1
|
Samoilenko M, Nedosekov V, Bertoni G. Testing the Tenacity of Small Ruminant Lentiviruses In Vitro to Assess the Potential Risk of Indirect Fomites' Transmission. Viruses 2025; 17:419. [PMID: 40143344 PMCID: PMC11946479 DOI: 10.3390/v17030419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
In 2011-2013, we isolated and characterized small ruminant lentiviruses (SRLVs) from two flocks, one of goats and the other of sheep, that had never been in direct contact. Phylogenetic analysis of these viruses indicated a common origin, which led us to hypothesize indirect transmission of these viruses between the two flocks. Since, to our knowledge, there are no published data on the tenacity of these viruses, we started this work. In the first part, we monitored the loss of infectivity of two prototypic SRLV strains, MVV 1514 and CAEV-CO, over time, in liquid suspension. As expected, the suspensions stored at 4 °C better preserved the infectivity of the viruses. Additionally, viruses resuspended in milk, the medium mirroring the in vivo situation, proved more tenacious than those maintained in a cell culture medium. These viruses, subjected to harsh treatments such as drying and resuspending, partially maintained their replication capacity. After an immediate loss of nearly 1 log10 TCID50 immediately after desiccation, the viruses maintained their replication capacity for at least three weeks when desiccated in milk. These results suggest that fomites, clothing, or pastures contaminated with secretions or milk from infected animals might mediate the infection of animals independently of direct contact.
Collapse
Affiliation(s)
- Maksym Samoilenko
- Institute of Virology and Immunology IVI, 3147 Mittelhäusern, Switzerland;
- Institute of Virology and Immunology IVI, 3012 Bern, Switzerland
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, University of Bern, 3012 Bern, Switzerland
- Department of Epizootiology, Microbiology and Virology, Faculty of Veterinary Medicine, National University of Life and Environmental Sciences of Ukraine, 03041 Kyiv, Ukraine;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Vitalii Nedosekov
- Department of Epizootiology, Microbiology and Virology, Faculty of Veterinary Medicine, National University of Life and Environmental Sciences of Ukraine, 03041 Kyiv, Ukraine;
- Royal Veterinary College, University of London, London NW1 0TU, UK
| | - Giuseppe Bertoni
- Institute of Virology and Immunology IVI, 3147 Mittelhäusern, Switzerland;
- Institute of Virology and Immunology IVI, 3012 Bern, Switzerland
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
2
|
Schaer J, Cvetnic Z, Sukalic T, Dörig S, Grisiger M, Iscaro C, Feliziani F, Pfeifer F, Origgi F, Zanoni RG, Abril CE. Evaluation of Serological Methods and a New Real-Time Nested PCR for Small Ruminant Lentiviruses. Pathogens 2022; 11:pathogens11020129. [PMID: 35215072 PMCID: PMC8875174 DOI: 10.3390/pathogens11020129] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
Small ruminant lentiviruses (SRLVs), i.e., CAEV and MVV, cause insidious infections with life-long persistence and a slowly progressive disease, impairing both animal welfare and productivity in affected herds. The complex diagnosis of SRLVs currently combines serological methods including whole-virus and peptide-based ELISAs and Immunoblot. To improve the current diagnostic protocol, we analyzed 290 sera of animals originating from different European countries in parallel with three commercial screening ELISAs, Immunoblot as a confirmatory assay and five SU5 peptide ELISAs for genotype differentiation. A newly developed nested real-time PCR was carried out for the detection and genotype differentiation of the virus. Using a heat-map display of the combined results, the drawbacks of the current techniques were graphically visualized and quantified. The immunoblot and the SU5-ELISAs exhibited either unsatisfactory sensitivity or insufficient reliability in the differentiation of the causative viral genotype, respectively. The new truth standard was the concordance of the results of two out of three screening ELISAs and the PCR results for serologically false negative samples along with genotype differentiation. Whole-virus antigen-based ELISA showed the highest sensitivity (92.2%) and specificity (98.9%) among the screening tests, whereas PCR exhibited a sensitivity of 75%.
Collapse
Affiliation(s)
- Jessica Schaer
- Institute of Virology and Immunology IVI, in Cooperation with the Vetsuisse-Faculty of the University of Bern, 3012 Bern, Switzerland; (J.S.); (R.G.Z.)
| | - Zeljko Cvetnic
- Regional Veterinary Department Križevci, Croatian Veterinary Institute, Zakmandijeva 10, 48260 Križevci, Croatia; (Z.C.); (T.S.)
| | - Tomislav Sukalic
- Regional Veterinary Department Križevci, Croatian Veterinary Institute, Zakmandijeva 10, 48260 Križevci, Croatia; (Z.C.); (T.S.)
| | - Sven Dörig
- Beratungs-und Gesundheitsdienst für Kleinwiederkäuer (BGK/SSPR), 3362 Niederoenz, Switzerland;
| | | | - Carmen Iscaro
- National Reference Laboratory for Ruminant Retroviruses, Istituto Zooprofilattico, Sperimentale dell’Umbria e delle Marche Togo Rosati, 06126 Perugia, Italy; (C.I.); (F.F.)
| | - Francesco Feliziani
- National Reference Laboratory for Ruminant Retroviruses, Istituto Zooprofilattico, Sperimentale dell’Umbria e delle Marche Togo Rosati, 06126 Perugia, Italy; (C.I.); (F.F.)
| | - Folke Pfeifer
- Tierseuchenkasse/Tiergesundheitsdienst Sachsen-Anhalt, 39116 Magdeburg, Germany;
| | - Francesco Origgi
- Institute of Veterinary Pathology, Vetsuisse-Faculty of the University of Bern, 3012 Bern, Switzerland;
| | - Reto Giacomo Zanoni
- Institute of Virology and Immunology IVI, in Cooperation with the Vetsuisse-Faculty of the University of Bern, 3012 Bern, Switzerland; (J.S.); (R.G.Z.)
| | - Carlos Eduardo Abril
- Institute of Virology and Immunology IVI, in Cooperation with the Vetsuisse-Faculty of the University of Bern, 3012 Bern, Switzerland; (J.S.); (R.G.Z.)
- Correspondence: ; Tel.: +41-31-631-2423
| |
Collapse
|
3
|
Shi J, Li X, Zhu M, Chi H, Song Y, Wang J, Huang J. The dUTPase of caprine arthritis-encephalitis virus negatively regulates interferon signaling pathway. IRANIAN JOURNAL OF VETERINARY RESEARCH 2021; 22:209-216. [PMID: 34777521 DOI: 10.22099/ijvr.2021.38240.5568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 05/19/2021] [Accepted: 06/27/2021] [Indexed: 09/30/2022]
Abstract
Background Deoxyuracil triphosphate nucleotide (dUTP) pyrophosphatase (dUTPase, DU) is an enzyme of caprine arthritis-encephalitis virus (CAEV) that minimizes incorporation of dUTP into the DNA. Caprine arthritis-encephalitis virus relies partly on its ability to escape from innate immunity to cause persistent infections. Interferon β (IFN-β) is an important marker for evaluating the innate immune system, and it has a broad spectrum of antiviral activity. Aims This study was conducted to investigate the details of the IFN-β response to CAEV infection. Methods The expression of IFN-β and the proliferation of Sendai virus (SeV) and vesicular stomatitis virus (VSV) were determined by real-time quantitative polymerase chain reaction (qPCR). The effect of DU on the IFN signaling pathway was evaluated using luciferase reporter assays. Results In our study, the expression of IFN-β was significantly inhibited and the proliferation of SeV and VSV was promoted in cells overexpressing CAEV-DU. DU affected interferon stimulated response element (ISRE) and IFN-β promoter activities induced by RIG-I/MDA5/MAVS/TBK1 pathway, while did not affect them induced by interferon regulatory factor 3 (IRF3-5D). Conclusion DU protein downregulated the production of IFN-β by inhibiting the activity of the signal transduction molecules upstream of IRF3, thereby, helping CAEV escape innate immunity. Findings of this work provide an evidence to understand the persistent infection and multiple system inflammation of CAEV.
Collapse
Affiliation(s)
- J Shi
- MSc Student in Biology, Department of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China.,These authors contributed equally to this work
| | - X Li
- Departmet of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China.,These authors contributed equally to this work
| | - M Zhu
- MSc Student in Biology, Department of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - H Chi
- MSc Student in Biology, Department of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Y Song
- MSc Student in Biology, Department of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - J Wang
- MSc Student in Biology, Department of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - J Huang
- Departmet of Microbiology & Immunology, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
4
|
Munis AM. Gene Therapy Applications of Non-Human Lentiviral Vectors. Viruses 2020; 12:v12101106. [PMID: 33003635 PMCID: PMC7599719 DOI: 10.3390/v12101106] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Recent commercialization of lentiviral vector (LV)-based cell therapies and successful reports of clinical studies have demonstrated the untapped potential of LVs to treat diseases and benefit patients. LVs hold notable and inherent advantages over other gene transfer agents based on their ability to transduce non-dividing cells, permanently transform target cell genome, and allow stable, long-term transgene expression. LV systems based on non-human lentiviruses are attractive alternatives to conventional HIV-1-based LVs due to their lack of pathogenicity in humans. This article reviews non-human lentiviruses and highlights their unique characteristics regarding virology and molecular biology. The LV systems developed based on these lentiviruses, as well as their successes and shortcomings, are also discussed. As the field of gene therapy is advancing rapidly, the use of LVs uncovers further challenges and possibilities. Advances in virology and an improved understanding of lentiviral biology will aid in the creation of recombinant viral vector variants suitable for translational applications from a variety of lentiviruses.
Collapse
Affiliation(s)
- Altar M Munis
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
5
|
Kara H, Chazal N, Bouaziz S. Is Uracil-DNA Glycosylase UNG2 a New Cellular Weapon Against HIV-1? Curr HIV Res 2020; 17:148-160. [PMID: 31433761 DOI: 10.2174/1570162x17666190821154331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/01/2019] [Accepted: 08/09/2019] [Indexed: 01/12/2023]
Abstract
Uracil-DNA glycosylase-2 (UNG2) is a DNA repair protein that removes uracil from single and double-stranded DNA through a basic excision repair process. UNG2 is packaged into new virions by interaction with integrase (IN) and is needed during the early stages of the replication cycle. UNG2 appears to play both a positive and negative role during HIV-1 replication; UNG2 improves the fidelity of reverse transcription but the nuclear isoform of UNG2 participates in the degradation of cDNA and the persistence of the cellular genome by repairing its uracil mismatches. In addition, UNG2 is neutralized by Vpr, which redirects it to the proteasome for degradation, suggesting that UNG2 may be a new cellular restriction factor. So far, we have not understood why HIV-1 imports UNG2 via its IN and why it causes degradation of endogenous UNG2 by redirecting it to the proteasome via Vpr. In this review, we propose to discuss the ambiguous role of UNG2 during the HIV-1 replication cycle.
Collapse
Affiliation(s)
- Hesna Kara
- Cibles Therapeutiques et Conception de Medicaments (CiTCoM), CNRS UMR8038, Faculte des Sciences Pharmaceutiques et Biologiques, Universite Paris Descartes, Paris, France
| | - Nathalie Chazal
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR9004, Universite de Montpellier, Montpellier, France
| | - Serge Bouaziz
- Cibles Therapeutiques et Conception de Medicaments (CiTCoM), CNRS UMR8038, Faculte des Sciences Pharmaceutiques et Biologiques, Universite Paris Descartes, Paris, France
| |
Collapse
|
6
|
Fu Y, Lu D, Su Y, Chi H, Wang J, Huang J. The Vif protein of caprine arthritis encephalitis virus inhibits interferon production. Arch Virol 2020; 165:1557-1567. [PMID: 32356187 DOI: 10.1007/s00705-020-04637-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 04/01/2020] [Indexed: 11/24/2022]
Abstract
Caprine arthritis-encephalitis (CAE) is a chronic progressive infectious disease caused by caprine arthritis-encephalitis virus (CAEV) that seriously threatens the goat industry. Chronic infection and life-long multi-tissue inflammation are the typical features of the disease. Innate antiviral immunity is essential for the host defense system that rapidly recognizes and eliminates invading viruses. Interferon β (IFN-β) is important for innate immunity and regulates immunity against a broad spectrum of viruses. To investigate the details of the IFN-β response to CAEV infection, the effects of six viral proteins and the molecular mechanisms by which they affect IFN-β production were analyzed. Overexpression of DU and Vif promote virus proliferation and inhibit the production of IFN-β. qRT-PCR and luciferase reporter assays showed that overexpression of Vif inhibits the expression of luciferase under the control of the ISRE, NF-κB or IFN-β promoter but does not affect the expression of IFN-β activated by IRF3, indicating that Vif negatively regulates IFN-β production by affecting upstream signal transduction of IRF3. Amino acids 149-164 of Vif were found to be necessary for the inhibitory effect of IFN-β production. Our results indicate that CAEV evades surveillance and clearance by intracellular innate immunity by downregulating IFN-β production.
Collapse
Affiliation(s)
- Yali Fu
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China
| | - Dong Lu
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China
| | - Yanxin Su
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China
| | - Heng Chi
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China
| | - Jiashun Wang
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, No. 92, Weijin road, Nankai District, Tianjin, 300072, China.
| |
Collapse
|
7
|
Molecular characterization of circulating strains of small ruminant lentiviruses in Brazil based on complete gag and pol genes. Small Rumin Res 2019. [DOI: 10.1016/j.smallrumres.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Grego E, Reina R, Lanfredini S, Tursi M, Favole A, Profiti M, Lungu MM, Perona G, Gay L, Stella MC, DeMeneghi D. Viral load, tissue distribution and histopathological lesions in goats naturally and experimentally infected with the Small Ruminant Lentivirus Genotype E (subtype E1 Roccaverano strain). Res Vet Sci 2018; 118:107-114. [PMID: 29421479 DOI: 10.1016/j.rvsc.2018.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 01/10/2018] [Accepted: 01/13/2018] [Indexed: 11/27/2022]
Abstract
Small Ruminant Lentivirus (SRLV) subtype E1, also known as Roccaverano strain, is considered a low pathogenic virus on the basis of natural genetic deletions, in vitro properties and on-farm observations. In order to gain more knowledge on this atypical lentivirus we investigated the in vivo tropism of Roccaverano strain in both, experimentally and naturally infected goats. Antibody responses were monitored as well as tissue distribution and viral load, evaluated by real time PCR on single spliced (gag/env) and multiple spliced (rev) RNA targets respectively, that were compared to histopathological lesions. Lymph nodes, spleen, alveolar macrophages and mammary gland turned out to be the main tissue reservoirs of genotype E1-provirus. Moreover, mammary gland and/or mammary lymph nodes acted as active replication sites in dairy goats, supporting the lactogenic transmission of this virus. Notably, a direct association between viral load and concomitant infection or inflammatory processes was evident within organs such as spleen, lung and testis. Our results validate the low pathogenicity designation of SRLV genotype E1 in vivo, and confirm the monocyte-macrophage cell lineage as the main virus reservoir of this genotype. Accordingly, SRLV genotype E displays a tropism towards all tissues characterized by an abundant presence of these cells, either for their own anatomical structure or for an occasional infectious/inflammatory status.
Collapse
Affiliation(s)
- E Grego
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy.
| | - R Reina
- Instituto de Agrobiotecnología, CSIC-UPNA, Gobierno de Navarra, Mutilva, Navarra 31192, Spain
| | - S Lanfredini
- European Cancer Stem Cell, Research Institute Hadyn Ellis, Building Maindy Road Cathays, CF24 4HQ Cardiff, UK
| | - M Tursi
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| | - A Favole
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Torino, Via Bologna 148, 10154 Torino, Italy
| | - M Profiti
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| | - M M Lungu
- Universitatea "Stefan cel Mare" dinSuceava, Departamentul de sanatate si dezvoltare umana, Str. Universitatii, 13, Suceava 720229, Romania
| | - G Perona
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| | - L Gay
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| | - M C Stella
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| | - D DeMeneghi
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy
| |
Collapse
|
9
|
Podolny Y, Herzig E, Hizi A. Insights into the molecular and biological features of the dUTPase-related gene of bovine immunodeficiency virus. Virology 2017; 506:55-63. [PMID: 28342388 DOI: 10.1016/j.virol.2017.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/06/2017] [Accepted: 03/16/2017] [Indexed: 01/20/2023]
Abstract
This study was stimulated by our previous research of the dUTPase-related protein from bovine immunodeficiency virus (BIV) (Voronin et al., 2014). Despite the lack of detectable enzymatic BIV dUTPase activity (both of the recombinant protein and in virions), mutating the dUTPase gene was deleterious to viral production. However, cDNA synthesis and integration were apparently unaffected. Consequently, we have studied here two important issues. First, we showed that in cDNA produced by the dUTPase-mutated virions, the incidence of mutations was not higher than that found in wild-type BIV-infected cells. Second, single mutations, introduced in preserved dUTPase residues Asp48 and Asn57 (in the putative dUTPase active site or close to it), have led to abortive BIV infections (except for the conservative Asp48Glu mutation). Therefore, we postulate that the BIV dUTPase-related protein has a critical role in retroviral replication at steps that take place after viral cDNA synthesis and integration.
Collapse
Affiliation(s)
- Yulia Podolny
- From the Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Eytan Herzig
- From the Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Amnon Hizi
- From the Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
10
|
Blatti-Cardinaux L, Sanjosé L, Zahno ML, Zanoni R, Reina R, Bertoni G. Detailed analysis of the promoter activity of an attenuated lentivirus. J Gen Virol 2016; 97:1699-1708. [PMID: 27114068 DOI: 10.1099/jgv.0.000489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In spite of an eradication campaign that eliminated clinical cases of caprine arthritis encephalitis virus-induced arthritis in the Swiss goat population, seroconversions are still observed. In the affected flocks, viruses belonging mainly to the small ruminant lentivirus A4 subtype are regularly isolated. These viruses are considered attenuated, except in the mammary gland, where high viral loads and histopathological lesions have been observed. We previously characterized and sequenced such field isolates, detecting several potentially attenuating mutations in their LTR. Here we present a detailed analysis of the promoter activity of these genetic elements, which was comparable to those of virulent isolates. An AP-1 binding site was shown to be crucial for promoter activity in reporter gene assays and also in the context of a replicating molecular clone. Other sites, such as AML(vis) and a conserved E-box, appeared to be less crucial. Analysis of a unique AP-4 site showed a clear discrepancy between results obtained with reporter gene assays and those with mutated viruses. Within the limits of this in vitro study, we did not find evidence pointing to the LTR as the genetic correlate of attenuation for these viruses. Finally, the limited replication of SRLV A4 in mammary cell culture could not explain the suggested mammary tropism. In contrast, and in view of the abundance of macrophages in the mammary gland, it is the striking replication capacity of SRLV A4 in these cells, unaffected by all LTR mutations tested, which may explain the apparent mammary tropism of these viruses.
Collapse
Affiliation(s)
- Laure Blatti-Cardinaux
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Leticia Sanjosé
- Instituto de Agrobiotecnología (CSIC-Universidad Pública de Navarra-Gobierno de Navarra) Avda, Pamplona, Spain
| | - Marie-Luise Zahno
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Zanoni
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ramses Reina
- Instituto de Agrobiotecnología (CSIC-Universidad Pública de Navarra-Gobierno de Navarra) Avda, Pamplona, Spain
| | - Giuseppe Bertoni
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Abstract
Despite having very limited coding capacity, RNA viruses are able to withstand challenge of antiviral drugs, cause epidemics in previously exposed human populations, and, in some cases, infect multiple host species. They are able to achieve this by virtue of their ability to multiply very rapidly, coupled with their extraordinary degree of genetic heterogeneity. RNA viruses exist not as single genotypes, but as a swarm of related variants, and this genomic diversity is an essential feature of their biology. RNA viruses have a variety of mechanisms that act in combination to determine their genetic heterogeneity. These include polymerase fidelity, error-mitigation mechanisms, genomic recombination, and different modes of genome replication. RNA viruses can vary in their ability to tolerate mutations, or “genetic robustness,” and several factors contribute to this. Finally, there is evidence that some RNA viruses exist close to a threshold where polymerase error rate has evolved to maximize the possible sequence space available, while avoiding the accumulation of a lethal load of deleterious mutations. We speculate that different viruses have evolved different error rates to complement the different “life-styles” they possess.
Collapse
Affiliation(s)
- J.N. Barr
- University of Leeds, Leeds, United Kingdom
| | - R. Fearns
- Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Blatti-Cardinaux L, Pisoni G, Stoffel MH, Zanoni R, Zahno ML, Bertoni G. Generation of a molecular clone of an attenuated lentivirus, a first step in understanding cytopathogenicity and virulence. Virology 2015; 487:50-8. [PMID: 26517396 DOI: 10.1016/j.virol.2015.09.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 09/23/2015] [Accepted: 09/30/2015] [Indexed: 11/20/2022]
Abstract
Small ruminant lentiviruses infect goats and sheep, inducing clinical disease in a minority of infected animals. Following an eradication campaign, clinical cases may disappear in a population. The complete elimination of these lentiviruses is however difficult to achieve and the spreading of less virulent strains often parallels the elimination of their virulent counterparts. Here, we characterized three such strains isolated from a flock in the post-eradication phase. We completely sequenced their genomes, showing that one of the isolates was most probably the product of a recombination event between the other two viruses. By comparing the sequences of these isolates with those of virulent strains, we found evidence that particular LTR mutations may explain their attenuated phenotype. Finally, we constructed an infectious molecular clone representative of these viruses, analyzing its replication characteristics in different target cells. This clone will permit us to explore the molecular correlates of cytopathogenicity and virulence.
Collapse
Affiliation(s)
- Laure Blatti-Cardinaux
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Giuliano Pisoni
- Università degli Studi di Milano, Department of Health, Animal Science and Food Safety, via Celoria 10, 20133 Milano, Italy
| | - Michael H Stoffel
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Zanoni
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marie-Luise Zahno
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Giuseppe Bertoni
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
13
|
Hizi A, Herzig E. dUTPase: the frequently overlooked enzyme encoded by many retroviruses. Retrovirology 2015; 12:70. [PMID: 26259899 PMCID: PMC4531489 DOI: 10.1186/s12977-015-0198-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/04/2015] [Indexed: 11/13/2022] Open
Abstract
Retroviruses are among the best studied viruses in last decades due to their pivotal involvement in cellular processes and, most importantly, in causing human diseases, most notably-acquired immunodeficiency syndrome (AIDS) that is triggered by human immunodeficiency viruses types 1 and 2 (HIV-1 and HIV-2, respectively). Numerous studied were conducted to understand the involvement of the three cardinal retroviral enzymes, reverse transcriptase, integrase and protease, in the life cycle of the viruses. These studies have led to the development of many inhibitors of these enzymes as anti-retroviral specific drugs that are used for routine treatments of HIV/AIDS patients. Interestingly, a fourth virus-encoded enzyme, the deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) is also found in several major retroviral groups. The presence and the importance of this enzyme to the life cycle of retroviruses were usually overlooked by most retrovirologists, although the occurrence of dUTPases, particularly in beta-retroviruses and in non-primate retroviruses, is known for more than 20 years. Only more recently, retroviral dUTPases were brought into the limelight and were shown in several cases to be essential for viral replication. Therefore, it is likely that future studies on this enzyme will advance our knowledge to a level that will allow designing novel, specific and potent anti-dUTPase drugs that are effective in combating retroviral diseases. The aim of this review is to give concise background information on dUTPases in general and to summarize the most relevant data on retroviral dUTPases and their involvement in the replication processes and pathogenicity of the viruses, as well as in possibly-associated human diseases.
Collapse
Affiliation(s)
- Amnon Hizi
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Eytan Herzig
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
14
|
Molecular characterization of the gag gene of caprine arthritis encephalitis virus from goats in the Philippines. Arch Virol 2015; 160:969-78. [PMID: 25655265 DOI: 10.1007/s00705-015-2359-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/30/2015] [Indexed: 10/24/2022]
Abstract
Caprine arthritis encephalitis virus (CAEV) causes caprine arthritis encephalitis syndrome, which is an emerging disease of goats in the Philippines. DNA sequence analysis showed homology of 86-93 % between Philippine CAEV and available CAEV sequences in GenBank. CAEV was detected using nested polymerase chain reaction (PCR), and new sets of primers were designed in order to amplify the gag gene, which is a highly conserved region of the viral genome. In addition, the Philippine CAEV isolate clustered in group B with the prototype caprine lentivirus. Based on amino acid sequence alignments, it is possible that the Philippine CAEV isolate is a new strain of CAEV, but it is also possible that it was already present in the country even before the start of goat importation. Molecular characterization of the CAEV gag gene is important for the development of a detection kit specific for the local strain of CAEV and the establishment of small ruminant lentivirus eradication programs in the Philippines. This study is the first report to describe the molecular characteristics of CAEV circulating in the Philippines.
Collapse
|
15
|
Voronin N, Herzig E, Hizi A. The dUTPase-related gene of bovine immunodeficiency virus is critical for viral replication, despite the lack of dUTPase activity of the encoded protein. Retrovirology 2014; 11:60. [PMID: 25117862 PMCID: PMC4261571 DOI: 10.1186/1742-4690-11-60] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/09/2014] [Indexed: 01/03/2023] Open
Abstract
Background Deoxyuridine 5′-triphosphate nucleotide-hydrolases (dUTPases) are essential for maintaining low intra-cellular dUTP/dTTP ratios. Therefore, many viruses encode this enzyme to prevent dUTP incorporation into their genomes instead of dTTP. Among the lentiviruses, the non-primate viruses express dUTPases. In bovine immunodeficiency virus (BIV), the putative dUTPase protein is only 74 residues-long, compared to ~130 residues in other lentiviruses. Results In this study, the recombinant BIV dUTPase, as well as infectious wild-type (WT) BIV virions, were shown to lack any detectable dUTPase activity. Controls of recombinant dUTPase from equine infectious anemia virus (EIAV) or of EIAV virions showed substantial dUTPase activities. To assess the importance of the dUTPase to BIV replication, we have generated virions of WT BIV or BIV with mutations in the dUTPase gene. The two mutant viral dUTPases were the double mutant D48E/N57S (in the putative enzyme active site and its vicinity) and a deletion of 36 residues. In dividing Cf2Th cells and under conditions where the WT virus was infectious and generated progeny virions, both mutant viruses were defective, as no progeny viruses were generated. Analyses of the integrated viral cDNA showed that cells infected with the mutant virions carry in their genomic DNA levels of integrated BIV DNA that are comparable to those in WT BIV-infected cells. Conclusions The herby presented results show that the two BIV mutants with the modified dUTPase gene could infect cells, as viral cDNA was synthesized and integrated into the host cell DNA. However, no virions were generated by cells infected by these mutants. The most likely explanation is that either the integrated cDNA of the mutants is defective (due to potential multiple mutations, introduced during reverse-transcription) or that the original dUTPase mutations have led to severe blocks in viral replication at steps post integration. These results emphasize the importance of the dUTPase-related sequence to BIV replication, despite the lack of any detectable catalytic activity. Electronic supplementary material The online version of this article (doi:10.1186/1742-4690-11-60) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Amnon Hizi
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
16
|
Deubelbeiss M, Blatti-Cardinaux L, Zahno ML, Zanoni R, Vogt HR, Posthaus H, Bertoni G. Characterization of small ruminant lentivirus A4 subtype isolates and assessment of their pathogenic potential in naturally infected goats. Virol J 2014; 11:65. [PMID: 24708706 PMCID: PMC3995504 DOI: 10.1186/1743-422x-11-65] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/13/2014] [Indexed: 11/10/2022] Open
Abstract
Background Small ruminant lentiviruses escaping efficient serological detection are still circulating in Swiss goats in spite of a long eradication campaign that essentially eliminated clinical cases of caprine arthritis encephalitis in the country. This strongly suggests that the circulating viruses are avirulent for goats. To test this hypothesis, we isolated circulating viruses from naturally infected animals and tested the in vitro and in vivo characteristics of these field isolates. Methods Viruses were isolated from primary macrophage cultures. The presence of lentiviruses in the culture supernatants was monitored by reverse transcriptase assay. Isolates were passaged in different cells and their cytopathogenic effects monitored by microscopy. Proviral load was quantified by real-time PCR using customized primer and probes. Statistical analysis comprised Analysis of Variance and Bonferroni Multiple Comparison Test. Results The isolated viruses belonged to the small ruminant lentiviruses A4 subtype that appears to be prominent in Switzerland. The 4 isolates replicated very efficiently in macrophages, displaying heterogeneous phenotypes, with two isolates showing a pronounced cytopathogenicity for these cells. By contrast, all 4 isolates had a poor replication capacity in goat and sheep fibroblasts. The proviral loads in the peripheral blood and, in particular, in the mammary gland were surprisingly high compared to previous observations. Nevertheless, these viruses appear to be of low virulence for goats except for the mammary gland were histopathological changes were observed. Conclusions Small ruminant lentiviruses continue to circulate in Switzerland despite a long and expensive caprine arthritis encephalitis virus eradication campaign. We isolated 4 of these lentiviruses and confirmed their phylogenetic association with the prominent A4 subtype. The pathological and histopathological analysis of the infected animals supported the hypothesis that these A4 viruses are of low pathogenicity for goats, with, however, a caveat about the potentially detrimental effects on the mammary gland. Moreover, the high proviral load detected indicates that the immune system of the animals cannot control the infection and this, combined with the phenotypic plasticity observed in vitro, strongly argues in favour of a continuous and precise monitoring of these SRLV to avoid the risk of jeopardizing a long eradication campaign.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Giuseppe Bertoni
- Institute of Virology and Immunology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Bhatia S, Patil SS, Sood R. Bovine immunodeficiency virus: a lentiviral infection. INDIAN JOURNAL OF VIROLOGY : AN OFFICIAL ORGAN OF INDIAN VIROLOGICAL SOCIETY 2013; 24:332-41. [PMID: 24426295 DOI: 10.1007/s13337-013-0165-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 09/16/2013] [Indexed: 12/11/2022]
Abstract
The bovine immunodeficiency virus (BIV) is a lentivirus which is known to infect cattle worldwide. Though serological and genomic evidence of BIV in cattle has been found throughout the world, isolation of the virus has been reported only from few places. Very little is known about its impact on animal health status, pathogenesis and mode of transmission. BIV is considered generally non-pathogenic and is not known to cause any serious disease in cattle. BIV is genetically and antigenically related to Jembrana disease virus (JDV), the cause of an acute disease in Bali cattle (Bos javanicus) and human immunodeficiency virus, the cause of acquired immunodeficiency syndrome in human. Therefore, it is important to monitor the presence of BIV in cattle to keep vigil over its possible evolution in its natural host to emerge as pathogenic lentivirus like JDV. Differentiation of BIV infection in cattle from the acutely pathogenic JDV is important for diagnosis of the latter. Currently, BIV is considered as a safe model for understanding the complex genome of lentiviruses. Further research on BIV is indeed needed to elucidate its possible role in animal health as well as for insight into the molecular mechanisms adopted by related lentiviruses.
Collapse
Affiliation(s)
- Sandeep Bhatia
- High Security Animal Disease Laboratory (HSADL), Indian Veterinary Research Institute (IVRI), Bhopal, India
| | - S S Patil
- Project Directorate on Animal Disease Monitoring and Surveillance (PD-ADMAS), Bengaluru, India
| | - R Sood
- High Security Animal Disease Laboratory (HSADL), Indian Veterinary Research Institute (IVRI), Bhopal, India
| |
Collapse
|
18
|
Ramírez H, Reina R, Amorena B, de Andrés D, Martínez HA. Small ruminant lentiviruses: genetic variability, tropism and diagnosis. Viruses 2013; 5:1175-1207. [PMID: 23611847 PMCID: PMC3705272 DOI: 10.3390/v5041175] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/09/2013] [Accepted: 04/12/2013] [Indexed: 02/05/2023] Open
Abstract
Small ruminant lentiviruses (SRLV) cause a multisystemic chronic disease affecting animal production and welfare. SRLV infections are spread across the world with the exception of Iceland. Success in controlling SRLV spread depends largely on the use of appropriate diagnostic tools, but the existence of a high genetic/antigenic variability among these viruses, the fluctuant levels of antibody against them and the low viral loads found in infected individuals hamper the diagnostic efficacy. SRLV have a marked in vivo tropism towards the monocyte/macrophage lineage and attempts have been made to identify the genome regions involved in tropism, with two main candidates, the LTR and env gene, since LTR contains primer binding sites for viral replication and the env-encoded protein (SU ENV), which mediates the binding of the virus to the host's cell and has hypervariable regions to escape the humoral immune response. Once inside the host cell, innate immunity may interfere with SRLV replication, but the virus develops counteraction mechanisms to escape, multiply and survive, creating a quasi-species and undergoing compartmentalization events. So far, the mechanisms of organ tropism involved in the development of different disease forms (neurological, arthritic, pulmonary and mammary) are unknown, but different alternatives are proposed. This is an overview of the current state of knowledge on SRLV genetic variability and its implications in tropism as well as in the development of alternative diagnostic assays.
Collapse
Affiliation(s)
- Hugo Ramírez
- Laboratory of Virology, Genetics and Molecular Biology, FES-Cuautitlán, UNAM C-4 Veterinary, Cuautitlán Izcalli, State of Mexico 54714, Mexico; E-Mail:
| | - Ramsés Reina
- Institute of Agrobiotechnology, CSIC-UPNA-Government of Navarra, Ctra. Mutilva Baja s/n, Navarra 31192, Spain; E-Mails: (R.R.); (B.A.); (D.A.)
| | - Beatriz Amorena
- Institute of Agrobiotechnology, CSIC-UPNA-Government of Navarra, Ctra. Mutilva Baja s/n, Navarra 31192, Spain; E-Mails: (R.R.); (B.A.); (D.A.)
| | - Damián de Andrés
- Institute of Agrobiotechnology, CSIC-UPNA-Government of Navarra, Ctra. Mutilva Baja s/n, Navarra 31192, Spain; E-Mails: (R.R.); (B.A.); (D.A.)
| | - Humberto A. Martínez
- Laboratory of Virology, Genetics and Molecular Biology, FES-Cuautitlán, UNAM C-4 Veterinary, Cuautitlán Izcalli, State of Mexico 54714, Mexico; E-Mail:
| |
Collapse
|
19
|
Uracil DNA glycosylase initiates degradation of HIV-1 cDNA containing misincorporated dUTP and prevents viral integration. Proc Natl Acad Sci U S A 2013; 110:E448-57. [PMID: 23341616 DOI: 10.1073/pnas.1219702110] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
HIV-1 reverse transcriptase discriminates poorly between dUTP and dTTP, and accordingly, viral DNA products become heavily uracilated when viruses infect host cells that contain high ratios of dUTP:dTTP. Uracilation of invading retroviral DNA is thought to be an innate immunity barrier to retroviral infection, but the mechanistic features of this immune pathway and the cellular fate of uracilated retroviral DNA products is not known. Here we developed a model system in which the cellular dUTP:dTTP ratio can be pharmacologically increased to favor dUTP incorporation, allowing dissection of this innate immunity pathway. When the virus-infected cells contained elevated dUTP levels, reverse transcription was found to proceed unperturbed, but integration and viral protein expression were largely blocked. Furthermore, successfully integrated proviruses lacked detectable uracil, suggesting that only nonuracilated viral DNA products were integration competent. Integration of the uracilated proviruses was restored using an isogenic cell line that had no detectable human uracil DNA glycosylase (hUNG2) activity, establishing that hUNG2 is a host restriction factor in cells that contain high dUTP. Biochemical studies in primary cells established that this immune pathway is not operative in CD4+ T cells, because these cells have high dUTPase activity (low dUTP), and only modest levels of hUNG activity. Although monocyte-derived macrophages have high dUTP levels, these cells have low hUNG activity, which may diminish the effectiveness of this restriction pathway. These findings establish the essential elements of this pathway and reconcile diverse observations in the literature.
Collapse
|
20
|
van der Kuyl AC, Berkhout B. The biased nucleotide composition of the HIV genome: a constant factor in a highly variable virus. Retrovirology 2012; 9:92. [PMID: 23131071 PMCID: PMC3511177 DOI: 10.1186/1742-4690-9-92] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/14/2012] [Indexed: 01/09/2023] Open
Abstract
Viruses often deviate from their hosts in the nucleotide composition of their genomes. The RNA genome of the lentivirus family of retroviruses, including human immunodeficiency virus (HIV), contains e.g. an above average percentage of adenine (A) nucleotides, while being extremely poor in cytosine (C). Such a deviant base composition has implications for the amino acids that are encoded by the open reading frames (ORFs), both in the requirement of specific tRNA species and in the preference for amino acids encoded by e.g. A-rich codons. Nucleotide composition does obviously affect the secondary and tertiary structure of the RNA genome and its biological functions, but it does also influence phylogenetic analysis of viral genome sequences, and possibly the activity of the integrated DNA provirus. Over time, the nucleotide composition of the HIV-1 genome is exceptionally conserved, varying by less than 1% per base position per isolate within either group M, N, or O during 1983–2009. This extreme stability of the nucleotide composition may possibly be achieved by negative selection, perhaps conserving semi-stable RNA secondary structure as reverse transcription would be significantly affected for a less A-rich genome where secondary structures are expected to be more stable and thus more difficult to unfold. This review will discuss all aspects of the lentiviral genome composition, both of the RNA and of its derived double-stranded DNA genome, with a focus on HIV-1, the nucleotide composition over time, the effects of artificially humanized codons as well as contributions of immune system pressure on HIV nucleotide bias.
Collapse
Affiliation(s)
- Antoinette C van der Kuyl
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam, Academic Medical Center of the University of Amsterdam, Meibergdreef 15, Amsterdam, AZ 1105, The Netherlands.
| | | |
Collapse
|
21
|
Blacklaws BA. Small ruminant lentiviruses: immunopathogenesis of visna-maedi and caprine arthritis and encephalitis virus. Comp Immunol Microbiol Infect Dis 2012; 35:259-69. [PMID: 22237012 DOI: 10.1016/j.cimid.2011.12.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 10/14/2022]
Abstract
The small ruminant lentiviruses include the prototype for the genus, visna-maedi virus (VMV) as well as caprine arthritis encephalitis virus (CAEV). Infection of sheep or goats with these viruses causes slow, progressive, inflammatory pathology in many tissues, but the most common clinical signs result from pathology in the lung, mammary gland, central nervous system and joints. This review examines replication, immunity to and pathogenesis of these viruses and highlights major differences from and similarities to some of the other lentiviruses.
Collapse
Affiliation(s)
- Barbara A Blacklaws
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK.
| |
Collapse
|
22
|
Jónsson SR, Andrésdóttir V. Propagating and detecting an infectious molecular clone of maedi-visna virus that expresses green fluorescent protein. J Vis Exp 2011:3483. [PMID: 22005550 PMCID: PMC3227179 DOI: 10.3791/3483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Maedi-visna virus (MVV) is a lentivirus of sheep, causing slowly progressive interstitial pneumonia and encephalitis. The primary target cells of MVV in vivo are considered to be of the monocyte lineage. Certain strains of MVV can replicate in other cell types, however. The green fluorescent protein is a commonly used marker for studying lentiviruses in living cells. We have nserted the egfp gene into the gene for dUTPase of MVV. The dUTPase gene is well conserved in most lentivirus strains of sheep and goats and has been shown to be important in replication of CAEV. However, dUTPase has been shown to be dispensable for replication of the molecular clone of MVV used in this study both in vitro and in vivo. MVV replication is strictly confined to cells of sheep or goat origin. We use a primary cell line from the choroid plexus of sheep (SCP cells) for transfection and propagation of the virus. The fluorescent MVV is fully infectious and EGFP expression is stable over at least 6 passages. There is good correlation between measurements of TCID₅₀ and EGFP. This virus should therefore be useful for rapid detection of infected cells in studies of cell tropism and pathogenicity in vitro and in vivo.
Collapse
|
23
|
Juganaru M, Reina R, Bertolotti L, Stella M, Profiti M, Armentano M, Bollo E, Amorena B, Rosati S. In vitro properties of small ruminant lentivirus genotype E. Virology 2011; 410:88-95. [DOI: 10.1016/j.virol.2010.10.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 07/15/2010] [Accepted: 10/17/2010] [Indexed: 10/18/2022]
|
24
|
Reina R, Juganaru MM, Profiti M, Cascio P, Cerruti F, Bertolotti L, De Meneghi D, Amorena B, Rosati S. Immunological parameters in goats experimentally infected with SRLV genotype E, strain Roccaverano. Vet Immunol Immunopathol 2010; 139:237-44. [PMID: 21122927 DOI: 10.1016/j.vetimm.2010.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/02/2010] [Accepted: 11/04/2010] [Indexed: 10/18/2022]
Abstract
Genotype E of small ruminant lentivirus has been recently described in goats from different breeds in Italy. Genotype E infection may differ from known genotypes since deletions of dUTPase and VPR proteins have been confirmed in different independent areas and goat breed, and play a key role on virus replication and pathogenesis. In particular, genotype E Roccaverano strain has been described as low pathogenic since does not lead to clinical symptoms in goats. In contrast, classical CAEV infected goats of the same area and breed presented arthritis. In this study, we have used intratracheal and intra-bone marrow routes to establish genotype E persistent infections. Humoral and cellular immune responses elicited in the host against genotype E and genotype B derived antigens were evaluated until 200 days post-inoculation. Compared to genotype B antigen, seroconversion against genotype E GAG P16-25 antigen was detected at 2-3 weeks after inoculation, significantly earlier and at higher titres. Interestingly, antibody avidity did not increase in the course of the experiment neither against P16-25 nor against SU5, both derived from genotype E. T cell proliferation against P25-GST fusion protein antigens derived from genotype E was firstly detected at 15 days post-inoculation and was maintained throughout time until week 20 post-infection, while T cell proliferation against the genotype B P25 was not produced by the end of the experiment at 20 weeks post-inoculation. The strength of reaction was also higher when using P25 E as stimulator antigen. In contrast with antibody and T cell proliferation, cytotoxic-T-lymphocyte (CTL) activity in the circulating lymphocytes (effector cells) using blood-derived macrophages (BDM) as target cells, was not strain specific being surprisingly higher against genotype B infected antigen presenting cells (APCs). This is the first study reporting experimentally induced immunological changes in SRLV genotype E infection and indicates that CTL activity may be the adaptive immune response able to induce protection against heterologous infection.
Collapse
Affiliation(s)
- Ramses Reina
- Instituto de Agrobiotecnología, CSIC-Universidad Pública de Navarra-Gobierno de Navarra, Mutilva Baja, Navarra, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fritz JV, Briant L, Mély Y, Bouaziz S, de Rocquigny H. HIV-1 viral protein r: from structure to function. Future Virol 2010. [DOI: 10.2217/fvl.10.47] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The viral protein r (Vpr) of HIV-1 binds several host proteins leading to pleiotropic functions, such as G2/M cell cycle arrest, apoptosis induction and gene transactivation. Vpr is encapsidated through the Gag C-terminus into the nascent viral particles, suggesting that Vpr plays several important functions in the early stages of the viral lifecycle. In this regard, Vpr interacts with nucleic acids and membranes to facilitate the preintegration complex migration and incorporation into the nucleus of nondividing cells. Thus, Vpr has to recruit several host and viral factors to promote its functions during HIV-1 pathogenesis. This article focuses on its interacting partners by giving an overview of the functional outcome of the different Vpr complexes, as well as the structural determinants of Vpr required for its binding properties.
Collapse
Affiliation(s)
- Joëlle V Fritz
- Department of Infectious Diseases, Virology, Universitätsklinikum, Im Neuenheimer Feld, 324, D-69120, Heidelberg, Germany
| | - Laurence Briant
- Université Montpellier 1, Centre d’études d’agents Pathogènes et Biotechnologies pour la Santé, CNRS, UMR 5236, CPBS, F-34965 Montpellier, France
| | - Yves Mély
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Université de Strasbourg, Faculté de Pharmacie, 74, Route du Rhin, 67401 ILLKIRCH Cedex, France
| | - Serge Bouaziz
- Laboratoire de Cristallographie et RMN Biologiques, CNRS UMR8015 UFR des Sciences Pharmaceutiques et Biologiques 4, Avenue de L’observatoire, 75006 Paris, France: Université de Strasbourg, Faculté de Pharmacie, 74, Route du Rhin, 67401 ILLKIRCH Cedex, France
| | | |
Collapse
|
26
|
Nuclear-export-signal-dependent protein translocation of dUTPase encoded by Singapore grouper iridovirus. Arch Virol 2010; 155:1069-76. [DOI: 10.1007/s00705-010-0684-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 04/26/2010] [Indexed: 11/25/2022]
|
27
|
Jones KL, Roche M, Gantier MP, Begum NA, Honjo T, Caradonna S, Williams BRG, Mak J. X4 and R5 HIV-1 have distinct post-entry requirements for uracil DNA glycosylase during infection of primary cells. J Biol Chem 2010; 285:18603-14. [PMID: 20371602 DOI: 10.1074/jbc.m109.090126] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been assumed that R5 and X4 HIV utilize similar strategies to support viral cDNA synthesis post viral entry. In this study, we provide evidence to show that R5 and X4 HIV have distinct requirements for host cell uracil DNA glycosylase (UNG2) during the early stage of infection. UNG2 has been previously implicated in HIV infection, but its precise role remains controversial. In this study we show that, although UNG2 is highly expressed in different cell lines, UNG2 levels are low in the natural host cells of HIV. Short interfering RNA knockdown of endogenous UNG2 in primary cells showed that UNG2 is required for R5 but not X4 HIV infection and that this requirement is bypassed when HIV enters the target cell via vesicular stomatitis virus envelope-glycoprotein-mediated endocytosis. We also show that short interfering RNA knockdown of UNG2 in virus-producing primary cells leads to defective R5 HIV virions that are unable to complete viral cDNA synthesis. Quantitative PCR analysis revealed that endogenous UNG2 levels are transiently up-regulated post HIV infection, and this increase in UNG2 mRNA is approximately 10-20 times higher in R5 versus X4 HIV-infected cells. Our data show that both virion-associated UNG2 and HIV infection-induced UNG2 expression are critical for reverse transcription during R5 but not X4 HIV infection. More importantly, we have made the novel observation that R5 and X4 HIV have distinct host cell factor requirements and differential capacities to induce gene expression during the early stages of infection. These differences may result from activation of distinct signaling cascades and/or infection of divergent T-lymphocyte subpopulations.
Collapse
Affiliation(s)
- Kate L Jones
- Centre for Virology, Burnet Institute, Melbourne, Victoria 3004, Australia
| | | | | | | | | | | | | | | |
Collapse
|
28
|
De Silva FS, Moss B. Effects of vaccinia virus uracil DNA glycosylase catalytic site and deoxyuridine triphosphatase deletion mutations individually and together on replication in active and quiescent cells and pathogenesis in mice. Virol J 2008; 5:145. [PMID: 19055736 PMCID: PMC2630940 DOI: 10.1186/1743-422x-5-145] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 12/02/2008] [Indexed: 01/24/2023] Open
Abstract
Background Low levels of uracil in DNA result from misincorporation of dUMP or cytosine deamination. Vaccinia virus (VACV), the prototype poxvirus, encodes two enzymes that can potentially reduce the amount of uracil in DNA. Deoxyuridine triphosphatase (dUTPase) hydrolyzes dUTP, generating dUMP for biosynthesis of thymidine nucleotides while decreasing the availability of dUTP for misincorporation; uracil DNA glycosylase (UNG) cleaves uracil N-glycosylic bonds in DNA initiating base excision repair. Studies with actively dividing cells showed that the VACV UNG protein is required for DNA replication but the UNG catalytic site is not, whereas the dUTPase gene can be deleted without impairing virus replication. Recombinant VACV with an UNG catalytic site mutation was attenuated in vivo, while a dUTPase deletion mutant was not. However, the importance of the two enzymes for replication in quiescent cells, their possible synergy and roles in virulence have not been fully assessed. Results VACV mutants lacking the gene encoding dUTPase or with catalytic site mutations in UNG and double UNG/dUTPase mutants were constructed. Replication of UNG and UNG/dUTPase mutants were slightly reduced compared to wild type or the dUTPase mutant in actively dividing cells. Viral DNA replication was reduced about one-third under these conditions. After high multiplicity infection of quiescent fibroblasts, yields of wild type and mutant viruses were decreased by 2-logs with relative differences similar to those observed in active fibroblasts. However, under low multiplicity multi-step growth conditions in quiescent fibroblasts, replication of the dUTPase/UNG mutant was delayed and 5-fold lower than that of either single mutant or parental virus. This difference was exacerbated by 1-day serial passages on quiescent fibroblasts, resulting in 2- to 3-logs lower titer of the double mutant compared to the parental and single mutant viruses. Each mutant was more attenuated than a revertant virus upon intranasal infection of mice. Conclusion VACV UNG and dUTPase activities are more important for replication in quiescent cells, which have low levels of endogenous UNG and dUTPase, than in more metabolically active cells and the loss of both is more detrimental than either alone. Both UNG and dUTPase activities are required for full virulence in mice.
Collapse
Affiliation(s)
- Frank S De Silva
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-3210, USA
| | | |
Collapse
|
29
|
Genome analysis of small-ruminant lentivirus genotype E: a caprine lentivirus with natural deletions of the dUTPase subunit, vpr-like accessory gene, and 70-base-pair repeat of the U3 region. J Virol 2008; 83:1152-5. [PMID: 18987157 DOI: 10.1128/jvi.01627-08] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The nucleotide sequence of the highly divergent small-ruminant lentivirus genotype E has been determined. The full genome consists of 8,418 nucleotides and lacks two large portions corresponding nearly to the entire dUTPase subunit of the pol and vpr-like accessory genes. Moreover, the 70-bp repeat of the U3 region of the long terminal repeat was observed to be deleted. Interestingly, this lentivirus genotype is able to persist in a local breed population, and retrospective analysis revealed its presence in milk samples collected in 1999. gag sequences obtained from a flock coinfected with the B1 and E genotypes revealed that the evolutionary rates of the two viruses were quite similar. Since a reduced viral load and/or disease progression was observed for viruses with artificially deleted dUTPase and vpr-like genes, it is proposed that this viral cluster be designated a low-pathogenicity caprine lentivirus.
Collapse
|
30
|
Németh-Pongrácz V, Snasel J, Rumlova M, Pichova I, Vértessy BG. Interacting partners of M-PMV nucleocapsid-dUTPase. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2007; 25:1197-200. [PMID: 17065090 DOI: 10.1080/15257770600894535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The nucleocapsid-dUTPase protein of Mason-Pfizer monkey virus is a truly bifunctional fusion enzyme. The exact role of this fusion protein in the viral life cycle is unclear. To explore its function, we started to identify interacting protein partners of the enzyme in vitro. Three viral proteins, integrase, capsid and nucleocapsid, were found to be capable of physical interaction with NC-dUTPase. Integrase protein is an important component within the preintegration complex; therefore the present results also suggest that NC-dUTPase might be associated with this complex.
Collapse
Affiliation(s)
- V Németh-Pongrácz
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
31
|
Zhao RY, Elder RT, Bukrinsky M. Interactions of HIV-1 viral protein R with host cell proteins. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2007; 55:233-60. [PMID: 17586317 DOI: 10.1016/s1054-3589(07)55007-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Richard Y Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
32
|
Németh-Pongrácz V, Barabás O, Fuxreiter M, Simon I, Pichová I, Rumlová M, Zábranská H, Svergun D, Petoukhov M, Harmat V, Klement É, Hunyadi-Gulyás É, Medzihradszky KF, Kónya E, Vértessy BG. Flexible segments modulate co-folding of dUTPase and nucleocapsid proteins. Nucleic Acids Res 2006; 35:495-505. [PMID: 17169987 PMCID: PMC1802613 DOI: 10.1093/nar/gkl1074] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The homotrimeric fusion protein nucleocapsid (NC)-dUTPase combines domains that participate in RNA/DNA folding, reverse transcription, and DNA repair in Mason-Pfizer monkey betaretrovirus infected cells. The structural organization of the fusion protein remained obscured by the N- and C-terminal flexible segments of dUTPase and the linker region connecting the two domains that are invisible in electron density maps. Small-angle X-ray scattering reveals that upon oligonucleotide binding the NC domains adopt the trimeric symmetry of dUTPase. High-resolution X-ray structures together with molecular modeling indicate that fusion with NC domains dramatically alters the conformation of the flexible C-terminus by perturbing the orientation of a critical β-strand. Consequently, the C-terminal segment is capable of double backing upon the active site of its own monomer and stabilized by non-covalent interactions formed with the N-terminal segment. This co-folding of the dUTPase terminal segments, not observable in other homologous enzymes, is due to the presence of the fused NC domain. Structural and genomic advantages of fusing the NC domain to a shortened dUTPase in betaretroviruses and the possible physiological consequences are envisaged.
Collapse
Affiliation(s)
| | | | | | | | - Iva Pichová
- Institute of Chemistry and Biochemistry, Czech Academy of SciencesPrague, Czech Republic
| | - Michalea Rumlová
- Institute of Chemistry and Biochemistry, Czech Academy of SciencesPrague, Czech Republic
| | - Helena Zábranská
- Institute of Chemistry and Biochemistry, Czech Academy of SciencesPrague, Czech Republic
| | - Dmitri Svergun
- European Molecular Biology Laboratory, Hamburg OutstationHamburg, Germany, and Institute of Crystallography, Russian Academy of Sciences, Moscow, Russia
| | - Maxim Petoukhov
- European Molecular Biology Laboratory, Hamburg OutstationHamburg, Germany, and Institute of Crystallography, Russian Academy of Sciences, Moscow, Russia
| | - Veronika Harmat
- Hungarian Academy of Sciences-Eotvos Lorand University, Protein Modeling Research GroupBudapest, Hungary
| | - Éva Klement
- Proteomics Laboratory, Biological Research CenterHungarian Academy of Sciences, Szeged, Hungary
| | - Éva Hunyadi-Gulyás
- Proteomics Laboratory, Biological Research CenterHungarian Academy of Sciences, Szeged, Hungary
| | | | | | - Beáta G. Vértessy
- To whom correspondence should be addressed. Tel: 36 12793116; Fax: 36 14665465;
| |
Collapse
|
33
|
Zhao Z, Ke F, Gui J, Zhang Q. Characterization of an early gene encoding for dUTPase in Rana grylio virus. Virus Res 2006; 123:128-37. [PMID: 16989917 DOI: 10.1016/j.virusres.2006.08.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 08/18/2006] [Accepted: 08/18/2006] [Indexed: 11/22/2022]
Abstract
dUTPase (DUT) is a ubiquitous and important enzyme responsible for regulating levels of dUTP. Here, an iridovirus DUT was identified and characterized from Rana grylio virus (RGV) which is a pathogen agent in pig frog. The DUT encodes a protein of 164aa with a predicted molecular mass of 17.4 kDa, and its transcriptional initiation site was determined by 5'RACE to start from the nucleotide A at 15 nt upstream of the initiation codon ATG. Sequence comparisons and multiple alignments suggested that RGV DUT was quite similar to other identified DUTs that function as homotrimers. Phylogenetic analysis implied that DUT horizontal transfers might have occurred between the vertebrate hosts and iridoviruses. Furthermore, its temporal expression pattern during RGV infection course was characterized by RT-PCR and Western blot analysis. It begins to transcribe and translate as early as 4h postinfection (p.i.), and remains detectable at 48 h p.i. DUT-EGFP fusion protein was observed in the cytoplasm of pEGFP-N3-Dut transfected EPC cells. Immunofluorescence also confirmed DUT cytoplasm localization in RGV-infected cells. Using drug inhibition analysis by a de novo protein synthesis inhibitor (cycloheximide) and a viral DNA replication inhibitor (cytosine arabinofuranoside), RGV DUT was classified as an early (E) viral gene during the in vitro infection. Moreover, RGV DUT overexpression was shown that there was no effect on RGV replication by viral replication kinetics assay.
Collapse
Affiliation(s)
- Zhe Zhao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Wuhan 430072, China
| | | | | | | |
Collapse
|
34
|
Barabás O, Németh V, Vértessy BG. Crystallization and preliminary X-ray studies of dUTPase from Mason-Pfizer monkey retrovirus. Acta Crystallogr Sect F Struct Biol Cryst Commun 2006; 62:399-401. [PMID: 16582495 PMCID: PMC2222558 DOI: 10.1107/s1744309106008931] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Accepted: 03/09/2006] [Indexed: 11/10/2022]
Abstract
Deoxyuridine 5'-triphosphate nucleotidohydrolase from Mason-Pfizer monkey retrovirus (M-PMV dUTPase) is a betaretroviral member of the dUTPase enzyme family. In the mature M-PMV virion, this enzyme is present as the C-terminal domain of the fusion protein nucleocapsid-dUTPase. The homotrimeric organization characteristic of dUTPases is retained in this bifunctional fusion protein. The fusion protein supposedly plays a role in adequate localization of dUTPase activity in the vicinity of nucleic acids during reverse transcription and integration. Here, the nucleocapsid-free dUTPase (48 426 Da) was cocrystallized with a dUTP substrate analogue using the hanging-drop vapour-diffusion method. The obtained crystals belong to the primitive hexagonal space group P6(3), with unit-cell parameters a = 60.6, b = 60.6, c = 63.6 angstroms, alpha = 90, beta = 90, gamma = 120 degrees. Native and PtCl4-derivative data sets were collected using synchrotron radiation to 1.75 and 2.3 angstroms, respectively. Phasing was successfully performed by isomorphous replacement combined with anomalous scattering.
Collapse
Affiliation(s)
- Orsolya Barabás
- Institute of Enzymology, BRC, Hungarian Academy of Sciences, Budapest, Karolina út 29, H-1113, Hungary
| | - Veronika Németh
- Institute of Enzymology, BRC, Hungarian Academy of Sciences, Budapest, Karolina út 29, H-1113, Hungary
| | - Beáta G. Vértessy
- Institute of Enzymology, BRC, Hungarian Academy of Sciences, Budapest, Karolina út 29, H-1113, Hungary
- Correspondence e-mail:
| |
Collapse
|
35
|
St-Louis MC, Cojocariu M, Archambault D. The molecular biology of bovine immunodeficiency virus: a comparison with other lentiviruses. Anim Health Res Rev 2005; 5:125-43. [PMID: 15984320 DOI: 10.1079/ahr200496] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bovine immunodeficiency virus (BIV) was first isolated in 1969 from a cow, R-29, with a wasting syndrome. The virus isolated induced the formation of syncytia in cell cultures and was structurally similar to maedi-visna virus. Twenty years later, it was demonstrated that the bovine R-29 isolate was indeed a lentivirus with striking similarity to the human immunodeficiency virus. Like other lentiviruses, BIV has a complex genomic structure characterized by the presence of several regulatory/accessory genes that encode proteins, some of which are involved in the regulation of virus gene expression. This manuscript aims to review biological and, more particularly, molecular aspects of BIV, with emphasis on regulatory/accessory viral genes/proteins, in comparison with those of other lentiviruses.
Collapse
Affiliation(s)
- Marie-Claude St-Louis
- University of Québec at Montréal, Department of Biological Sciences, Montréal, Québec, Canada
| | | | | |
Collapse
|
36
|
Zhang Y, Moriyama H, Homma K, Van Etten JL. Chlorella virus-encoded deoxyuridine triphosphatases exhibit different temperature optima. J Virol 2005; 79:9945-53. [PMID: 16014955 PMCID: PMC1181562 DOI: 10.1128/jvi.79.15.9945-9953.2005] [Citation(s) in RCA: 9] [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
A putative deoxyuridine triphosphatase (dUTPase) gene from chlorella virus PBCV-1 was cloned, and the recombinant protein was expressed in Escherichia coli. The recombinant protein has dUTPase activity and requires Mg(2+) for optimal activity, while it retains some activity in the presence of other divalent cations. Kinetic studies of the enzyme revealed a K(m) of 11.7 microM, a turnover k(cat) of 6.8 s(-1), and a catalytic efficiency of k(cat)/K(m) = 5.8 x 10(5) M(-1) s(-1). dUTPase genes were cloned and expressed from two other chlorella viruses IL-3A and SH-6A. The two dUTPases have similar properties to PBCV-1 dUTPase except that IL-3A dUTPase has a lower temperature optimum (37 degrees C) than PBCV-1 dUTPase (50 degrees C). The IL-3A dUTPase differs from the PBCV-1 enzyme by nine amino acids, including two amino acid substitutions, Glu81-->Ser81 and Thr84-->Arg84, in the highly conserved motif III of the proteins. To investigate the difference in temperature optima between the two enzymes, homology modeling and docking simulations were conducted. The results of the simulation and comparisons of amino acid sequence suggest that adjacent amino acids are important in the temperature optima. To confirm this suggestion, three site-directed amino acid substitutions were made in the IL-3A enzyme: Thr84-->Arg84, Glu81-->Ser81, and Glu81-->Ser81 plus Thr84-->Arg84. The single substitutions affected the optimal temperature for enzyme activity. The temperature optimum increased from 37 to 55 degrees C for the enzyme containing the two amino acid substitutions. We postulate that the change in temperature optimum is due to reduction in charge and balkiness in the active cavity that allows more movement of the ligand and protein before the enzyme and substrate complex is formed.
Collapse
Affiliation(s)
- Yuanzheng Zhang
- Department of Plant Pathology, University of Nebraska-Lincoln, 68583-0722, USA
| | | | | | | |
Collapse
|
37
|
Priet S, Gros N, Navarro JM, Boretto J, Canard B, Quérat G, Sire J. HIV-1-associated uracil DNA glycosylase activity controls dUTP misincorporation in viral DNA and is essential to the HIV-1 life cycle. Mol Cell 2005; 17:479-90. [PMID: 15721252 DOI: 10.1016/j.molcel.2005.01.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Revised: 11/26/2004] [Accepted: 01/21/2005] [Indexed: 01/13/2023]
Abstract
Uracilation of DNA represents a constant threat to the survival of many organisms including viruses. Uracil may appear in DNA either by cytosine deamination or by misincorporation of dUTP. The HIV-1-encoded Vif protein controls cytosine deamination by preventing the incorporation of host-derived APOBEC3G cytidine deaminase into viral particles. Here, we show that the host-derived uracil DNA glycosylase UNG2 enzyme, which is recruited into viral particles by the HIV-1-encoded integrase domain, is essential to the viral life cycle. We demonstrate that virion-associated UNG2 catalytic activity can be replaced by the packaging of heterologous dUTPase into virion, indicating that UNG2 acts to counteract dUTP misincorporation in the viral genome. Therefore, HIV-1 prevents incorporation of dUTP in viral cDNA by UNG2-mediated uracil excision followed by a dNTP-dependent, reverse transcriptase-mediated endonucleolytic cleavage and finally by strand-displacement polymerization. Our findings indicate that pharmacologic strategies aimed toward blocking UNG2 packaging should be explored as potential HIV/AIDS therapeutics.
Collapse
Affiliation(s)
- Stéphane Priet
- Pathogénie des Infections à Lentivirus, INSERM U372, Université de la Méditerranée, 163 Avenue de Luminy, BP 178, 13276 Marseille Cedex 9, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Neuberger MS, Di Noia JM, Beale RCL, Williams GT, Yang Z, Rada C. Somatic hypermutation at A.T pairs: polymerase error versus dUTP incorporation. Nat Rev Immunol 2005; 5:171-8. [PMID: 15688043 DOI: 10.1038/nri1553] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Somatic hypermutation of immunoglobulin genes occurs at both C.G pairs and A.T pairs. Mutations at C.G pairs are created by activation-induced deaminase (AID)-catalysed deamination of C residues to U residues. Mutations at A.T pairs are probably produced during patch repair of the AID-generated U.G lesion, but they occur through an unknown mechanism. Here, we compare the popular suggestion of nucleotide mispairing through polymerase error with an alternative possibility, mutation through incorporation of dUTP (or another non-canonical nucleotide).
Collapse
Affiliation(s)
- Michael S Neuberger
- Medical Research Council Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Chen R, Le Rouzic E, Kearney JA, Mansky LM, Benichou S. Vpr-mediated incorporation of UNG2 into HIV-1 particles is required to modulate the virus mutation rate and for replication in macrophages. J Biol Chem 2004; 279:28419-25. [PMID: 15096517 DOI: 10.1074/jbc.m403875200] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human immunodeficiency virus type 1 is able to infect nondividing cells, such as macrophages, and the viral Vpr protein has been shown to participate in this process. Here, we investigated the impact of the recruitment into virus particles of the nuclear form of uracil DNA glycosylase (UNG2), a cellular DNA repair enzyme, on the virus mutation rate and on replication in macrophages. We demonstrate that the interaction of Vpr with UNG2 led to virion incorporation of a catalytically active enzyme that is directly involved with Vpr in modulating the virus mutation rate. The lack of UNG in virions during virus replication in primary monocyte-derived macrophages further exacerbated virus mutant frequencies to an 18-fold increase compared with the 4-fold increase measured in actively dividing cells. Because the presence of UNG is also critical for efficient infection of macrophages, these observations extend the role of Vpr to another early step of the virus life cycle, e.g. viral DNA synthesis, that is essential for replication of human immunodeficiency virus type 1 in nondividing cells.
Collapse
Affiliation(s)
- Renxiang Chen
- Ohio State University Biochemistry Graduate Program, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
40
|
Kovári J, Barabás O, Takács E, Békési A, Dubrovay Z, Pongrácz V, Zagyva I, Imre T, Szabó P, Vértessy BG. Altered active site flexibility and a structural metal-binding site in eukaryotic dUTPase: kinetic characterization, folding, and crystallographic studies of the homotrimeric Drosophila enzyme. J Biol Chem 2004; 279:17932-44. [PMID: 14724274 DOI: 10.1074/jbc.m313643200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
dUTPase is responsible for preventive DNA repair via exclusion of uracil. Developmental regulation of the Drosophila enzyme is suggested to be involved in thymine-less apoptosis. Here we show that in addition to conserved dUTPase sequence motifs, the gene of Drosophila enzyme codes for a unique Ala-Pro-rich segment. Kinetic and structural analyses of the recombinant protein and a truncation mutant show that the Ala-Pro segment is flexible and has no regulatory role in vitro. The homotrimer enzyme unfolds reversibly as a trimeric entity with a melting temperature of 54 degrees C, 23 degrees C lower than Escherichia coli dUTPase. In contrast to the bacterial enzyme, Mg(2+) binding modulates conformation of fly dUTPase, as identified by spectroscopy and by increment in melting temperature. A single well folded, but inactive, homotrimeric core domain is generated through three distinct steps of limited trypsinolysis. In fly, but not in bacterial dUTPase, binding of the product dUMP induces protection against proteolysis at the tryptic site reflecting formation of the catalytically competent closed conformer. Crystallographic analysis argues for the presence of a stable monomer of Drosophila dUTPase in crystal phase. The significant differences between prototypes of eukaryotic and prokaryotic dUTPases with respect to conformational flexibility of the active site, substrate specificity, metal ion binding, and oligomerization in the crystal phase are consistent with alteration of the catalytic mechanism and hydropathy of subunit interfaces.
Collapse
Affiliation(s)
- Júlia Kovári
- Institute of Enzymology, Biological Research Center (BRC), Hungarian Academy of Sciences, POB 7, H-1518, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Barabás O, Rumlová M, Erdei A, Pongrácz V, Pichová I, Vértessy BG. dUTPase and nucleocapsid polypeptides of the Mason-Pfizer monkey virus form a fusion protein in the virion with homotrimeric organization and low catalytic efficiency. J Biol Chem 2003; 278:38803-12. [PMID: 12869552 DOI: 10.1074/jbc.m306967200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Betaretroviruses encode dUTPase, an essential factor in DNA metabolism and repair, in the pro open reading frame located between gag and pol. Ribosomal frame-shifts during expression of retroviral proteins provide a unique possibility for covalent joining of nucleocapsid (NC) and dUTPase within Gag-Pro polyproteins. By developing an antibody against the prototype betaretrovirus Mason-Pfizer monkey virus dUTPase, we demonstrate that i) the NC-dUTPase fusion protein exists both within the virions and infected cells providing the only form of dUTPase, and ii) the retroviral protease does not cleave NC-dUTPase either in the virion or in vitro. We show that recombinant betaretroviral NC-dUTPase and dUTPase are both inefficient catalysts compared with all other dUTPases. Dynamic light scattering and gel filtration confirm that the homotrimeric organization, common among dUTPases, is retained in the NC-dUTPase fusion protein. The betaretroviral dUTPase has been crystallized and single crystals contain homotrimers. Oligonucleotide and Zn2+ binding is well retained in the fusion protein, which is the first example of acquisition of a functional nucleic acid binding module by the DNA repair factor dUTPase. Binding of the hexanucleotide ACTGCC or the octanucleotide (TG)4 to NC-dUTPase modulates enzymatic function, indicating that the low catalytic activity may be compensated by adequate localization.
Collapse
Affiliation(s)
- Orsolya Barabás
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, POB 7, H-1518, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
42
|
Bahr U, Schöndorf E, Handermann M, Darai G. Molecular anatomy of Tupaia (tree shrew) adenovirus genome; evolution of viral genes and viral phylogeny. Virus Genes 2003; 27:29-48. [PMID: 12913356 DOI: 10.1023/a:1025120418159] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Adenoviruses are globally spread and infect species in all five taxons of vertebrates. Outstanding attention is focused on adenoviruses because of their transformation potential, their possible usability as vectors in gene therapy and their applicability in studies dealing with, e.g. cell cycle control, DNA replication, transcription, splicing, virus-host interactions, apoptosis, and viral evolution. The accumulation of genetic data provides the basis for the increase of our knowledge about adenoviruses. The Tupaia adenovirus (TAV) infects members of the genus Tupaiidae that are frequently used as laboratory animals in behavior research dealing with questions about biological and molecular processes of stress in mammals, in neurobiological and physiological studies, and as model organisms for human hepatitis B and C virus infections. In the present study the TAV genome underwent an extensive analysis including determination of codon usage, CG depletion, gene content, gene arrangement, potential splice sites, and phylogeny. The TAV genome has a length of 33,501 bp with a G+C content of 49.96%. The genome termini show a strong CG depletion that could be due to methylation of these genome regions during the viral replication cycle. The analysis of the coding capacity of the complete TAV genome resulted in the identification of 109 open reading frames (ORFs), of which 38 were predicted to be real viral genes. TAV was classified within the genus Mastadenovirus characterized by typical gene content, arrangement, and homology values of 29 conserved ORFs. Phylogenetic trees show that TAV is part of a separate evolutionary lineage and no mastadenovirus species can be considered as the most related. In contrast to other mastadenoviruses a direct ancestor of TAV captured a DUT gene from its mammalian host, presumably controlling local dUTP levels during replication and enhance viral replication in non-dividing host tissues. Furthermore, TAV possesses a second DNA-binding protein gene, that is likely to play a role in the determination of the host range. In view of these data it is conceivable that TAV underwent evolutionary adaptations to its biological environment resulting in the formation of special genomic components that provided TAV with the ability to expand its host range during viral evolution.
Collapse
Affiliation(s)
- Udo Bahr
- Hygiene-Institute, Department of Virology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Federal Republic of Germany
| | | | | | | |
Collapse
|
43
|
Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, Trono D. Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 2003; 424:99-103. [PMID: 12808466 DOI: 10.1038/nature01709] [Citation(s) in RCA: 1185] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Accepted: 05/13/2003] [Indexed: 11/09/2022]
Abstract
Viral replication usually requires that innate intracellular lines of defence be overcome, a task usually accomplished by specialized viral gene products. The virion infectivity factor (Vif) protein of human immunodeficiency virus (HIV) is required during the late stages of viral production to counter the antiviral activity of APOBEC3G (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G; also known as CEM15), a protein expressed notably in human T lymphocytes. When produced in the presence of APOBEC3G, vif-defective virus is non-infectious. APOBEC3G is closely related to APOBEC1, the central component of an RNA-editing complex that deaminates a cytosine residue in apoB messenger RNA. APOBEC family members also have potent DNA mutator activity through dC deamination; however, whether the editing potential of APOBEC3G has any relevance to HIV inhibition is unknown. Here, we demonstrate that it does, as APOBEC3G exerts its antiviral effect during reverse transcription to trigger G-to-A hypermutation in the nascent retroviral DNA. We also find that APOBEC3G can act on a broad range of retroviruses in addition to HIV, suggesting that hypermutation by editing is a general innate defence mechanism against this important group of pathogens.
Collapse
MESH Headings
- APOBEC-3G Deaminase
- Antiviral Agents/metabolism
- Base Sequence
- Cell Line
- Cytidine Deaminase
- DNA, Viral/biosynthesis
- DNA, Viral/genetics
- Gene Products, vif/deficiency
- Gene Products, vif/genetics
- Gene Products, vif/metabolism
- HIV-1/genetics
- HIV-1/physiology
- Humans
- Molecular Sequence Data
- Mutagenesis/genetics
- Nucleoside Deaminases
- Point Mutation/genetics
- Proteins/genetics
- Proteins/metabolism
- RNA Editing/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Repressor Proteins
- Transcription, Genetic/genetics
- Viral Load
- Virus Replication
- vif Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Bastien Mangeat
- Department of Genetics and Microbiology, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
44
|
Priet S, Navarro JM, Gros N, Quérat G, Sire J. Differential incorporation of uracil DNA glycosylase UNG2 into HIV-1, HIV-2, and SIV(MAC) viral particles. Virology 2003; 307:283-9. [PMID: 12667798 DOI: 10.1016/s0042-6822(02)00073-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously reported that the host uracil DNA glycosylase UNG2 enzyme is incorporated into HIV-1 virions via a specific association with the viral integrase (IN) domain of Gag-Pol precursor. In this study, we investigated whether UNG2 was packaged into two phylogenetically closely related primate lentiviruses, HIV-2(ROD) and SIV(MAC239). We demonstrated by GST-pull-down and coprecipitation assays that INs from HIV-1, HIV-2(ROD), and SIV(MAC239) associated with UNG2, although the interaction of UNG2 with HIV-2(ROD) IN and SIV(MAC239) IN was less strong than with HIV-1 IN. We then showed by Western blotting that highly purified HIV-2 and SIV(MAC) viral particles did not incorporate host UNG2, contrasting with the presence of UNG2 in HIV-1 viral particles. Finally, we showed that HIV-1/SIV chimeric viruses in which residues 6 to 202 of HIV-1 IN were replaced by the SIV counterpart were impaired for packaging of UNG2, indicating that the incorporation of host UNG2 into viral particles is the hallmark of the HIV-1 strain. Moreover, we found that HIV-1/SIV IN chimeric viruses were deficient for viral propagation.
Collapse
Affiliation(s)
- Stéphane Priet
- Pathogénie des Infections à Lentivirus, INSERM U372, Marseille, France
| | | | | | | | | |
Collapse
|
45
|
Klarmann GJ, Chen X, North TW, Preston BD. Incorporation of uracil into minus strand DNA affects the specificity of plus strand synthesis initiation during lentiviral reverse transcription. J Biol Chem 2003; 278:7902-9. [PMID: 12458216 DOI: 10.1074/jbc.m207223200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many retroviruses either encode dUTP pyrophosphatase (dUTPase) or package host-derived uracil DNA glycosylase as a means to limit the accumulation of uracil in DNA strands, suggesting that uracil is detrimental to one or more steps in the viral life cycle. In the present study, the effects of DNA uracilation on (-) strand DNA synthesis, RNase H activity, and (+) strand DNA synthesis were investigated in a cell-free system. This system uses the activities of purified human immunodeficiency virus type 1 (HIV-1) reverse transcriptase to convert single-stranded RNA to double-stranded DNA in a single reaction mixture. Substitution of dUTP for dTTP had no effect on (-) strand synthesis but significantly decreased yields of (+) strand DNA. Mapping of nascent (+) strand 5' ends revealed that this was due to decreased initiation from polypurine tracts with a concomitant increase in initiation at non-polypurine tract sites. Aberrant initiation correlated with a change in RNase H cleavage specificity when assayed on preformed RNA-DNA duplexes containing uracilated DNA, suggesting that appropriate "selection" of the (+) strand primer is affected. Collectively, these data suggest that accumulation of uracil in retroviral DNA may disrupt the viral life cycle by altering the specificity of (+) strand DNA synthesis initiation during reverse transcription.
Collapse
Affiliation(s)
- George J Klarmann
- Eccles Institute of Human Genetics and the Department of Biochemistry, University of Utah, Salt Lake City 84112, USA.
| | | | | | | |
Collapse
|
46
|
Priet S, Navarro JM, Gros N, Querat G, Sire J. Functional role of HIV-1 virion-associated uracil DNA glycosylase 2 in the correction of G:U mispairs to G:C pairs. J Biol Chem 2003; 278:4566-71. [PMID: 12458223 DOI: 10.1074/jbc.m209311200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human monocytes/macrophages are target cells for HIV-1 infection. As other non-dividing cells, they are characterized by low and imbalanced intracellular dNTP pool levels and an excess of dUTP. The replication of HIV-1 in this cellular context favors misincorporation of uracil residues into viral DNA because of the use of dUTP in place of dCTP. We have previously reported that the host uracil DNA glycosylase enzyme UNG2 is packaged into HIV-1 viral particles via a specific association with the integrase domain of the Gag-Pol precursor. In this study, we investigated whether virion-associated UNG2 plays a role similar to that of its cellular counterpart. We show that the L172A mutation of integrase impaired the packaging of UNG2 into viral particles. Using a primer-template DNA substrate containing G:U mispairs, we demonstrate that wild-type viral lysate has the ability to repair G:U mismatched pairs to G:C matched pairs, in contrast to UNG2-deficient viral lysate. Moreover, no correction of G:T mispairs by wild-type HIV-1 viral lysate was observed, which argues for the specificity of the repair process. We also show that UNG2 physically associates with the viral reverse transcriptase enzyme. Altogether our data indicate for the first time that a uracil repair pathway is specifically associated with HIV-1 viral particles. However, the molecular mechanism of this process remains to be characterized further.
Collapse
Affiliation(s)
- Stephane Priet
- Pathogénie des Infections à Lentivirus, INSERM U372, 163 avenue de Luminy, BP 178, 13276 Marseille Cedex 09, France
| | | | | | | | | |
Collapse
|
47
|
Abstract
Herpesviruses and poxviruses are known to encode the DNA repair enzyme uracil-DNA glycosylase (UNG), an enzyme involved in the base excision repair pathway that specifically removes the RNA base uracil from DNA, while at least one retrovirus (human immunodeficiency virus type 1) packages cellular UNG into virus particles. In these instances, UNG is implicated as being important in virus replication. However, a clear understanding of the role(s) of UNG in virus replication remains elusive. Herpesviruses, poxviruses and some retroviruses encode dUTPase, an enzyme that can minimize the misincorporation of uracil into DNA. The encoding of dUTPase by these viruses also implies their importance in virus replication. An understanding at the molecular level of how these viruses replicate in non-dividing cells should provide clues to the biological relevance of UNG and dUTPase function in virus replication.
Collapse
Affiliation(s)
- Renxiang Chen
- Ohio State University Biochemistry Graduate Program, Ohio State University, USA2
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research, and Comprehensive Cancer Center, Ohio State University Medical Center, 2078 Graves Hall, 333 West 10th Ave, Columbus, OH 43210, USA1
| | - Huating Wang
- Molecular, Cellular, and Developmental Biology Graduate Program, Ohio State University, USA3
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research, and Comprehensive Cancer Center, Ohio State University Medical Center, 2078 Graves Hall, 333 West 10th Ave, Columbus, OH 43210, USA1
| | - Louis M Mansky
- Molecular, Cellular, and Developmental Biology Graduate Program, Ohio State University, USA3
- Ohio State University Biochemistry Graduate Program, Ohio State University, USA2
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research, and Comprehensive Cancer Center, Ohio State University Medical Center, 2078 Graves Hall, 333 West 10th Ave, Columbus, OH 43210, USA1
| |
Collapse
|
48
|
Cottone R, Büttner M, McInnes CJ, Wood AR, Rziha HJ. Orf virus encodes a functional dUTPase gene. J Gen Virol 2002; 83:1043-1048. [PMID: 11961258 DOI: 10.1099/0022-1317-83-5-1043] [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: 11/18/2022] Open
Abstract
The present study is the first report on the functional activity of a parapoxvirus-encoded dUTPase. The dUTPase gene of the attenuated orf virus (ORFV), strain D1701, was expressed as a bacterial thioredoxin fusion protein. In vitro assays showed that ORFV dUTPase was highly specific for dUTP as substrate. The enzyme was active over a broad pH range (pH 6.0-9.0), with maximal enzymatic activity at pH 7.0 in the presence of Mg(2+) cations. Kinetic studies of the recombinant ORFV dUTPase revealed an apparent K(m) of 4.0 microM, which is more similar to that of the mammalian or African swine fever virus enzyme than to the K(m) of vaccinia virus dUTPase. Enzyme activity was also found with purified ORFV particles, indicating its virion association.
Collapse
Affiliation(s)
- R Cottone
- Federal Research Centre for Virus Diseases of Animals, Institute for Immunology, Paul-Ehrlich-Straße 28, D-72076 Tübingen, Federal Republic of Germany1
| | - M Büttner
- Federal Research Centre for Virus Diseases of Animals, Institute for Immunology, Paul-Ehrlich-Straße 28, D-72076 Tübingen, Federal Republic of Germany1
| | - C J McInnes
- Moredun Research Institute, International Research Centre, Pentlands Science Park, Penicuik, Midlothian EH26 OPZ, UK2
| | - A R Wood
- Moredun Research Institute, International Research Centre, Pentlands Science Park, Penicuik, Midlothian EH26 OPZ, UK2
| | - H-J Rziha
- Federal Research Centre for Virus Diseases of Animals, Institute for Immunology, Paul-Ehrlich-Straße 28, D-72076 Tübingen, Federal Republic of Germany1
| |
Collapse
|
49
|
Rohll JB, Mitrophanous KA, Martin-Rendon E, Ellard FM, Radcliffe PA, Mazarakis ND, Kingsman SM. Design, production, safety, evaluation, and clinical applications of nonprimate lentiviral vectors. Methods Enzymol 2002; 346:466-500. [PMID: 11883086 DOI: 10.1016/s0076-6879(02)46072-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Jonathan B Rohll
- Department of Biochemistry, Oxford BioMedica (UK) Limited, Oxford OX4 4GA, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
50
|
Rutkoski J, Werenicz R, Reischak D, Wendelstein A, Moojen V, Ravazzolo A. Detecção da infecção pelo vírus da artrite-encefalite caprina: imunodifusão em ágar e reação em cadeia da polimerase com "primers" degenerados. ARQ BRAS MED VET ZOO 2001. [DOI: 10.1590/s0102-09352001000600001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
O objetivo deste trabalho foi analisar amostras de soro e de células sangüíneas de caprinos para detecção de anticorpos e DNA proviral do vírus da artrite-encefalite caprina (CAEV), respectivamente. Utilizou-se a técnica de imunodifusão em ágar (AGID) e a reação em cadeia da polimerase (PCR) com "primers" degenerados. Foram analisadas amostras de diferentes procedências: 39 de Mato Grosso do Sul (MS), 19 de São Paulo (SP) e 22 do Ceará (CE), dessas últimas, 12 oriundas de animais importados do Canadá. Os resultados de AGID e PCR foram discordantes, pois o primeiro permitiu a detecção de 25 animais soropositivos, enquanto a PCR detectou DNA proviral de CAEV em 16 amostras. Pela PCR foi possível identificar animais infectados cujos testes sorológicos foram negativos pelo AGID: oito amostras do MS e um do CE. São discutidos diferentes aspectos que poderiam estar envolvidos na discordância dos resultados.
Collapse
|