determined from the study of viral and cytokine mRNA expression at important sites of neuropathogenesis. First, only limited viral and cytokine mRNA expression was detected in the nasal mucosa, suggesting that other sites may serve as the primary replication site. Second, PRV reached the trigeminal ganglion (TG) and brain stem rapidly upon infection but, compared to 2-week-old pigs, viral replication was less pronounced in 15-week-old pigs, and the decrease in viral mRNA expression was not preceded by or associated with an increased cytokine expression. Third, extensive viral replication associated with a robust expression of cytokine mRNA was detected in the olfactory bulbs of pigs from both age categories and correlated with the observed neurological disease. Our results suggest that age-dependent differences in PRV-induced clinical signs are probably due to enhanced viral replication and associated immunopathology in immature TG and the central nervous system neurons of 2-week-old pigs and that neurological disease is related with extensive viral replication and an associated immune response in the olfactory bulb. IMPORTANCE It is well known that alphaherpesvirus infections of humans and animals result in more severe clinical disease in newborns than in older individuals and that this is probably related to differences in neuropathogenesis. The underlying mechanisms, however, remain unclear. Pseudorabies virus infection of its natural host, the pig, provides a suitable infection model to study this more profoundly. We show here that the severe neurological disease observed in 2-week-old pigs does not appear to be related to a hampered innate immune response but is more likely to reflect the immature development state of the trigeminal ganglia (TG) and central nervous system (CNS) neurons, resulting in an inefficient suppression of viral replication. In 15-week-old pigs, viral replication was efficiently suppressed in the TG and CNS without induction of an extensive immune response. Furthermore, our results provide evidence that neurological disease could, at least in part, be related to viral replication and associated immunopathology in the olfactory bulb.
Collapse
|
15
|
Tombácz D, Csabai Z, Oláh P, Balázs Z, Likó I, Zsigmond L, Sharon D, Snyder M, Boldogkői Z. Full-Length Isoform Sequencing Reveals Novel Transcripts and Substantial Transcriptional Overlaps in a Herpesvirus. PLoS One 2016; 11:e0162868. [PMID: 27685795 PMCID: PMC5042381 DOI: 10.1371/journal.pone.0162868] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/30/2016] [Indexed: 11/19/2022] Open
Abstract
Whole transcriptome studies have become essential for understanding the complexity of genetic regulation. However, the conventionally applied short-read sequencing platforms cannot be used to reliably distinguish between many transcript isoforms. The Pacific Biosciences (PacBio) RS II platform is capable of reading long nucleic acid stretches in a single sequencing run. The pseudorabies virus (PRV) is an excellent system to study herpesvirus gene expression and potential interactions between the transcriptional units. In this work, non-amplified and amplified isoform sequencing protocols were used to characterize the poly(A+) fraction of the lytic transcriptome of PRV, with the aim of a complete transcriptional annotation of the viral genes. The analyses revealed a previously unrecognized complexity of the PRV transcriptome including the discovery of novel protein-coding and non-coding genes, novel mono- and polycistronic transcription units, as well as extensive transcriptional overlaps between neighboring and distal genes. This study identified non-coding transcripts overlapping all three replication origins of the PRV, which might play a role in the control of DNA synthesis. We additionally established the relative expression levels of gene products. Our investigations revealed that the whole PRV genome is utilized for transcription, including both DNA strands in all coding and intergenic regions. The genome-wide occurrence of transcript overlaps suggests a crosstalk between genes through a network formed by interacting transcriptional machineries with a potential function in the control of gene expression.
Collapse
Affiliation(s)
- Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
- Department of Genetics, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Zsolt Csabai
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Oláh
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zsolt Balázs
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - István Likó
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Laura Zsigmond
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Donald Sharon
- Department of Genetics, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Michael Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Zsolt Boldogkői
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
- * E-mail:
| |
Collapse
|
16
|
Oláh P, Tombácz D, Póka N, Csabai Z, Prazsák I, Boldogkői Z. Characterization of pseudorabies virus transcriptome by Illumina sequencing. BMC Microbiol 2015; 15:130. [PMID: 26129912 PMCID: PMC4487798 DOI: 10.1186/s12866-015-0470-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/19/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pseudorabies virus is a widely-studied model organism of the Herpesviridae family, with a compact genome arrangement of 72 known coding sequences. In order to obtain an up-to-date genetic map of the virus, a combination of RNA-sequencing approaches were applied, as recent advancements in high-throughput sequencing methods have provided a wealth of information on novel RNA species and transcript isoforms, revealing additional layers of transcriptome complexity in several viral species. RESULTS The total RNA content and polyadenylation landscape of pseudorabies virus were characterized for the first time at high coverage by Illumina high-throughput sequencing of cDNA samples collected during the lytic infectious cycle. As anticipated, nearly all of the viral genome was transcribed, with the exception of loci in the large internal and terminal repeats, and several small intergenic repetitive sequences. Our findings included a small novel polyadenylated non-coding RNA near an origin of replication, and the single-base resolution mapping of 3' UTRs across the viral genome. Alternative polyadenylation sites were found in a number of genes and a novel alternative splice site was characterized in the ep0 gene, while previously known splicing events were confirmed, yielding no alternative splice isoforms. Additionally, we detected the active polyadenylation of transcripts earlier believed to be transcribed as part of polycistronic RNAs. CONCLUSION To the best of our knowledge, the present work has furnished the highest-resolution transcriptome map of an alphaherpesvirus to date, and reveals further complexities of viral gene expression, with the identification of novel transcript boundaries, alternative splicing of the key transactivator EP0, and a highly abundant, novel non-coding RNA near the lytic replication origin. These advances provide a detailed genetic map of PRV for future research.
Collapse
Affiliation(s)
- Péter Oláh
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - Nándor Póka
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - Zsolt Csabai
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - István Prazsák
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| | - Zsolt Boldogkői
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
17
|
Xiang K, Cheng Y, Zhou M, Sun L, Ji Y, Wang Y, Zhang B, Luo Y, Ju C. Production of monoclonal antibody against EP0 protein of pseudorabies virus and determination of its recognized epitope. Monoclon Antib Immunodiagn Immunother 2014; 33:409-13. [PMID: 25545210 DOI: 10.1089/mab.2014.0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Early protein 0 (EP0) is especially important for modulating PRV gene expression and reactivation from the latent state, but the mechanisms have not been elucidated. In this study, six monoclonal antibodies (MAbs) against EP0 protein of PRV were generated and their characterizations were investigated. Western blot analysis showed all six MAbs could react with immunizing antigen, but only 2B12 and 2C6 could react with native EP0 protein from PRV-infected cells. ELISA additivity tests revealed that at least three epitopes in EP0 were defined by six MAbs. The epitope recognized by MAb 2B12 was further identified in 287-292 aa of EP0 protein using a series of expressed overlapping peptides. These MAbs may provide valuable tools for further research of the functions of EP0 in PRV infection.
Collapse
Affiliation(s)
- Keyu Xiang
- 1 College of Veterinary Medicine, South China Agricultural University , Guangzhou, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
A 2.5-kilobase deletion containing a cluster of nine microRNAs in the latency-associated-transcript locus of the pseudorabies virus affects the host response of porcine trigeminal ganglia during established latency. J Virol 2014; 89:428-42. [PMID: 25320324 DOI: 10.1128/jvi.02181-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED The alphaherpesvirus pseudorabies virus (PrV) establishes latency primarily in neurons of trigeminal ganglia when only the transcription of the latency-associated transcript (LAT) locus is detected. Eleven microRNAs (miRNAs) cluster within the LAT, suggesting a role in establishment and/or maintenance of latency. We generated a mutant (M) PrV deleted of nine miRNA genes which displayed properties that were almost identical to those of the parental PrV wild type (WT) during propagation in vitro. Fifteen pigs were experimentally infected with either WT or M virus or were mock infected. Similar levels of virus excretion and host antibody response were observed in all infected animals. At 62 days postinfection, trigeminal ganglia were excised and profiled by deep sequencing and quantitative RT-PCR. Latency was established in all infected animals without evidence of viral reactivation, demonstrating that miRNAs are not essential for this process. Lower levels of the large latency transcript (LLT) were found in ganglia infected by M PrV than in those infected by WT PrV. All PrV miRNAs were expressed, with highest expression observed for prv-miR-LLT1, prv-miR-LLT2 (in WT ganglia), and prv-miR-LLT10 (in both WT and M ganglia). No evidence of differentially expressed porcine miRNAs was found. Fifty-four porcine genes were differentially expressed between WT, M, and control ganglia. Both viruses triggered a strong host immune response, but in M ganglia gene upregulation was prevalent. Pathway analyses indicated that several biofunctions, including those related to cell-mediated immune response and the migration of dendritic cells, were impaired in M ganglia. These findings are consistent with a function of the LAT locus in the modulation of host response for maintaining a latent state. IMPORTANCE This study provides a thorough reference on the establishment of latency by PrV in its natural host, the pig. Our results corroborate the evidence obtained from the study of several LAT mutants of other alphaherpesviruses encoding miRNAs from their LAT regions. Neither PrV miRNA expression nor high LLT expression levels are essential to achieve latency in trigeminal ganglia. Once latency is established by PrV, the only remarkable differences are found in the pattern of host response. This indicates that, as in herpes simplex virus, LAT functions as an immune evasion locus.
Collapse
|
19
|
Tombácz D, Tóth JS, Boldogkoi Z. Effects of deletion of the early protein 0 gene of pseudorabies virus on the overall viral gene expression. Gene 2012; 493:235-42. [PMID: 22178766 DOI: 10.1016/j.gene.2011.11.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 08/17/2011] [Accepted: 11/14/2011] [Indexed: 11/29/2022]
Abstract
Real-time RT-PCR analysis was applied to evaluate the impact of deletion of the early protein 0 (EP0) gene of pseudorabies virus (PRV) on the global expression of the viral transcripts during lytic infection in cultured porcine kidney cells. Our analysis showed that EP0 exerted an inhibitory effect on the transcription of the PRV genes in the early stage of infection, and alternating stimulatory and inhibitory effects on the viral gene expressions in the late stage of infection. The data also suggested that a general function of EP0 might be to reverse the kinetics of expression of early viral genes. We also observed that EP0 facilitated the development of correlations in the transcription kinetics between the immediate early 180 gene and the PRV transcripts, indicating that a major function of EP0 could be to modify the effects of the IE180 protein on the PRV transcriptome.
Collapse
Affiliation(s)
- Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. st. 4., Szeged, H-6720, Hungary.
| | | | | |
Collapse
|
20
|
Pseudorabies virus infected porcine epithelial cell line generates a diverse set of host microRNAs and a special cluster of viral microRNAs. PLoS One 2012; 7:e30988. [PMID: 22292087 PMCID: PMC3264653 DOI: 10.1371/journal.pone.0030988] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 12/29/2011] [Indexed: 12/29/2022] Open
Abstract
Pseudorabies virus (PRV) belongs to Alphaherpesvirinae subfamily that causes huge economic loss in pig industry worldwide. It has been recently demonstrated that many herpesviruses encode microRNAs (miRNAs), which play crucial roles in viral life cycle. However, the knowledge about PRV-encoded miRNAs is still limited. Here, we report a comprehensive analysis of both viral and host miRNA expression profiles in PRV-infected porcine epithelial cell line (PK-15). Deep sequencing data showed that the ∼4.6 kb intron of the large latency transcript (LLT) functions as a primary microRNA precursor (pri-miRNA) that encodes a cluster of 11 distinct miRNAs in the PRV genome, and 209 known and 39 novel porcine miRNAs were detected. Viral miRNAs were further confirmed by stem-loop RT-PCR and northern blot analysis. Intriguingly, all of these viral miRNAs exhibited terminal heterogeneity both at the 5′ and 3′ ends. Seven miRNA genes produced mature miRNAs from both arms and two of the viral miRNA genes showed partially overlapped in their precursor regions. Unexpectedly, a terminal loop-derived small RNA with high abundance and one special miRNA offset RNA (moRNA) were processed from a same viral miRNA precursor. The polymorphisms of viral miRNAs shed light on the complexity of host miRNA-processing machinery and viral miRNA-regulatory mechanism. The swine genes and PRV genes were collected for target prediction of the viral miRNAs, revealing a complex network formed by both host and viral genes. GO enrichment analysis of host target genes suggests that PRV miRNAs are involved in complex cellular pathways including cell death, immune system process, metabolic pathway, indicating that these miRNAs play significant roles in virus-cells interaction of PRV and its hosts. Collectively, these data suggest that PRV infected epithelial cell line generates a diverse set of host miRNAs and a special cluster of viral miRNAs, which might facilitate PRV replication in cells.
Collapse
|
21
|
Antifungal activity of 2α,3β-functionalized steroids stereoselectively increases with the addition of oligosaccharides. Bioorg Med Chem Lett 2011; 21:7379-86. [PMID: 22047693 DOI: 10.1016/j.bmcl.2011.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 11/21/2022]
Abstract
Invasive fungal infections pose a significant problem to the immune-compromised. Moreover, increased resistance to common antifungals requires development of novel compounds that can be used to treat invasive fungal infections. Naturally occurring steroidal glycosides have been shown to possess a range of functional antimicrobial properties, but synthetic methodology for their development hinders thorough exploration of this class of molecules and the structural components required for broad spectrum antifungal activity. In this report, we outline a novel approach to the synthesis of glycoside-linked functionalized 2α,3β-cholestane and spirostane molecules and present data from in vitro screenings of the antifungal activities against human fungal pathogens and as well as mammalian cell toxicity of these derivatives.
Collapse
|
22
|
Tóth JS, Tombácz D, Takács IF, Boldogkoi Z. The effects of viral load on pseudorabies virus gene expression. BMC Microbiol 2010; 10:311. [PMID: 21134263 PMCID: PMC3016322 DOI: 10.1186/1471-2180-10-311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 12/06/2010] [Indexed: 05/08/2023] Open
Abstract
Background Herpesvirus genes are classified into distinct kinetic groups on the basis of their expression dynamics during lytic growth of the virus in cultured cells at a high, typically 10 plaque-forming units/cell multiplicity of infection (MOI). It has been shown that both the host response and the success of a pathogen are dependent on the quantity of particles infecting an organism. This work is a continuation of an earlier study [1], in which we characterized the overall expression of PRV genes following low-MOI infection. In the present study, we have addressed the question of whether viral gene expressions are dependent on the multiplicity of infection by comparing gene expressions under low and high-MOI conditions. Results In the present study, using a real-time RT-PCR assay, we address the question of whether the expression properties of the pseudorabies virus (PRV) genes are dependent on the number of virion particles infecting a single cell in a culture. Our analysis revealed a significant dependence of the gene expression on the MOI in most of these genes. Specifically, we found that most of the examined viral genes were expressed at a lower level at a low MOI (0.1) than at a high MOI (10) experiment in the early stage of infection; however, this trend reversed by six hour post-infection in more than half of the genes. Furthermore, in the high-MOI infection, several PRV genes substantially declined within the 4 to 6-h infection period, which was not the case in the low-MOI infection. In the low-MOI infection, the level of antisense transcript (AST), transcribed from the antiparallel DNA strand of the immediate-early 180 (ie180) gene, was comparable to that of ie180 mRNA, while in the high-MOI experiment (despite the 10 times higher copy number of the viral genome in the infected cells) the amount of AST dropped by more than two log values at the early phase of infection. Furthermore, our analysis suggests that adjacent PRV genes are under a common regulation. This is the first report on the effect of the multiplicity of infection on genome-wide gene expression of large DNA viruses, including herpesviruses. Conclusion Our results show a strong dependence of the global expression of PRV genes on the MOI. Furthermore, our data indicate a strong interrelation between the expressions of ie180 mRNA and AST, which determines the expression properties of the herpesvirus genome and possibly the replication strategy (lytic or latent infection) of the virus in certain cell types.
Collapse
Affiliation(s)
- Judit S Tóth
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Hungary
| | | | | | | |
Collapse
|
23
|
Tombácz D, Tóth JS, Petrovszki P, Boldogkoi Z. Whole-genome analysis of pseudorabies virus gene expression by real-time quantitative RT-PCR assay. BMC Genomics 2009; 10:491. [PMID: 19852823 PMCID: PMC2775753 DOI: 10.1186/1471-2164-10-491] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 10/23/2009] [Indexed: 01/26/2023] Open
Abstract
Background Pseudorabies virus (PRV), a neurotropic herpesvirus of pigs, serves as an excellent model system with which to investigate the herpesvirus life cycle both in cultured cells and in vivo. Real-time RT-PCR is a very sensitive, accurate and reproducible technique that can be used to detect very small amounts of RNA molecules, and it can therefore be applied for analysis of the expression of herpesvirus genes from the very early period of infection. Results In this study, we have developed and applied a quantitative reverse transcriptase-based real-time PCR technique in order to profile transcription from the whole genome of PRV after lytic infection in porcine kidney cells. We calculated the relative expression ratios in a novel way, which allowed us to compare different PRV genes with respect to their expression dynamics, and to divide the PRV genes into distinct kinetic classes. This is the first publication on the whole-genome analysis of the gene expression of an alpha-herpesvirus by qRT2-PCR. We additionally established the kinetic properties of uncharacterized PRV genes and revised or confirmed data on PRV genes earlier examined by traditional methods such as Northern blot analysis. Our investigations revealed that genes with the same expression properties form clusters on the PRV genome: nested overlapping genes belong in the same kinetic class, while most convergent genes belong in different kinetic classes. Further, we detected inverse relationships as concerns the expressions of EP0 and IE180 mRNAs and their antisense partners. Conclusion Most (if not all) PRV genes begin to be expressed from the onset of viral expression. No sharp boundary was found between the groups of early and late genes classified on the basis of their requirement for viral DNA synthesis. The expressions of the PRV genes were analyzed, categorized and compared by qRT2-PCR assay, with the average of the minimum cycle threshold used as a control for the calculation of a particular R value. In principle, this new calculation technique is applicable for the analysis of gene expression in all temporally changing genetic systems.
Collapse
Affiliation(s)
- Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, H-6720, Hungary.
| | | | | | | |
Collapse
|
24
|
Sorrell I, White A, Pedersen AB, Hails RS, Boots M. The evolution of covert, silent infection as a parasite strategy. Proc Biol Sci 2009; 276:2217-26. [PMID: 19324776 PMCID: PMC2677597 DOI: 10.1098/rspb.2008.1915] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Many parasites and pathogens cause silent/covert infections in addition to the more obvious infectious disease-causing pathology. Here, we consider how assumptions concerning superinfection, protection and seasonal host birth and transmission rates affect the evolution of such covert infections as a parasite strategy. Regardless of whether there is vertical infection or effects on sterility, overt infection is always disadvantageous in relatively constant host populations unless it provides protection from superinfection. If covert infections are protective, all individuals will enter the covert stage if there is enough vertical transmission, and revert to overt infections after a ‘latent’ period (susceptible, exposed, infected epidemiology). Seasonal variation in transmission rates selects for non-protective covert infections in relatively long-lived hosts with low birth rates typical of many mammals. Variable host population density caused by seasonal birth rates may also select for covert transmission, but in this case it is most likely in short-lived fecund hosts. The covert infections of some insects may therefore be explained by their outbreak population dynamics. However, our models consistently predict proportions of covert infection, which are lower than some of those observed in nature. Higher proportions of covert infection may occur if there is a direct link between covert infection and overt transmission success, the covert infection is protective or the covert state is the result of suppression by the host. Relatively low proportions of covert transmission may, however, be explained as a parasite strategy when transmission opportunities vary.
Collapse
Affiliation(s)
- Ian Sorrell
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| | | | | | | | | |
Collapse
|
25
|
Characterization of pseudorabies virus (PrV) cleavage-encapsidation proteins and functional complementation of PrV pUL32 by the homologous protein of herpes simplex virus type 1. J Virol 2009; 83:3930-43. [PMID: 19193798 DOI: 10.1128/jvi.02636-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cleavage and encapsidation of newly replicated herpes simplex virus type 1 (HSV-1) DNA requires several essential viral gene products that are conserved in sequence within the Herpesviridae. However, conservation of function has not been analyzed in greater detail. For functional characterization of the UL6, UL15, UL28, UL32, and UL33 gene products of pseudorabies virus (PrV), the respective deletion mutants were generated by mutagenesis of the virus genome cloned as a bacterial artificial chromosome (BAC) in Escherichia coli and propagated in transgenic rabbit kidney cells lines expressing the deleted genes. Neither of the PrV mutants was able to produce plaques or infectious progeny in noncomplementing cells. DNA analyses revealed that the viral genomes were replicated but not cleaved into monomers. By electron microscopy, only scaffold-containing immature but not DNA-containing mature capsids were detected in the nuclei of noncomplementing cells infected with either of the mutants. Remarkably, primary envelopment of empty capsids at the nuclear membrane occasionally occurred, and enveloped tegument-containing light particles were formed in the cytoplasm and released into the extracellular space. Immunofluorescence analyses with monospecific antisera of cells transfected with the respective expression plasmids indicated that pUL6, pUL15, and pUL32 were able to enter the nucleus. In contrast, pUL28 and pUL33 were predominantly found in the cytoplasm. Only pUL6 could be unequivocally identified and localized in PrV-infected cells and in purified virions, whereas the low abundance or immunogenicity of the other proteins hampered similar studies. Yeast two-hybrid analyses revealed physical interactions between the PrV pUL15, pUL28, and pUL33 proteins, indicating that, as in HSV-1, a tripartite protein complex might catalyze cleavage and encapsidation of viral DNA. Whereas the pUL6 protein is supposed to form the portal for DNA entry into the capsid, the precise role of the UL32 gene product during this process remains to be elucidated. Interestingly, the defect of UL32-negative PrV could be completely corrected in trans by the homologous protein of HSV-1, demonstrating similar functions. However, trans-complementation of UL32-negative HSV-1 by the PrV protein was not observed.
Collapse
|
26
|
Transcriptional suppression of IE180 and TK promoters by the EP0 of pseudorabies virus strains Ea and Fa. Virus Genes 2009; 38:269-75. [DOI: 10.1007/s11262-008-0320-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 12/18/2008] [Indexed: 11/26/2022]
|
27
|
Gray WL. Simian varicella in old world monkeys. Comp Med 2008; 58:22-30. [PMID: 19793453 PMCID: PMC2703154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Revised: 10/03/2007] [Accepted: 10/25/2007] [Indexed: 05/28/2023]
Abstract
Simian varicella virus (SVV) causes a natural erythematous disease in Old World monkeys and is responsible for simian varicella epizootics that occur sporadically in facilities housing nonhuman primates. This review summarizes the biology of SVV and simian varicella as a veterinary disease of nonhuman primates. SVV is closely related to varicella-zoster virus, the causative agent of human varicella and herpes zoster. Clinical signs of simian varicella include fever, vesicular skin rash, and hepatitis. Simian varicella may range from a mild infection to a severe and life-threatening disease, and epizootics may have high morbidity and mortality rates. SVV establishes a lifelong latent infection in neural ganglia of animals in which the primary disease resolves, and the virus may reactivate later in life to cause a secondary disease corresponding to herpes zoster. Prompt diagnosis is important for control and prevention of epizootics. Antiviral treatment for simian varicella may be effective if administered early in the course of infection.
Collapse
Affiliation(s)
- Wayne L Gray
- Department of Microbiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
28
|
Ou Y, Davis KA, Traina-Dorge V, Gray WL. Simian varicella virus expresses a latency-associated transcript that is antisense to open reading frame 61 (ICP0) mRNA in neural ganglia of latently infected monkeys. J Virol 2007; 81:8149-56. [PMID: 17507490 PMCID: PMC1951321 DOI: 10.1128/jvi.00407-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Simian varicella virus (SVV) and varicella-zoster virus (VZV) are closely related alphaherpesviruses that cause varicella (chickenpox) in nonhuman primates and humans, respectively. After resolution of the primary disease, SVV and VZV establish latent infection of neural ganglia and may later reactivate to cause a secondary disease (herpes zoster). This study investigated SVV gene expression in neural ganglia derived from latently infected vervet monkeys. SVV transcripts were detected in neural ganglia, but not in liver or lung tissues, of latently infected animals. A transcript mapping to open reading frame (ORF) 61 (herpes simplex virus type 1 [HSV-1] ICP0 homolog) was consistently detected in latently infected trigeminal, cervical, and lumbar ganglia by reverse transcriptase PCR. Further analysis confirmed that this SVV latency-associated transcript (LAT) was oriented antisense to the gene 61 mRNA. SVV ORF 21 transcripts were also detected in 42% of neural ganglia during latency. In contrast, SVV ORF 28, 29, 31, 62, and 63 transcripts were not detected in ganglia, liver, or lung tissues of latently infected animals. The results demonstrate that viral gene expression is limited during SVV latency and that a LAT antisense to an ICP0 homolog is expressed. In this regard, SVV gene expression during latency is similar to that of HSV-1 and other neurotropic animal alphaherpesviruses but differs from that reported for VZV.
Collapse
Affiliation(s)
- Yang Ou
- Dept. of Microbiology and Immunology, Slot 511, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA
| | | | | | | |
Collapse
|
29
|
Pomeranz LE, Reynolds AE, Hengartner CJ. Molecular biology of pseudorabies virus: impact on neurovirology and veterinary medicine. Microbiol Mol Biol Rev 2005; 69:462-500. [PMID: 16148307 PMCID: PMC1197806 DOI: 10.1128/mmbr.69.3.462-500.2005] [Citation(s) in RCA: 649] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pseudorabies virus (PRV) is a herpesvirus of swine, a member of the Alphaherpesvirinae subfamily, and the etiological agent of Aujeszky's disease. This review describes the contributions of PRV research to herpesvirus biology, neurobiology, and viral pathogenesis by focusing on (i) the molecular biology of PRV, (ii) model systems to study PRV pathogenesis and neurovirulence, (iii) PRV transsynaptic tracing of neuronal circuits, and (iv) veterinary aspects of pseudorabies disease. The structure of the enveloped infectious particle, the content of the viral DNA genome, and a step-by-step overview of the viral replication cycle are presented. PRV infection is initiated by binding to cellular receptors to allow penetration into the cell. After reaching the nucleus, the viral genome directs a regulated gene expression cascade that culminates with viral DNA replication and production of new virion constituents. Finally, progeny virions self-assemble and exit the host cells. Animal models and neuronal culture systems developed for the study of PRV pathogenesis and neurovirulence are discussed. PRV serves asa self-perpetuating transsynaptic tracer of neuronal circuitry, and we detail the original studies of PRV circuitry mapping, the biology underlying this application, and the development of the next generation of tracer viruses. The basic veterinary aspects of pseudorabies management and disease in swine are discussed. PRV infection progresses from acute infection of the respiratory epithelium to latent infection in the peripheral nervous system. Sporadic reactivation from latency can transmit PRV to new hosts. The successful management of PRV disease has relied on vaccination, prevention, and testing.
Collapse
Affiliation(s)
- Lisa E Pomeranz
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08540, USA.
| | | | | |
Collapse
|
30
|
BONSALL MB, SAIT SM, HAILS RS. Invasion and dynamics of covert infection strategies in structured insect-pathogen populations. J Anim Ecol 2005. [DOI: 10.1111/j.1365-2656.2005.00945.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Kim SK, Albrecht RA, O'Callaghan DJ. A negative regulatory element (base pairs -204 to -177) of the EICP0 promoter of equine herpesvirus 1 abolishes the EICP0 protein's trans-activation of its own promoter. J Virol 2004; 78:11696-706. [PMID: 15479811 PMCID: PMC523287 DOI: 10.1128/jvi.78.21.11696-11706.2004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The early EICP0 protein is a powerful trans-activator that activates all classes of equine herpesvirus 1 (EHV-1) promoters but, unexpectedly, trans-activates its own promoter very weakly. Transient transfection assays that employed constructs harboring deletions within the EICP0 promoter indicated that EICP0 cis-acting sequences within bp -224 to -158 relative to the first ATG abolished the EICP0 protein's trans-activation of its own promoter. When inserted into the promoters of other EHV-1 genes, this sequence also downregulated activation of the immediate-early IE(-169/+73), early thymidine kinase TK(-215/+97), and late glycoprotein K gK(-83/+14) promoters, indicating that the cis-acting sequence (-224 to -158) downregulated expression of representative promoters of all classes of EHV-1 genes and contains a negative regulatory element (NRE). To define the cis-acting element(s), three synthetic oligonucleotides (Na [bp -224 to -195], Nb [bp -204 to -177], and Nc [bp -185 to -156]) were synthesized and cloned upstream of the EICP0(-157/-21) promoter. Of the three synthetic sequences, only the Nb oligonucleotide caused the downregulation of the EICP0 promoter. The NRE was identified as a 28-bp element to lie at -204 to -177 that encompassed the sequence of ([-204]AGATACAGATGTTCGATAAATTGGAACC[-177]). Gel shift assays performed with mouse L-M, rabbit RK-13, and human HeLa cell nuclear extracts and gamma-(32)P-labeled wild-type and mutant NREs demonstrated that a ubiquitous nuclear protein(s) (NRE-binding protein, NREBP) binds specifically to a sequence (bp -193 to -183) in the NRE. The NREBP is also present in the nucleus of EHV-1-infected cells; however, the amount of NREBP in EHV-1-infected L-M cells that bound to the Nb oligonucleotide was reduced compared to that in uninfected L-M cells. Transient transfection assays showed that deletions or mutations within the NREBP-binding site abolished the NRE activity of the EICP0 promoter. These results suggested that the NREBP may mediate the NRE activity of the EICP0 promoter and may function in the coordinate expression of EHV-1 genes.
Collapse
Affiliation(s)
- Seong K Kim
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, P.O. Box 33932, Shreveport, LA 71130-3932, USA
| | | | | |
Collapse
|
32
|
Boldogköi Z, Sík A, Dénes A, Reichart A, Toldi J, Gerendai I, Kovács KJ, Palkovits M. Novel tracing paradigms--genetically engineered herpesviruses as tools for mapping functional circuits within the CNS: present status and future prospects. Prog Neurobiol 2004; 72:417-45. [PMID: 15177785 DOI: 10.1016/j.pneurobio.2004.03.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2003] [Accepted: 03/29/2004] [Indexed: 11/17/2022]
Abstract
The mammalian CNS is composed of an extremely complex meshwork of highly ordered interconnections among billions of neurons. To understand the diverse functions of this neuronal network we need to differentiate between functionally related and nonrelated elements. A powerful labeling method for defining intricate neural circuits is based on the utilization of neurotropic herpesviruses, including pseudorabies virus and herpes simplex virus type 1. The recent development of genetically engineered tracing viruses can open the way toward the conception of novel tract-tracing paradigms. These new-generation tracing viruses may facilitate the clarification of problems, which were inaccessible to earlier approaches. This article first presents a concise review of the general aspects of neuroanatomical tracing protocols. Subsequently, it discusses the molecular biology of alpha-herpesviruses, and the genetic manipulation and gene expression techniques that are utilized for the construction of virus-based tracers. Finally, it describes the current utilization of genetically modified herpesviruses for circuit analysis, and the future directions in their potential applications.
Collapse
Affiliation(s)
- Zsolt Boldogköi
- Laboratory of Neuromorphology, Department of Anatomy, Faculty of Medicine, Semmelweis University and Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- Sebastian Maurer-Stroh
- IMP-Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria.
| | | |
Collapse
|
34
|
Klupp BG, Hengartner CJ, Mettenleiter TC, Enquist LW. Complete, annotated sequence of the pseudorabies virus genome. J Virol 2004; 78:424-40. [PMID: 14671123 PMCID: PMC303424 DOI: 10.1128/jvi.78.1.424-440.2004] [Citation(s) in RCA: 246] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We have obtained the complete DNA sequence of pseudorabies virus (PRV), an alphaherpesvirus also known as Aujeszky's disease virus or suid herpesvirus 1, using sequence fragments derived from six different strains (Kaplan, Becker, Rice, Indiana-Funkhauser, NIA-3, and TNL). The assembled PRV genome sequence comprises 143,461 nucleotides. As expected, it matches the predicted gene arrangement, genome size, and restriction enzyme digest patterns. More than 70 open reading frames were identified with homologs in related alphaherpesviruses; none were unique to PRV. RNA polymerase II transcriptional control elements in the PRV genome, including core promoters, splice sites, and polyadenylation sites, were identified with computer prediction programs. The correlation between predicted and experimentally determined transcription start and stop sites was excellent. The transcriptional control architecture is characterized by three key features: core transcription elements shared between genes, yielding divergent transcripts and a large number of coterminal transcripts; bifunctional transcriptional elements, yielding head-to-tail transcripts; and short repetitive sequences that could function as insulators against improperly terminated transcripts. Many of these features are conserved in the alphaherpesvirus subfamily and have important implications for gene array analyses.
Collapse
Affiliation(s)
- Barbara G Klupp
- Institute of Molecular Biology, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, D-17493 Greifswald-Insel Riems, Germany
| | | | | | | |
Collapse
|
35
|
Gilden DH, Cohrs RJ, Mahalingam R. Clinical and molecular pathogenesis of varicella virus infection. Viral Immunol 2004; 16:243-58. [PMID: 14583142 DOI: 10.1089/088282403322396073] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Varicella zoster virus (VZV) is a neurotropic human herpesvirus that infects nearly all humans and causes chickenpox (varicella). After chickenpox, VZV becomes latent in cranial nerve, dorsal root, and autonomic nervous system ganglia along the entire neuraxis. Virus reactivation produces shingles (zoster), characterized by pain and rash usually restricted to 1-3 dermatomes. Zoster is often complicated by postherpetic neuralgia (PHN), pain that persists for months to years after rash resolves. Virus may also spread to the spinal cord and blood vessels of the brain, producing a unifocal or multifocal vasculopathy, particularly in immunocompromised individuals. The increased incidence of zoster in elderly and immunocompromised individuals appears to be due to a VZV-specific host immunodeficiency. PHN may reflect a chronic VZV ganglionitis, and VZV vasculopathy is due to productive virus infection in cerebral arteries. Strategies that might boost host cell-mediated immunity to VZV are discussed, as well as the physical state of viral nucleic acid during latency and the possible mechanisms by which herpesvirus latency is maintained and virus is reactivated. A current summary of varicella latency and pathogenesis produced by simian varicella virus (SVV), the counterpart of human VZV, points to the usefulness of a primate model of natural infection to study varicella latency, as well as the experimental model of intratracheal inoculation to study the effectiveness of antiviral agents in driving persistent varicella virus into a latent state.
Collapse
Affiliation(s)
- Donald H Gilden
- Department of Neurology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | |
Collapse
|
36
|
Taharaguchi S, Yoshino S, Amagai K, Ono E. The latency-associated transcript promoter of pseudorabies virus directs neuron-specific expression in trigeminal ganglia of transgenic mice. J Gen Virol 2003; 84:2015-2022. [PMID: 12867631 DOI: 10.1099/vir.0.19080-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The latency-associated transcript (LAT) promoter of pseudorabies virus (PRV) is unique among viral promoters in that it remains active in trigeminal ganglia during the latent state. It is not known which the viral or host proteins regulate expression of the PRV LAT gene in latently infected neurons. To determine whether host transcriptional proteins in neurons can regulate the PRV LAT promoter in vivo, three transgenic mouse lines containing the PRV LAT promoter (LAP; LAP1 and LAP2) linked to the chloramphenicol acetyltransferase (CAT) gene were generated. All of the transgenic mouse lines, in the absence of the viral proteins, displayed strong expression of the transgene in trigeminal ganglia in addition to other neuronal tissues such as cerebral cortex, cerebellum, hippocampus and olfactory bulb. Expression of the transgene in neurons of trigeminal ganglia was demonstrated by in situ hybridization. These data provide direct evidence that neuronal transcription factors are sufficient to activate the PRV LAP in vivo and that the promoter is neuron-specific.
Collapse
Affiliation(s)
- Satoshi Taharaguchi
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Saori Yoshino
- G-in Techno Science, Sapporo 001-0015, Japan
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Keiko Amagai
- Sankyo Labo Service Corporation, Tokyo 132-0023, Japan
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Etsuro Ono
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| |
Collapse
|
37
|
Burden JP, Nixon CP, Hodgkinson AE, Possee RD, Sait SM, King LA, Hails RS. Covert infections as a mechanism for long-term persistence of baculoviruses. Ecol Lett 2003. [DOI: 10.1046/j.1461-0248.2003.00459.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
38
|
Kim SK, Jang HK, Albrecht RA, Derbigny WA, Zhang Y, O'Callaghan DJ. Interaction of the equine herpesvirus 1 EICP0 protein with the immediate-early (IE) protein, TFIIB, and TBP may mediate the antagonism between the IE and EICP0 proteins. J Virol 2003; 77:2675-85. [PMID: 12552007 PMCID: PMC141080 DOI: 10.1128/jvi.77.4.2675-2685.2003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The equine herpesvirus 1 (EHV-1) immediate-early (IE) and EICP0 proteins are potent trans-activators of EHV-1 promoters; however, in transient-transfection assays, the IE protein inhibits the trans-activation function of the EICP0 protein. Assays with IE mutant proteins revealed that its DNA-binding domain, TFIIB-binding domain, and nuclear localization signal may be important for the antagonism between the IE and EICP0 proteins. In vitro interaction assays with the purified IE and EICP0 proteins indicated that these proteins interact directly. At late times postinfection, the IE and EICP0 proteins colocalized in the nuclei of infected equine cells. Transient-transfection assays showed that the EICP0 protein trans-activated EHV-1 promoters harboring only a minimal promoter region (TATA box and cap site), suggesting that the EICP0 protein trans-activates EHV-1 promoters by interactions with general transcription factor(s). In vitro interaction assays revealed that the EICP0 protein interacted directly with the basal transcription factors TFIIB and TBP and that the EICP0 protein (amino acids [aa] 143 to 278) mediated the interaction with aa 125 to 174 of TFIIB. Our unpublished data showed that the IE protein interacts with the same domain (aa 125 to 174) of TFIIB and with TBP. Taken together, these results suggested that interaction of the EICP0 protein with the IE protein, TFIIB, and TBP may mediate the antagonism between the IE and EICP0 proteins.
Collapse
Affiliation(s)
- Seong K Kim
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | | | | | | | | | | |
Collapse
|
39
|
Boldogköi Z, Reichart A, Tóth IE, Sik A, Erdélyi F, Medveczky I, Llorens-Cortes C, Palkovits M, Lenkei Z. Construction of recombinant pseudorabies viruses optimized for labeling and neurochemical characterization of neural circuitry. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 109:105-18. [PMID: 12531520 DOI: 10.1016/s0169-328x(02)00546-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this study we have modified the neuroinvasiveness of pseudorabies virus strain Bartha, a commonly utilized trans-synaptic tract-tracer. In addition, we sought to facilitate detection of cellular mRNAs in neurons infected with the virus. In order to modify spreading characteristics, we inserted the lacZ or the GFP (green fluorescent protein) genes into the genomic loci containing the putative latency-associated transcript promoter (P(LAT2)), resulting in the disruption of the promoter function. Following rat kidney injection, mutant viruses labeled central autonomic neurons in a slower and much more restricted manner than the parent Bartha strain. Since both reporter genes were controlled by the human cytomegalovirus immediate early (IE) 1 promoter, they exhibited IE expression kinetics. This property proved to be important for the co-detection of reporter proteins with neuronal mRNAs, readily detected at early but not at late stage of infection, as shown in tyrosine-hydroxylase expressing A5 catecholaminergic neurons and in serotonin transporter expressing raphe magnus neurons.
Collapse
Affiliation(s)
- Zsolt Boldogköi
- Laboratory of Neuromorphology, Department of Anatomy, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ou CJ, Wong ML, Huang C, Chang TJ. Suppression of promoter activity of the LAT gene by IE180 of pseudorabies virus. Virus Genes 2002; 25:227-39. [PMID: 12881635 DOI: 10.1023/a:1020959521745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The latency-associated transcript (LAT) gene is the only viral genomic region that is abundantly transcribed during pseudorabies virus (PrV) latent infection. The mechanism of reactivation of PrV from latency remains unknown. To analyze the regulation mechanism of the LAT promoter, we constructed a series of recombinant vectors in which various sequences upstream of LAT were linked to the chloramphenicol acetyltransferase (CAT) gene. Transcriptional efficiency was examined by cotransfection with plasmids carrying the PrV IE, EP0, or gD gene, respectively. Results showed that the activity of PrV LAT promoter was dramatically repressed by the IE180 protein and a TATA box and a putative IE180 binding site within the promoter were involved in this repression. To dissect the functional domains of IE180, we compared the relative repressive abilities of IE180 variants to the LAT promoter by transient transfection assays. Mutational analysis demonstrated that almost the whole IE180 (amino acid residues 1-1440) are essential for its repression to LAT promoter. To explore the possible mechanism of repression, an electrophoretic mobility shift assay (EMSA) using nuclear extracts from neuronal cells was performed and formation of protein-DNA complexes between IE180 and the oligonucleotide probe (-46 to -19, relative to the start site of LAT transcription) was demonstrated. The association of IE180 with the region encompassing the putative IE180 binding site and the TATA box upstream of PrV LAT gene was further confirmed by supershift of EMSA complexes using IE180 specific antibody. Thus, our results suggested that IE180 repressed the LAT promoter via an interaction between IE180, LAT promoter and cellular protein(s).
Collapse
Affiliation(s)
- Chia-Jen Ou
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung 402, Taiwan, ROC
| | | | | | | |
Collapse
|
41
|
Ou CJ, Wong ML, Chang TJ. A TEF-1-element is required for activation of the promoter of pseudorabies virus glycoprotein X gene by IE180. Virus Genes 2002; 25:241-53. [PMID: 12881636 DOI: 10.1023/a:1020915706724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The pseudorabies virus (PRV) immediate-early regulatory protein IE180 is able to transactivate the viral early and late genes. Using chloramphenicol acetyltransferase (CAT) assay, we investigated the transactivation function of IE180 to the promoter of PRV glycoprotein X (gX) gene, and our results showed that IE180 could significantly increase the expression of CAT gene which was under the control of gX promoter. To further identify the activation domains of IE180 protein that interact with the gX promoter sequences, various truncated mutants of IE180 gene and gX promoter gene were constructed and analyzed by CAT and gel retardation assay. Results revealed that the N-terminal amino acid residues from 133 to 736 of IE180 could interact with the binding site of transcriptional enhancer factor-1 (TEF-1) that resides in the gX promoter. Formation of protein-DNA complexes between the IE180 protein and the TEF-1 element of the gX promoter was observed using electrophoretic mobility shift assay (EMSA) as well as Southwestern blot analysis. These results indicated that a direct interaction occurred between IE180 and the TEF-1 element; and this interaction was abolished if the TEF-1 element was mutated. The association of IE180 with the TEF-1 element was further confirmed by the supershift of EMSA complexes using IE180 specific antibody. Taken together, our results suggested that formation of a complex between the IE180 protein and TEF-1 element in the gX promoter region was involved in the transcriptional regulation of the gX gene.
Collapse
Affiliation(s)
- Chia-Jen Ou
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung 402, Taiwan, ROC
| | | | | |
Collapse
|
42
|
Chang YY, Wong ML, Lin HW, Chang TJ. Cloning and regulation of the promoter of pseudorabies virus (TNL strain) glycoprotein E gene. Virus Genes 2002; 24:235-41. [PMID: 12086144 DOI: 10.1023/a:1015376431948] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The nucleotide sequence upstream to the glycoprotein E (gE) gene of pseudorabies virus (PrV, TNL strain) was cloned from the genomic virus DNA by polymerase chain reaction (PCR) and its DNA sequences were determined. The DNA segment, which was supposed to contain the gE promoter, was subcloned into a chloramphenicol acetyltransferase (CAT) reporter gene and the resulting plasmid was named pgEp-B-CAT. To examine the promoter function of this upstream sequence of gE gene, we transfected pgEp-B-CAT DNA into L-M cells and the promoter activity was analyzed by CAT assay. Results showed that our DNA fragment could exhibit promoter activity. Furthermore, we transfected L-M cells with pgEp-B-CAT for 48 h, then superinfected cells with pseudorabies virus, and performed CAT assay. It was found that PrV superinfection could slightly enhance the activity of gE promoter, suggesting that factors produced during viral infection could stimulate the promoter. To explore the possible mechanism of regulation at transcriptional level, the pgEp-B-CAT plasmid were cotransfected with eukaryotic vectors expressing viral regulatory proteins IE or EP0, and results indicated that the gE promoter was activated by IE protein whereas it was inhibited by EP0 protein. Moreover, the effect of exogenous IE or EP0 on the protein level of gE in PrV-infected cells was examined; conclusion similar to that of CAT assay were obtained.
Collapse
Affiliation(s)
- Yuan-Yen Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | | | | | | |
Collapse
|
43
|
Boldogköi Z, Szabó A, Vrbová G, Nógrádi A. Pseudorabies virus-based gene delivery to rat embryonic spinal cord grafts. Hum Gene Ther 2002; 13:719-29. [PMID: 11936971 DOI: 10.1089/104303402317322285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The construction and application of recombinant pseudorabies viruses (PrVs) for the delivery of beta-galactosidase and/or green fluorescent protein (GFP) genes to rat embryonic spinal cord cells are reported here. These viruses were specifically designed to infect embryonic spinal cord neurons, which can be grafted into a lesioned spinal cord in order to restore the lost functions of the host cord. The recombinant viruses were constructed in two steps. The small subunit of the ribonucleotide reductase (RR) gene was first abolished by a frameshift mutation and an expression cassette containing the lacZ gene alone or together with the GFP gene was then inserted in place of the early protein 0 (EP0) gene of PrV. The reporter gene cassettes were positioned downstream from the PrV latency-associated promoter. Using an ex vivo system, we infected embryonic spinal cord explants with these viruses and found that neither vRREP0lac nor vRREP0lacgfp exerted any cytotoxic effect at all. It was also revealed that these viruses infect embryonic cells with high efficiency, and that infected neurons grafted into the spinal cord express the inserted reporter genes for periods of up to 12 weeks. This system offers a new approach for foreign gene transfer to neurons grafted into the CNS.
Collapse
Affiliation(s)
- Zsolt Boldogköi
- Laboratory of Neuromorphology, Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary
| | | | | | | |
Collapse
|
44
|
Taharaguchi S, Kobayashi T, Yoshino S, Ono E. Analysis of regulatory functions for the region located upstream from the latency-associated transcript (LAT) promoter of pseudorabies virus in cultured cells. Vet Microbiol 2002; 85:197-208. [PMID: 11852187 DOI: 10.1016/s0378-1135(01)00513-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The latency-associated transcript (LAT) promoter of pseudorabies virus (PrV) is unique among the many promoters of the viral genome in that it remains active during the latent state. The regulatory mechanism of PrV LAT gene expression is complex and different between latency and lytic infection of cultured cells. Although two different sequences, LAP1 and LAP2, are thought to be involved in LAT gene expression, the function of the upstream region of the LAT promoter (LAP1 and LAP2) remains an enigma, even in cultured cells. To analyze the function of the upstream region, it is necessary to examine the effects of the upstream sequence on LAT gene expression in the absence of other viral proteins. Transient expression assays were performed by employing a series of reporter plasmids in which various sequences upstream of the LAT promoter (from nucleotide positions -592 to +423 relative to the transcriptional start site of the large latency transcript (LLT)) were linked to the chloramphenicol acetyltransferase (CAT) gene in cells of neuronal and non-neuronal origin. We identified a region (from nucleotide positions -3606 to -1386) that was capable of repressing the LAT promoter activity in Vero cells by analyzing CAT gene expression of the series of reporter plasmids. This effect was not observed in Neuro-2a cells. We have also shown that the LAT promoter activity of the reporter plasmid containing the upstream region was repressed by the immediate-early gene product IE180 in Vero cells, but not in Neuro-2a cells. These results suggest that the upstream region of the LAT promoter may have a role in repressing LAT gene expression in cultured non-neuronal cells.
Collapse
Affiliation(s)
- Satoshi Taharaguchi
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic, Medicine, Hokkaido University, 060-0815, Sapporo, Japan
| | | | | | | |
Collapse
|
45
|
Borchers K, Field HJ. Neuronal latency in human and animal herpesvirus infections. Curr Top Microbiol Immunol 2001; 253:61-94. [PMID: 11417140 DOI: 10.1007/978-3-662-10356-2_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- K Borchers
- Institut für Virologie, Freie Universität Berlin, Königin-Luise-Strasse 49, 14195 Berlin, Germany.
| | | |
Collapse
|
46
|
Tasaki T, Taharaguchi S, Kobayashi T, Yoshino S, Ono E. Inhibition of pseudorabies virus replication by a dominant-negative mutant of early protein 0 expressed in a tetracycline-regulated system. Vet Microbiol 2001; 78:195-203. [PMID: 11165064 DOI: 10.1016/s0378-1135(00)00301-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pseudorabies virus (PRV) early protein 0 (EP0) consisting of 410 amino acids is a transactivator of viral genes. A mutant consisting of amino acids 1-113 exhibits dominant-negative properties. In order to assess the antiviral potential of the EP0 mutant, Vero cells were transformed with the EP0 mutant gene expressed in a tetracycline-regulated system. The transformed cell lines showed marked resistance to PRV infection when expression of the EP0 mutant gene was induced. In the transformed cell line infected with PRV, synthesis of the immediate-early protein (IE180) and of EP0 was inhibited, whereas the levels of IE and EP0 messenger RNA (mRNA) were not decreased, as compared with those of the control cell line. The present results suggest that the EP0 mutant may not alter the efficiency of the viral gene transcription but rather translation efficiency of the viral mRNA.
Collapse
Affiliation(s)
- T Tasaki
- Laboratory of Animal Experiment for Disease Model, Institute for Genetic Medicine, Hokkaido University, 060-0815, Sapporo, Japan
| | | | | | | | | |
Collapse
|
47
|
Boldogköi Z, Braun A, Fodor I. Replication and virulence of early protein 0 and long latency transcript deficient mutants of the Aujeszky's disease (pseudorabies) virus. Microbes Infect 2000; 2:1321-8. [PMID: 11018448 DOI: 10.1016/s1286-4579(00)01285-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Early protein 0 (EP0)-deficient recombinant Aujeszky's disease viruses, Ka-ep0lac and Ba-ep0lac derived from strains Kaplan and Bartha, respectively, were constructed to explore the impact of the mutation on replication, virulence and latency of the virus. Inactivation of the EP0 gene resulted in a mutation of long latency transcript (Cheung et al., 1991) that is located on the complementary DNA strand of EP0 and immediate early protein (IE)175 genes. In infection of immortalized porcine kidney cells, the growth rate and yield of both EP0(-) mutant strains were significantly smaller than that of wild-type virus. Ka-ep0lac was found to be highly virulent, while Ba-ep0lac showed an attenuated phenotype in mice. PCR assay and immunohistochemistry showed that the Ba-ep0lac virus was able to establish latency in the mouse trigeminal ganglia. However, latent virus was not able to reactivate in explant reactivation assays. Accordingly, latent Ba-ep0lac has the potential to be exploited as vectors for the delivery of foreign genes to the nervous system.
Collapse
Affiliation(s)
- Z Boldogköi
- Laboratory of Molecular Virology, Agricultural Biotechnology Center, Gödöllö, 2100 Hungary
| | | | | |
Collapse
|
48
|
Jin L, Schnitzlein WM, Scherba G. Identification of the pseudorabies virus promoter required for latency-associated transcript gene expression in the natural host. J Virol 2000; 74:6333-8. [PMID: 10864643 PMCID: PMC112139 DOI: 10.1128/jvi.74.14.6333-6338.2000] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Expression of the latency-associated transcript (LAT) gene is a hallmark of alphaherpesvirus latency, and yet its control and function remain an enigma. Resolution of this problem will require verification and subsequent elimination or disabling of elements regulating LAT gene transcription so that the influence of the resultant RNA can be evaluated. Toward this end, we generated a novel pseudorabies virus (PrV) recombinant in which a 282-bp region containing the LAP1 (first latency-active promoter) consensus sequence was replaced by a reporter cassette. Despite this substitution, replication of the recombinant was comparable to that of the parental and rescuant viruses both in cultured mammalian cells and in the natural host, swine. Furthermore, production of the LAT gene-associated 2.0- and 8.0-kb RNAs during an in vitro lytic infection of cultured neuronal cells was unaffected. However, the otherwise constitutively produced and processed 8.4-kb LAT was not detected in porcine trigeminal ganglia latently infected with this novel recombinant, although the viral genome was shown to be present. Therefore, LAP1 is apparently the basal promoter for PrV LAT gene expression during viral latency but is not required for such activity during an in vitro lytic infection of neuronal cells. More importantly, the ability of PrV to persist in a latent state in the absence of LAT suggests that other factors are responsible for this event in the natural host.
Collapse
Affiliation(s)
- L Jin
- Department of Veterinary Pathobiology, University of Illinois, Urbana, Illinois 61802, USA
| | | | | |
Collapse
|
49
|
Fuchs W, Ehrlich C, Klupp BG, Mettenleiter TC. Characterization of the replication origin (Ori(S)) and adjoining parts of the inverted repeat sequences of the pseudorabies virus genome. J Gen Virol 2000; 81:1539-43. [PMID: 10811937 DOI: 10.1099/0022-1317-81-6-1539] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The DNA sequence of a 2.4 kbp fragment located in the internal and terminal inverted repeat sequences of the pseudorabies virus genome determined in this study closes a gap between the previously described genes for the ICP4 and ICP22 homologues. The novel sequence contains no conserved herpesvirus open reading frames. Northern blot and cDNA analyses revealed a viral immediate-early transcript of 1.8 kb, which is spliced by the removal of two small introns close to its 5' end and which presumably represents the mRNA of the downstream open reading frame encoding the ICP22 homologue. Upstream of the transcribed region, an imperfect set of three directly repeated sequences was identified. Each of them contains a complementary pair of the alphaherpesvirus origin-binding protein recognition motif GTTCGCAC, spaced by AT-rich sequences. In vitro studies confirmed that the DNA fragment analysed includes a functional origin of viral DNA replication.
Collapse
Affiliation(s)
- W Fuchs
- Institute of Molecular Biology, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, D-17498 Insel Riems, Germany.
| | | | | | | |
Collapse
|
50
|
Boldogköi Z, Erdélyi F, Fodor I. A putative latency promoter/enhancer (P(LAT2)) region of pseudorabies virus contains a virulence determinant. J Gen Virol 2000; 81:415-20. [PMID: 10644840 DOI: 10.1099/0022-1317-81-2-415] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Contradictory data have recently been reported on the role of the unique long-internal repeat junction area of pseudorabies (Aujeszky's disease) virus (PrV) genome in the virulence of the virus. To investigate the basis of the difference, four recombinant PrVs mutated at the outer region of inverted repeats that involved a putative latency promoter (P(LAT2)) were constructed in this study. Propagation characteristics of mutant viruses in cultured cells were similar to those of the wild-type virus. However, a 757 bp deletion at this location caused significant reduction in the virulence of PrV after intraperitoneal inoculation of mice and a moderate decrease in the virulence after intracranial inoculation. These results indicate that the P(LAT2) region is an important virulence determinant that may be implicated in the neuroinvasive capability of the virus.
Collapse
Affiliation(s)
- Z Boldogköi
- Institute for Biochemistry, Laboratory of Gene Technology, Agricultural Biotechnology Center, PO Box 411, H-2101 Gödöllö, Hungary
| | | | | |
Collapse
|