Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Virol ; 96(9): e0032122, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35412345

ABSTRACT

Circular RNAs (circRNAs) are a recently rediscovered class of functional noncoding RNAs that are involved in gene regulation and cancer development. Next-generation sequencing approaches identified circRNA fragments and sequences underlying circularization events in virus-induced cancers. In the present study, we performed viral circRNA expression analysis and full-length sequencing in infections with Marek's disease virus (MDV), which serves as a model for herpesvirus-induced tumorigenesis. We established inverse PCRs to identify and characterize circRNA expression from the repeat regions of the MDV genome during viral replication, latency, and reactivation. We identified a large variety of viral circRNAs through precise mapping of full-length circular transcripts and detected matching sequences with several viral genes. Hot spots of circRNA expression included the transcriptional unit of the major viral oncogene encoding the Meq protein and the latency-associated transcripts (LATs). Moreover, we performed genome-wide bioinformatic analyses to extract back-splice junctions from lymphoma-derived samples. Using this strategy, we found that circRNAs were abundantly expressed in vivo from the same key virulence genes. Strikingly, the observed back-splice junctions do not follow a unique canonical pattern, compatible with the U2-dependent splicing machinery. Numerous noncanonical junctions were observed in viral circRNA sequences characterized from in vitro and in vivo infections. Given the importance of the genes involved in the transcription of these circRNAs, our study contributes to our understanding and complexity of this deadly pathogen. IMPORTANCE Circular RNAs (circRNAs) were rediscovered in recent years both in physiological and pathological contexts, such as in cancer. Viral circRNAs are encoded by at least two human herpesviruses, the Epstein Barr virus and the Kaposi's Sarcoma-associated herpesvirus, both associated with the development of lymphoma. Marek's disease virus (MDV) is a well-established animal model to study virus-induced lymphoma but circRNA expression has not been reported for MDV yet. Our study provided the first evidence of viral circRNAs that were expressed at key steps of the MDV lifecycle using genome-wide analyses of circRNAs. These circRNAs were primarily found in transcriptional units that corresponded to the major MDV virulence factors. In addition, we established a bioinformatics pipeline that offers a new tool to identify circular RNAs in other herpesviruses. This study on the circRNAs provided important insights into major MDV virulence genes and herpesviruses-mediated gene dysregulation.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 2, Gallid , Marek Disease , RNA, Circular , Animals , Chickens , Genome-Wide Association Study , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/pathogenicity , Lymphoma/virology , Marek Disease/virology , Oncogene Proteins, Viral/genetics , RNA, Circular/genetics , RNA, Untranslated/genetics , Virulence/genetics
2.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32967954

ABSTRACT

Gallid herpesvirus type 2 (GaHV-2) is an oncogenic alphaherpesvirus that induces malignant T-cell lymphoma in chicken. GaHV-2 encodes a viral telomerase RNA subunit (vTR) that plays a crucial role in virus-induced tumorigenesis, enhances telomerase activity, and possesses functions independent of the telomerase complex. vTR is driven by a robust viral promoter, highly expressed in virus-infected cells, and regulated by two c-Myc response elements (c-Myc REs). The regulatory mechanisms involved in controlling vTR and other genes during viral replication and latency remain poorly understood but are crucial to understanding this oncogenic herpesvirus. Therefore, we investigated DNA methylation patterns of CpG dinucleotides found in the vTR promoter and measured the impact of methylation on telomerase activity. We demonstrated that telomerase activity was considerably increased following viral reactivation. Furthermore, CpG sites within c-Myc REs showed specific changes in methylation after in vitro reactivation and in infected animals over time. Promoter reporter assays indicated that one of the c-Myc REs is involved in regulating vTR transcription, and that methylation strongly influenced vTR promoter activity. To study the importance of the CpG sites found in c-Myc REs in virus-induced tumorigenesis, we generated recombinant virus containing mutations in CpG sites of c-Myc REs together with the revertant virus by two-step Red-mediated mutagenesis. Introduced mutations in the vTR promoter did not affect the replication properties of the recombinant viruses in vitro In contrast, replication of the mutant virus in infected chickens was severely impaired, and tumor formation completely abrogated. Our data provides an in-depth characterization of c-Myc oncoprotein REs and the involvement of DNA methylation in transcriptional regulation of vTR.IMPORTANCE Previous studies demonstrated that telomerase RNAs possess functions that promote tumor development independent of the telomerase complex. vTR is a herpesvirus-encoded telomerase RNA subunit that plays a crucial role in virus-induced tumorigenesis and is expressed by a robust viral promoter that is highly regulated by the c-Myc oncoprotein binding to the E-boxes. Here, we demonstrated that the DNA methylation patterns in the functional c-Myc response elements of the vTR promoter change upon reactivation from latency, and that demethylation strongly increases telomerase activity in virus-infected cells. Moreover, the introduction of mutation in the CpG dinucleotides of the c-Myc binding sites resulted in decreased vTR expression and complete abrogation of tumor formation. Our study provides further confirmation of the involvement of specific DNA methylation patterns in the regulation of vTR expression and vTR importance for virus-induced tumorigenesis.


Subject(s)
DNA Methylation/physiology , Herpesvirus 2, Gallid/genetics , Promoter Regions, Genetic , RNA, Viral/genetics , Telomerase/genetics , Animals , Carcinogenesis/genetics , Cell Line , Chickens , Gene Expression Regulation, Viral , Herpesvirus 2, Gallid/enzymology , Herpesvirus 2, Gallid/pathogenicity , Marek Disease/virology , Mutagenesis, Site-Directed , Mutation , RNA , Virus Replication
3.
J Gen Virol ; 99(9): 1286-1300, 2018 09.
Article in English | MEDLINE | ID: mdl-30067174

ABSTRACT

Herpesviruses have a lifecycle consisting of successive lytic, latent and reactivation phases. Only three infected cell proteins (ICPs) have been described for the oncogenic Marek's disease virus (or Gallid herpes virus 2, GaHV-2): ICP4, ICP22 and ICP27. We focus here on ICP22, confirming its cytoplasmic location and showing that ICP22 is expressed during productive phases of the lifecycle, via a bicistronic transcript encompassing the US10 gene. We also identified the unique promoter controlling ICP22 expression, and its core promoter, containing functional responsive elements including E-box, ETS-1 and GATA elements involved in ICP22 transactivation. ICP22 gene expression was weakly regulated by DNA methylation and activated by ICP4 or ICP27 proteins. We also investigated the function of GaHV-2 ICP22. We found that this protein repressed transcription from its own promoter and from those of IE ICP4 and ICP27, and the late gK promoter. Finally, we investigated posttranscriptional ICP22 regulation by GaHV-2 microRNAs. We found that mdv1-miR-M5-3p and -M1-5p downregulated ICP22 mRNA expression during latency, whereas, unexpectedly, mdv1-miR-M4-5p upregulated the expression of the protein ICP22, indicating a tight regulation of ICP22 expression by microRNAs.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesvirus 2, Gallid/physiology , Viral Proteins/metabolism , Animals , Cell Line , Chickens , DNA Methylation , Promoter Regions, Genetic , Response Elements , Viral Proteins/genetics , Virus Replication
4.
J Gen Virol ; 99(3): 355-368, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29458534

ABSTRACT

Transcriptional and post-transcriptional mechanisms are involved in the switch between the lytic, latent and reactivation phases of the viral cycle in herpesviruses. During the productive phases, herpesvirus gene expression is characterized by a temporally regulated cascade of immediate early (IE), early (E) and late (L) genes. In alphaherpesviruses, the major product of the IE ICP4 gene is a transcriptional regulator that initiates the cascade of gene expression that is essential for viral replication. In this study, we redefine the infected cell protein 4 (ICP4) gene of the oncogenic Marek's disease virus (MDV or gallid herpesvirus 2) as a 9438 nt gene ended with four alternative poly(A) signals and controlled by two alternative promoters containing essentially ubiquitous functional response elements (GC, TATA and CCAAT boxes). The distal promoter is associated with ICP4 gene expression during the lytic and the latent phases, whereas the proximal promoter is associated with the expression of this gene during the reactivation phase. Both promoters are regulated by DNA methylation during the viral cycle and are hypermethylated during latency. Transcript analyses showed ICP4 to consist of three exons and two introns, the alternative splicing of which is associated with five predicted nested ICP4ORFs. We show that the ICP4 gene is highly and specifically regulated by transcriptional and post-transcriptional mechanisms during the three phases of the GaHV-2 viral cycle, with a clear difference in expression between the lytic phase and reactivation from latency in our model.

5.
J Gen Virol ; 97(9): 2399-2410, 2016 09.
Article in English | MEDLINE | ID: mdl-27411695

ABSTRACT

Herpesvirus gene expression is temporally regulated, with immediate early (IE), early (E) and late (L) genes. ICP27, which is involved in post-transcriptional regulation, is the only IE gene product conserved in all herpesviruses. We show here that the ICP27 transcript of the oncogenic Marek's disease virus shares the same polyadenylation signal as the bicistronic glycoprotein K-ICP27 transcript and is regulated by alternative promoter usage, with transcription from its own promoter (pICP27) or that of gK (pgK). The pgK can generate a spliced ICP27 transcript yielding an N-terminal-deleted ICP27 isoform (ICP27ΔN) that, like ICP27, co-localizes with the SR protein in infected cells, but with a diffuse nuclear distribution. The pICP27 includes functional responsive elements (REs) for SP1, AP1 and CREB, is essentially active during the lytic phase and leads to exclusive expression of the native form of ICP27. The alternative promoter, pgK, including active REs for GATA, P53 and CREB, preferentially generates the gK transcript during the lytic phase and the spliced ICP27 transcript (ICP27ΔN) during the latent phase. An analysis of the DNA methylation marks of each promoter showed that pgK was systematically demethylated, whereas pICP27 was methylated during latency and demethylated during the lytic stage. Thus, MDV ICP27 gene expression is dependent on alternative promoters, the usage of which is regulated by DNA methylation, which differs between viral stages.


Subject(s)
Gene Expression Regulation, Viral , Mardivirus/genetics , Mardivirus/metabolism , Promoter Regions, Genetic , Protein Isoforms/biosynthesis , Transcription, Genetic , Viral Proteins/biosynthesis , Animals , Cell Line , Chickens , Protein Isoforms/genetics , Viral Proteins/genetics
6.
Arch Virol ; 161(5): 1353-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26887970

ABSTRACT

A large sheep flock screened over a two-year period showed active spreading of Schmallenberg virus (SBV) during the summers of 2011 and 2012. Transplacental infections were observed during the two associated lambing periods (the winters of 2012 and 2013). Analysis of small (S) segment sequences of 38 SBV-positive samples, collected during periods of viral spreading and lambing revealed intra-herd sequences diversity and sub-consensus variability occurring after transplacental infections. In comparison with the nucleoprotein (N), which appeared to be conserved, the non-structural protein (NSs) showed the highest level of variability at the time of viral emergence and over the two-year analysis period.


Subject(s)
Bunyaviridae Infections/veterinary , Orthobunyavirus/genetics , Sheep Diseases/virology , Animals , Bunyaviridae Infections/virology , Genetic Variation/genetics , Open Reading Frames/genetics , RNA, Viral/genetics , Sheep
7.
Emerg Infect Dis ; 19(7): 1115-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23764090

ABSTRACT

Reemergence of Schmallenberg virus (SBV) occurred among lambs (n = 50) in a sheep flock in Belgium between mid-July and mid-October 2012. Bimonthly assessment by quantitative reverse transcription PCR and seroneutralization demonstrated that 100% of lambs were infected. Viremia duration may be longer in naturally infected than in experimentally infected animals.


Subject(s)
Bunyaviridae Infections/veterinary , Communicable Diseases, Emerging/veterinary , Disease Outbreaks , Orthobunyavirus/genetics , Sheep Diseases/epidemiology , Animals , Belgium/epidemiology , Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/virology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Female , Genes, Viral , Molecular Diagnostic Techniques , Prevalence , Real-Time Polymerase Chain Reaction , Seroepidemiologic Studies , Sheep , Sheep Diseases/virology , Sheep, Domestic/virology
8.
J Gen Virol ; 94(Pt 6): 1168-1174, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23364190

ABSTRACT

Detected for the first time in 2011, Schmallenberg virus (SBV) is an orthobunyavirus of the Simbu serogroup that caused a large outbreak in European ruminants. In a tight time frame, data have been obtained on SBV epidemiology and the clinical pictures associated with this new viral infection, but little information is available on the molecular biology of SBV. In this study, SBV sequence variability was characterized from the central nervous system of two stillborn lambs in a naturally infected herd. A hypervariable region (HVR) was detected in the N-terminal region of the SBV Gc glycoprotein through sequencing and analysis of the two full-length genomes representative of intra-herd SBV dissemination. In vitro growth assays coupled with full-length genome sequencing were performed on the two isolates after successive cellular passages, showing an in vitro adaptation of SBV and mutation accumulation inside the HVR in the absence of immune selective pressure.


Subject(s)
Bunyaviridae Infections/veterinary , Genetic Variation , Orthobunyavirus/genetics , Sheep Diseases/virology , Animals , Base Sequence , Bunyaviridae Infections/virology , Glycoproteins/genetics , Molecular Sequence Data , Mutation , Orthobunyavirus/isolation & purification , Sheep , Viral Proteins/genetics
9.
J Gen Virol ; 94(Pt 11): 2399-2405, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939979

ABSTRACT

Schmallenberg virus (SBV) is an emerging arbovirus infecting ruminants in Europe. SBV belongs to the Bunyaviridae family within the Simbu serogroup. Its genome comprises three segments, small (S), medium (M) and large (L), that together encode six proteins and contain NTRs. NTRs are involved in initiation and termination of transcription and in genome packaging. This study explored the 3' mRNA termini of SBV and related Simbuviruses. In addition, the 5' termini of SBV messenger RNA (mRNA) were characterized. For the three SBV segments, cap-snatching was found to initiate mRNA transcription both in vivo and in vitro. The presence of extraneous nucleotides between host RNA leaders and the viral termini fits with the previously described prime-and-realign theory. At the 3' termini, common features were identified for SBV and related Simbuviruses. However, different patterns were observed for the termini of the three segments from the same virus type.


Subject(s)
3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Orthobunyavirus/genetics , Orthobunyavirus/metabolism , RNA, Messenger/genetics , Simbu virus/genetics , Transcription, Genetic , 3' Untranslated Regions/physiology , 5' Untranslated Regions/physiology , Base Sequence , Molecular Sequence Data , Orthobunyavirus/classification , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Transcription Initiation, Genetic , Transcription Termination, Genetic
10.
Vet Surg ; 42(5): 551-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23711241

ABSTRACT

OBJECTIVE: To describe magnetic resonance imaging (MRI) anatomy of the ovine stifle and investigate meniscotibial and cruciate ligaments anatomy. STUDY DESIGN: Descriptive ex vivo study. ANIMALS: Pelvic limbs (n = 44) from 22 adult Texel ewes. METHODS: Forty limbs (n = 40) were scanned using 3 Tesla MRI before gross anatomic dissection. Two other limb pairs were frozen and transected to obtain sections that were compared with MRI images for identification of anatomic structures. RESULTS: In all stifles, the craniomedial bundle of the cranial cruciate ligament inserted caudally to the cranial attachment of the medial meniscus. No transverse intermeniscal ligament was identified in 80% of stifles, whereas a few small ligamentous fibers were seen crossing from 1 cranial horn to the other in 20% of stifles. There was good differentiation of menisci, ligaments, and synovial cavities on MRI images. Two bundles were identified in all cranial cruciate ligaments on MRI. Sensitivity and specificity of 3T MRI for detection of transverse intermeniscal ligament were 42% and 84%, respectively. CONCLUSION: 3T MRI provided well defined reference images for menisci, synovial cavities, and most ligaments.


Subject(s)
Magnetic Resonance Imaging/veterinary , Sheep/anatomy & histology , Stifle/diagnostic imaging , Animals , Cadaver , Female , Ligaments/anatomy & histology , Radiography , Stifle/anatomy & histology
11.
Med Sci (Paris) ; 39(4): 344-350, 2023 Apr.
Article in French | MEDLINE | ID: mdl-37094267

ABSTRACT

Circular RNAs (circRNA), as ancient as the first viruses, take an important part in the host-pathogen relationship. After the first description of dysregulated cellular circRNAs upon viral infection, numerous circRNAs of viral origin were identified and characterized. They are impacting both viral and cellular cycles and are associated with virus-induced oncogenesis, immune system regulation and cell differentiation. While the naïve reader might get swamped by discovering this new field of RNA biology, it seems that these RNA rings are actually full of surprises and wonders at both a functional and a biogenesis level.


Title: Quel cirque, ces virus. Abstract: Les ARN circulaires (ARNcirc) font partie intégrante de la relation hôte-pathogène. Après la description de la dérégulation d'ARNcirc cellulaires lors d'infections virales, de nombreux ARNcirc d'origine virale ont été identifiés et caractérisés. Des rôles régulateurs, aussi bien du cycle cellulaire que du cycle viral, leur ont été attribués. Ils sont associés à l'oncogenèse viro-induite, à la régulation du système immunitaire et à la différenciation cellulaire. Ces boucles d'ARN, aussi archaïques que les premiers virus, réservent bien des surprises aux chercheurs tant au niveau de leurs fonctions que de leurs biogenèses !


Subject(s)
Virus Diseases , Viruses , Humans , RNA, Circular , RNA/genetics , Virus Diseases/genetics , Genes, Viral , Viruses/genetics
12.
RNA ; 16(11): 2263-76, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20881002

ABSTRACT

The tumor suppressor protein p53 plays a role in cellular responses to cancer-initiating events by regulating progress through the cell cycle. Several recent studies have shown that p53 transactivates expression of the members of the proapoptotic microRNA-34 family, which are underexpressed in several cancers. We demonstrate here that the latency-associated cluster of microRNAs (miRNA) encoded by an oncogenic herpesvirus, gallid herpesvirus 2 (GaHV-2), is a direct target of p53. Robust transcriptional activity was induced in three avian cell lines by a sequence mapping 600 base pairs (bp) upstream of the cluster of miRNAs. We found transcription start sites for the pri-miRNA transcript at the 3' end of this transcription-inducing sequence. The promoter has no consensus core promoter element, but is organized into a variable number of tandem repeats of 60-bp harboring p53-responsive elements (RE). The minimal functional construct consists of two tandem repeats. Mutagenesis to change the sequence of the p53 RE abolished transcriptional activity, whereas p53 induction enhanced mature miRNA expression. The identification of a viral miRNA promoter regulated by p53 is biologically significant, because all avirulent GaHV-2 strains described to date lack the corresponding regulatory sequence, whereas all virulent, very virulent, and hypervirulent strains possess at least two tandem repeats harboring the p53 RE.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Gallid/genetics , MicroRNAs/genetics , Promoter Regions, Genetic , Tandem Repeat Sequences , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Chickens , Herpesvirus 2, Gallid/pathogenicity , Humans , MicroRNAs/metabolism , Molecular Sequence Data , Multigene Family , Polymorphism, Genetic , Protein Binding , Tumor Suppressor Protein p53/genetics , Virulence
13.
Transbound Emerg Dis ; 69(3): 1625-1633, 2022 May.
Article in English | MEDLINE | ID: mdl-33949132

ABSTRACT

Schmallenberg virus emerged in 2011 in Europe. The epicentre of primordial spreading was the region straddling Germany, the Netherlands and Belgium. One of the key questions is whether the newcomer would establish a lasting presence on the continent. The apparent seroprevalence in southern Belgium wild deer populations was followed for 6 years. Two years of intense circulation were revealed, 2012 and 2016, characterized by a peak seroprevalence in the two studied populations (Capreolus capreolus and Cervus elaphus). Between the peak years and after 2016, apparent seroprevalences declined rapidly among adults and became nil among juveniles. The general pattern of apparent seroprevalence evolution observed is consistent with a cyclic circulation of Schmallenberg virus, similar to what is observed for other Orthobunyaviruses in endemic areas. These data also suggest that wild cervids play no central role in the circulation dynamics of the virus.


Subject(s)
Bunyaviridae Infections , Deer , Orthobunyavirus , Animals , Belgium/epidemiology , Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/veterinary , Seroepidemiologic Studies
14.
BMC Vet Res ; 7: 19, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21592326

ABSTRACT

BACKGROUND: Interspecific recombinant viruses R1ΔgC and R2ΔgI were isolated after in vitro co-infection with BoHV-1 and BoHV-5, two closely related alphaherpesviruses that infect cattle. The genetic characterization of R1ΔgC and R2ΔgI showed that they are composed of different sections of the parental genomes. The aim of this study was the characterization of the in vivo behavior of these recombinants in the natural host. RESULTS: Four groups of four 3-month-old calves of both genders were intranasally inoculated with either the recombinant or parental viruses. A control group of two animals was also included. Viral excretion and clinical signs were monitored after infection. Histopathological examination of the central nervous system (CNS) was performed and the establishment of latency in trigeminal ganglia was analyzed by PCR. The humoral response was also evaluated using ELISA tests. Three out of four animals from the BoHV-5 infected group excreted virus for 4-10 days. Two calves shed R1ΔgC virus for one day. In R2ΔgI and BoHV-1.2ΔgCΔgI groups, infectious virus was isolated only after two or three blind passages. None of the infected animals developed neurological signs, although those infected with BoHV-5 showed histopathological evidence of viral infection. Latent viral DNA was detected in at least one calf from each infected group. Serum and/or mucosal antibodies were detected in all groups. CONCLUSION: Both BoHV-1/-5 recombinants and the BoHV-1 parental strain are attenuated in calves, although they are able to replicate in animals at low rates and to establish latent infections.


Subject(s)
Cattle Diseases/virology , Encephalitis, Viral/veterinary , Herpesviridae Infections/veterinary , Herpesvirus 1, Bovine/genetics , Herpesvirus 5, Bovine/genetics , Meningoencephalitis/veterinary , Animals , Cattle , Cattle Diseases/immunology , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/pathogenicity , Herpesvirus 1, Bovine/physiology , Herpesvirus 5, Bovine/pathogenicity , Herpesvirus 5, Bovine/physiology , Immunity, Humoral/immunology , In Vitro Techniques , Male , Meningoencephalitis/immunology , Meningoencephalitis/virology , Recombination, Genetic/genetics , Trigeminal Ganglion/virology , Virus Latency/genetics , Virus Replication/genetics
15.
BMC Vet Res ; 7: 8, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21299866

ABSTRACT

BACKGROUND: Bovine herpesvirus 5 (BoHV-5) is a member of the subfamily Alphaherpesvirinae responsible for meningo-encephalitis in young cattle. The first case of bovine meningo-encephalitis associated with a herpesvirus infection was reported in Australia. The current geographical distribution of BoHV-5 infection is mainly restricted to South America, especially Brazil and Argentina. Outbreaks of BoHV-5 are regularly observed in Argentina suggesting the circulation of the virus in the bovine population. RESULTS: Seventeen field strains of BoHV-5 isolated from 1984 to now were confirmed by differential PCR and subjected to restriction endonuclease analysis (REA). Viral DNA was cleaved with BstEII which allows the differentiation among subtypes a, b and non a, non b. According to the REA with BstEII, only one field strain showed a pattern similar to the Argentinean A663 strain (prototype of BoHV-5b). All other isolates showed a clear pattern similar to the Australian N569 strain (prototype of BoHV-5a) consistent with the subtypes observed in Brazil, the other South-American country where BoHV-5 is known to be prevalent. The genomic region of subtype b responsible for the distinct pattern was determined and amplified by PCR; specifically a point mutation was identified in glycoprotein B gene, on the BstEII restriction site, which generates the profile specific of BoHV-5b. CONCLUSIONS: This is the first report of circulation of BoHV-5a in Argentina as the prevailing subtype. Therefore the circulation of BoHV-5b was restricted to a few years in Argentina, speculating that this subtype was not able to be maintained in the bovine population. The mutation in the gB gene is associated with the difference in the restriction patterns between subtypes "a" and "b".


Subject(s)
Cattle Diseases/virology , Disease Outbreaks/veterinary , Encephalitis/veterinary , Herpesviridae Infections/veterinary , Herpesvirus 5, Bovine/isolation & purification , Animals , Argentina/epidemiology , Cattle , Cattle Diseases/epidemiology , DNA, Viral/chemistry , DNA, Viral/genetics , Encephalitis/epidemiology , Encephalitis/virology , Herpesviridae Infections/epidemiology , Herpesviridae Infections/virology , Herpesvirus 5, Bovine/genetics , Point Mutation/genetics , Polymerase Chain Reaction/veterinary , Restriction Mapping/veterinary , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
16.
Microorganisms ; 9(6)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205549

ABSTRACT

During latency, herpesvirus infection results in the establishment of a dormant state in which a restricted set of viral genes are expressed. Together with alterations of the viral genome, several host genes undergo epigenetic silencing during latency. These epigenetic dysregulations of cellular genes might be involved in the development of cancer. In this context, Gallid alphaherpesvirus 2 (GaHV-2), causing Marek's disease (MD) in susceptible chicken, was shown to impair the expression of several cellular microRNAs (miRNAs). We decided to focus on gga-miR-126, a host miRNA considered a tumor suppressor through signaling pathways controlling cell proliferation. Our objectives were to analyze the cause and the impact of miR-126 silencing during GaHV-2 infection. This cellular miRNA was found to be repressed at crucial steps of the viral infection. In order to determine whether miR-126 low expression level was associated with specific epigenetic signatures, DNA methylation patterns were established in the miR-126 gene promoter. Repression was associated with hypermethylation at a CpG island located in the miR-126 host gene epidermal growth factor like-7 (EGFL-7). A strategy was developed to conditionally overexpress miR-126 and control miRNAs in transformed CD4+ T cells propagated from Marek's disease (MD) lymphoma. This functional assay showed that miR-126 restoration specifically diminishes cell proliferation. We identified CT10 regulator of kinase (CRK), an adaptor protein dysregulated in several human malignancies, as a candidate target gene. Indeed, CRK protein levels were markedly reduced by the miR-126 restoration.

17.
BMC Mol Biol ; 11: 67, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20813039

ABSTRACT

BACKGROUND: The c-myb proto-oncogene is the founding member of a family of transcription factors involved principally in haematopoiesis, in diverse organisms, from zebrafish to mammals. Its deregulation has been implicated in human leukaemogenesis and other cancers. The expression of c-myb is tightly regulated by post-transcriptional mechanisms involving microRNAs. MicroRNAs are small, highly conserved non-coding RNAs that inhibit translation and decrease mRNA stability by binding to regulatory motifs mostly located in the 3'UTR of target mRNAs conserved throughout evolution. MYB is an evolutionarily conserved miR-150 target experimentally validated in mice, humans and zebrafish. However, the functional miR-150 sites of humans and mice are orthologous, whereas that of zebrafish is different. RESULTS: We identified the avian mature miRNA-150-5P, Gallus gallus gga-miR-150 from chicken leukocyte small-RNA libraries and showed that, as expected, the gga-miR-150 sequence was highly conserved, including the seed region sequence present in the other miR-150 sequences listed in miRBase. Reporter assays showed that gga-miR-150 acted on the avian MYB 3'UTR and identified the avian MYB target site involved in gga-miR-150 binding. A comparative in silico analysis of the miR-150 target sites of MYB 3'UTRs from different species led to the identification of a single set of putative target sites in amphibians and zebrafish, whereas two sets of putative target sites were identified in chicken and mammals. However, only the target site present in the chicken MYB 3'UTR that was identical to that in zebrafish was functional, despite the additional presence of mammalian target sites in chicken. This specific miR-150 site usage was not cell-type specific and persisted when the chicken c-myb 3'UTR was used in the cell system to identify mammalian target sites, showing that this miR-150 target site usage was intrinsic to the chicken c-myb 3'UTR. CONCLUSION: Our study of the avian MYB/gga-miR-150 interaction shows a conservation of miR-150 target site functionality between chicken and zebrafish that does not extend to mammals.


Subject(s)
3' Untranslated Regions , Chickens/genetics , Genes, myb , MicroRNAs/genetics , Proto-Oncogene Proteins c-myb/genetics , Zebrafish/genetics , Animals , Base Sequence , Gene Expression Regulation , Genes, Reporter , Humans , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myb/metabolism
18.
J Gen Virol ; 91(Pt 11): 2723-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702654

ABSTRACT

Based on sequencing data, norovirus (NoV) recombinants have been described, but no experimental evidence of recombination in NoVs has been documented. Using the murine norovirus (MNV) model, we investigated the occurrence of genetic recombination between two co-infecting wild-type MNV isolates in RAW cells. The design of a PCR-based genotyping tool allowed accurate discrimination between the parental genomes and the detection of a viable recombinant MNV (Rec MNV) in the progeny viruses. Genetic analysis of Rec MNV identified a homologous-recombination event located at the ORF1-ORF2 overlap. Rec MNV exhibited distinct growth curves and produced smaller plaques than the wild-type MNV in RAW cells. Here, we demonstrate experimentally that MNV undergoes homologous recombination at the previously described recombination hot spot for NoVs, suggesting that the MNV model might be suitable for in vitro studies of NoV recombination. Moreover, the results show that exchange of genetic material between NoVs can generate viruses with distinct biological properties from the parental viruses.


Subject(s)
Norovirus/growth & development , Norovirus/genetics , RNA, Viral/genetics , Recombination, Genetic , Animals , Cell Line , Cluster Analysis , Genotype , Macrophages/virology , Mice , Microbial Viability , Molecular Sequence Data , Norovirus/pathogenicity , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Analysis, DNA , Sequence Homology , Viral Plaque Assay , Virulence
19.
J Virol ; 83(7): 3127-37, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19153224

ABSTRACT

Phylogenetic studies of the emergence and spread of natural recombinants in herpesviruses infecting humans and animals have been reported recently. However, despite an ever-increasing amount of evidence of recombination in herpesvirus history, the recombination process and the consequences on the genetic diversity of the progeny remain poorly characterized. We addressed this issue by using multiple single-nucleotide polymorphisms (SNPs) differentiating the two subtypes of an alphaherpesvirus, bovine herpesvirus 1 (BoHV-1). Analysis of a large sample of progeny virions obtained in a single growth cycle of coinfected BoHV-1 strains provided a prospective investigation of the recombination dynamics by using SNPs as recombination markers. We found that the simultaneous infection with two closely related herpesviruses results in a highly diversified recombination mosaic. From the analysis of multiple recombinants arising in the progeny, we provide the first evidence of genetic interference influencing the recombination process in herpesviruses. In addition, we report striking differences in the levels of recombination frequency observed along the BoHV-1 genome. With particular emphasis on the genetic structure of a progeny virus population rising in vitro, our data show to which extent recombination participates to the genetic diversification of herpesviruses.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 1, Bovine/growth & development , Herpesvirus 1, Bovine/genetics , Recombination, Genetic , Viral Interference , Animals , Cattle , Cell Line , DNA, Viral/chemistry , DNA, Viral/genetics , Genetic Markers , Genotype , Herpesvirus 1, Bovine/isolation & purification , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
20.
Arch Virol ; 155(11): 1823-37, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20680360

ABSTRACT

Mdv1-miR-M4 is one of 25 microRNAs (miRNAs) expressed by Marek's disease virus (MDV-1), an oncogenic alphaherpesvirus that induces fatal T-cell lymphoma in chickens. Mdv1-miR-M4 was shown to be the second functional viral ortholog of miR-155, a cellular miRNA that plays a crucial role in several physiological and pathological processes in lymphocyte biology. In this study, we investigated a panel of putative mdv1-miR-M4 targets involved in gene networks affecting both cellular and viral life cycles. Using luciferase reporter assays, we showed that mdv1-miR-M4-5P and miR-155 efficiently targeted a common set of 3' untranslated regions (3'UTR) of six cellular genes (GPM6B, RREB1, c-Myb, MAP3K7IP2, PU.1 and C/EBP). In addition, we also investigated the interactions between mdv1-miR-M4-5P and mdv1-miR-M43P and viral mRNAs encoding UL28 and UL32 in both reporter and western blot assays. Mdv1-miR-M4 specifically inhibited the translation of these two viral proteins, which are involved in the cleavage/packaging of herpesvirus DNA.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation/physiology , Mardivirus/genetics , Mardivirus/metabolism , MicroRNAs/metabolism , Animals , Base Sequence , Cell Line , Chickens , Leukocytes/metabolism , MicroRNAs/genetics
SELECTION OF CITATIONS
SEARCH DETAIL