Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yongxiu Yao is active.

Publication


Featured researches published by Yongxiu Yao.


PLOS Pathogens | 2011

Critical Role of the Virus-Encoded MicroRNA-155 Ortholog in the Induction of Marek's Disease Lymphomas

Yuguang Zhao; Hongtao Xu; Yongxiu Yao; Lorraine P. Smith; Lydia Kgosana; James Green; Lawrence Petherbridge; Susan J. Baigent; Venugopal Nair

Notwithstanding the well-characterised roles of a number of oncogenes in neoplastic transformation, microRNAs (miRNAs) are increasingly implicated in several human cancers. Discovery of miRNAs in several oncogenic herpesviruses such as KSHV has further highlighted the potential of virus-encoded miRNAs to contribute to their oncogenic capabilities. Nevertheless, despite the identification of several possible cancer-related genes as their targets, the direct in vivo role of virus-encoded miRNAs in neoplastic diseases such as those induced by KSHV is difficult to demonstrate in the absence of suitable models. However, excellent natural disease models of rapid-onset Mareks disease (MD) lymphomas in chickens allow examination of the oncogenic potential of virus-encoded miRNAs. Using viruses modified by reverse genetics of the infectious BAC clone of the oncogenic RB-1B strain of MDV, we show that the deletion of the six-miRNA cluster 1 from the viral genome abolished the oncogenicity of the virus. This loss of oncogenicity appeared to be primarily due to the single miRNA within the cluster, miR-M4, the ortholog of cellular miR-155, since its deletion or a 2-nucleotide mutation within its seed region was sufficient to inhibit the induction of lymphomas. The definitive role of this miR-155 ortholog in oncogenicity was further confirmed by the rescue of oncogenic phenotype by revertant viruses that expressed either the miR-M4 or the cellular homolog gga-miR-155. This is the first demonstration of the direct in vivo role of a virus-encoded miRNA in inducing tumors in a natural infection model. Furthermore, the use of viruses deleted in miRNAs as effective vaccines against virulent MDV challenge, enables the prospects of generating genetically defined attenuated vaccines.


Journal of Virology | 2009

A functional microRNA-155 ortholog encoded by the oncogenic Marek's disease virus.

Yuguang Zhao; Yongxiu Yao; Hongtao Xu; Luke S. Lambeth; Lorraine P. Smith; Lydia Kgosana; Xiaowei Wang; Venugopal Nair

ABSTRACT Kaposis sarcoma-associated herpesvirus-encoded microRNA (miRNA) MiR-K12-11 was recently shown to be a functional ortholog of miR-155, a miRNA that plays a major role in lymphoid malignancies and the modulation of immune responses. Here we show that miR-M4, encoded by the highly oncogenic Mareks disease virus of chickens, shares common targets with miR-155 and thus is also a functional ortholog of miR-155, the first one identified in an alphaherpesvirus. The observation that two distinct oncogenic herpesviruses associated with distinct types of lymphomas in different species encode functional miR-155 orthologs suggested the importance of this miRNA in regulatory pathways and the biology of lymphomagenesis.


Journal of Virology | 2007

Marek's Disease Virus Type 2 (MDV-2)-Encoded MicroRNAs Show No Sequence Conservation with Those Encoded by MDV-1

Yongxiu Yao; Yuguang Zhao; Hongtao Xu; Lorraine P. Smith; Charles H. Lawrie; Alain Sewer; Mihaela Zavolan; Venugopal Nair

ABSTRACT MicroRNAs (miRNAs) are increasingly being recognized as major regulators of gene expression in many organisms, including viruses. Among viruses, members of the family Herpesviridae account for the majority of the currently known virus-encoded miRNAs. The highly oncogenic Mareks disease virus type 1 (MDV-1), an avian herpesvirus, has recently been shown to encode eight miRNAs clustered in the MEQ and LAT regions of the viral genome. The genus Mardivirus, to which MDV-1 belongs, also includes the nononcogenic but antigenically related MDV-2. As MDV-1 and MDV-2 are evolutionarily very close, we sought to determine if MDV-2 also encodes miRNAs. For this, we cloned, sequenced, and analyzed a library of small RNAs from the lymphoblastoid cell line MSB-1, previously shown to be coinfected with both MDV-1 and MDV-2. Among the 5,099 small RNA sequences determined from the library, we identified 17 novel MDV-2-specific miRNAs. Out of these, 16 were clustered in a 4.2-kb long repeat region that encodes R-LORF2 to R-LORF5. The single miRNA outside the cluster was located in the short repeat region, within the C-terminal region of the ICP4 homolog. The expression of these miRNAs in MSB-1 cells and infected chicken embryo fibroblasts was further confirmed by Northern blotting analysis. The identification of miRNA clusters within the repeat regions of MDV-2 demonstrates conservation of the relative genomic positions of miRNA clusters in MDV-1 and MDV-2, despite the lack of sequence homology among the miRNAs of the two viruses. The identification of these novel miRNAs adds to the growing list of virus-encoded miRNAs.


Journal of General Virology | 2009

Differential expression of microRNAs in Marek's disease virus-transformed T-lymphoma cell lines.

Yongxiu Yao; Yuguang Zhao; Lorraine P. Smith; Charles H. Lawrie; Nigel J. Saunders; Michael Watson; Venugopal Nair

MicroRNAs (miRNAs) are increasingly recognized to play crucial roles in regulation of gene expression in different biological events, including many sporadic forms of cancer. However, despite the involvement of several viruses in inducing cancer, only a limited number of studies have been carried out to examine the miRNA expression signatures in virus-induced neoplasia, particularly in herpesvirus-induced tumours where virus-encoded miRNAs also contribute significantly to the miRNome of the tumour cell. Mareks disease (MD) is a naturally occurring, rapid-onset CD4+ T-cell lymphoma of poultry, induced by the highly contagious Mareks disease virus (MDV). High levels of expression of virus-encoded miRNAs and altered expression of several host-encoded miRNAs were demonstrated in the MDV-transformed lymphoblastoid cell line MSB-1. In order to identify the miRNA expression signature specific to MDV-transformed cells, we examined the global miRNA expression profiles in seven distinct MDV-transformed cell lines by microarray analysis. This study revealed that, in addition to the high levels of MDV-encoded miRNAs, these MD tumour-derived lymphoblastoid cell lines showed altered expression of several host-encoded miRNAs. Comparison of the miRNA expression profiles of these cell lines with the MDV-negative, retrovirus-transformed AVOL-1 cell line showed that miR-150 and miR-223 are downregulated irrespective of the viral aetiology, whereas downregulation of miR-155 was specific for MDV-transformed tumour cells. Thus, increased expression of MDV-encoded miRNAs with specific downregulation of miR-155 can be considered as unique expression signatures for MD tumour cells. Analysis of the functional targets of these miRNAs would contribute to the understanding of the molecular pathways of MD oncogenicity.


Journal of Virological Methods | 2008

Analysis of the expression profiles of Marek's disease virus-encoded microRNAs by real-time quantitative PCR.

Hongtao Xu; Yongxiu Yao; Yuguang Zhao; Lorraine P. Smith; Susan J. Baigent; Venugopal Nair

MicroRNAs (miRNAs) are a large class of endogenous approximately 22-nucleotide long non-coding RNAs involved in post-transcriptional silencing of gene expression by translational repression or direct cleavage of the target mRNAs. Several hundreds of miRNAs have now been identified in a wide range of organisms including many viruses. Mareks disease virus serotype 1 (MDV-1) is a highly contagious oncogenic herpesvirus that induces fatal T-cell lymphomas in chickens. MDV-1 has been shown recently to encode several miRNAs clustered in the Meq and LAT region of the viral genome. The majority of these miRNAs are expressed at very high levels in infected chicken embryo fibroblasts (CEF) as well as in MDV-transformed lymphomas and lymphoblastoid cell lines derived from tumours. As part of the studies to evaluate the functions of these miRNAs, a quantitative RT-PCR to analyse the expression of three MDV-encoded miRNAs, viz. miR-4, miR-8 and miR-12 in CEF infected with oncogenic strain RB-1B and vaccine strain CVI988 was developed. The study shows that these miRNAs are expressed at very high levels in the tumour tissues compared to the non-tumour tissues from the infected birds indicating that expression levels of MDV-1 miRNAs can be used as biomarkers for transformation.


Cancer Cell International | 2010

MicroRNA-26a-mediated regulation of interleukin-2 expression in transformed avian lymphocyte lines

Hongtao Xu; Yongxiu Yao; Lorraine P. Smith; Venugopal Nair

BackgroundMicro(mi)RNAs are a class of small non-coding RNAs that play critical roles in the induction of various cancers, including lymphomas induced by oncogenic viruses. While some of the miRNAs are oncogenic, miRNAs such as miR-26a are consistently downregulated in a number of cancers, demonstrating their potential tumor suppressor functions. Global miRNA expression profiles of a number of virus-transformed avian lymphoma cell lines have shown downregulation of gga-miR-26a expression, irrespective of molecular mechanisms of transformation or the viral aetiology. The neoplastic transformation of lymphocytes by many viruses accompanies high levels of proliferative responses, mostly mediated through cytokines such as IL-2. Chicken IL-2 can modulate T-cell proliferation and cytotoxicity in vitro and in vivo and dysregulation of IL-2 expression is observed in diseases such as leukaemia.ResultsThe expression levels of gga-miR-26a in chicken lymphoma cells transformed by 3 distinct avian oncogenic viruses, viz Mareks disease virus (MDV), avian leukosis virus (ALV) and Reticuloendotheliosis virus (REV) were consistently downregulated compared to the levels in the normal lymphocytes. This downregulation of miR-26a regardless of the viral etiology and molecular mechanisms of transformation was consistent with the tumor suppressor role of this miRNA. Notwithstanding this well-established role in cancer, we demonstrate the additional role of this miRNA in directly targeting chicken IL-2 through reporter and biochemical assays. The downregulation of miR-26a can relieve the suppressive effect of this miRNA on IL-2 expression.ConclusionsWe show that miR-26a is globally downregulated in a number of avian lymphoma cells irrespective of the mechanisms of transformation, reiterating the highly conserved tumor suppressor function of this miRNA. However, with the potential for directly targeting chicken IL-2, the downregulation of miR-26a in these tumor cells could relieve the inhibitory effect on IL-2 expression assisting in the proliferative features of the transformed lymphocyte lines.


Journal of Virology | 2014

An avian retrovirus uses canonical expression and processing mechanisms to generate viral microRNA

Yongxiu Yao; Lorraine P. Smith; Venugopal Nair; Mick Watson

ABSTRACT To date, the vast majority of known virus-encoded microRNAs (miRNAs) are derived from polymerase II transcripts encoded by DNA viruses. A recent demonstration that the bovine leukemia virus, a retrovirus, uses RNA polymerase III to directly transcribe the pre-miRNA hairpins to generate viral miRNAs further supports the common notion that the canonical pathway of miRNA biogenesis does not exist commonly among RNA viruses. Here, we show that an exogenous virus-specific region, termed the E element or XSR, of avian leukosis virus subgroup J (ALV-J), a member of avian retrovirus, encodes a novel miRNA, designated E (XSR) miRNA, using the canonical miRNA biogenesis pathway. Detection of novel microRNA species derived from the E (XSR) element, a 148-nucleotide noncoding RNA with hairpin structure, showed that the E (XSR) element has the potential to function as a microRNA primary transcript, demonstrating a hitherto unknown function with possible roles in myeloid leukosis associated with ALV-J.


Viruses | 2014

Role of Virus-Encoded microRNAs in Avian Viral Diseases

Yongxiu Yao; Venugopal Nair

With total dependence on the host cell, several viruses have adopted strategies to modulate the host cellular environment, including the modulation of microRNA (miRNA) pathway through virus-encoded miRNAs. Several avian viruses, mostly herpesviruses, have been shown to encode a number of novel miRNAs. These include the highly oncogenic Marek’s disease virus-1 (26 miRNAs), avirulent Marek’s disease virus-2 (36 miRNAs), herpesvirus of turkeys (28 miRNAs), infectious laryngotracheitis virus (10 miRNAs), duck enteritis virus (33 miRNAs) and avian leukosis virus (2 miRNAs). Despite the closer antigenic and phylogenetic relationship among some of the herpesviruses, miRNAs encoded by different viruses showed no sequence conservation, although locations of some of the miRNAs were conserved within the repeat regions of the genomes. However, some of the virus-encoded miRNAs showed significant sequence homology with host miRNAs demonstrating their ability to serve as functional orthologs. For example, mdv1-miR-M4-5p, a functional ortholog of gga-miR-155, is critical for the oncogenicity of Marek’s disease virus. Additionally, we also describe the potential association of the recently described avian leukosis virus subgroup J encoded E (XSR) miRNA in the induction of myeloid tumors in certain genetically-distinct chicken lines. In this review, we describe the advances in our understanding on the role of virus-encoded miRNAs in avian diseases.


Journal of General Virology | 2012

Novel microRNAs encoded by duck enteritis virus

Yongxiu Yao; Lorraine P. Smith; Lawrence Petherbridge; Mick Watson; Venugopal Nair

Duck enteritis virus (DEV) is an important herpesvirus pathogen associated with acute, highly contagious lethal disease in waterfowls. Using a deep sequencing approach on RNA from infected chicken embryo fibroblast cultures, we identified several novel DEV-encoded micro (mi)RNAs. Unlike most mardivirus-encoded miRNAs, DEV-encoded miRNAs mapped mostly to the unique long region of the genome. The precursors of DEV miR-D18 and miR-D19 overlapped with each other, suggesting similarities to miRNA-offset RNAs, although only the DEV-miR-D18-3p was functional in reporter assays. Identification of these novel miRNAs will add to the growing list of virus-encoded miRNAs enabling the exploration of their roles in pathogenesis.


Frontiers in Genetics | 2013

MicroRNA expression profiles in avian haemopoietic cells.

Yongxiu Yao; Jane Charlesworth; Venugopal Nair; Mick Watson

MicroRNAs (miRNAs) are small, abundant, non-coding RNAs that modulate gene expression by interfering with translation or stability of mRNA transcripts in a sequence-specific manner. A total of 734 precursor and 996 mature miRNAs have so far been identified in the chicken genome. A number of these miRNAs are expressed in a cell type-specific manner, and understanding their function requires detailed examination of their expression in different cell types. We carried out deep sequencing of small RNA populations isolated from stimulated or transformed avian haemopoietic cell lines to determine the changes in the expression profiles of these important regulatory molecules during these biological events. There were significant changes in the expression of a number of miRNAs, including miR-155, in chicken B cells stimulated with CD40 ligand. Similarly, avian leukosis virus (ALV)-transformed DT40 cells also showed changes in miRNA expression in relation to the naïve cells. Embryonic stem cell line BP25 demonstrated a distinct cluster of upregulated miRNAs, many of which were shown previously to be involved in embryonic stem cell development. Finally, chicken macrophage cell line HD11 showed changes in miRNA profiles, some of which are thought to be related to the transformation by v-myc transduced by the virus. This work represents the first publication of a catalog of microRNA expression in a range of important avian cells and provides insights into the potential roles of miRNAs in the hematopoietic lineages of cells in a model non-mammalian species.

Collaboration


Dive into the Yongxiu Yao's collaboration.

Top Co-Authors

Avatar

Venugopal Nair

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar

Lorraine P. Smith

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar

Yuguang Zhao

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Hongtao Xu

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Na Tang

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar

Susan J. Baigent

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar

Lydia Kgosana

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar

Mick Watson

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar

Yaoyao Zhang

Institute for Animal Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge