Network


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

Hotspot


Dive into the research topics where Xi-Juan Liu is active.

Publication


Featured researches published by Xi-Juan Liu.


Journal of Virology | 2015

MicroRNA miR-21 Attenuates Human Cytomegalovirus Replication in Neural Cells by Targeting Cdc25a

Ya-Ru Fu; Xi-Juan Liu; Xiao-Jun Li; Zhang-Zhou Shen; Bo Yang; Cong-Cong Wu; Jiafu Li; Ling-Feng Miao; Han-Qing Ye; Guan-Hua Qiao; Simon Rayner; Stéphane Chavanas; Christian Davrinche; William J. Britt; Qiyi Tang; Michael A. McVoy; Edward S. Mocarski; Min-Hua Luo

ABSTRACT Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, primarily manifesting as neurological disorders. HCMV infection alters expression of cellular microRNAs (miRs) and induces cell cycle arrest, which in turn modifies the cellular environment to favor virus replication. Previous observations found that HCMV infection reduces miR-21 expression in neural progenitor/stem cells (NPCs). Here, we show that infection of NPCs and U-251MG cells represses miR-21 while increasing the levels of Cdc25a, a cell cycle regulator and known target of miR-21. These opposing responses to infection prompted an investigation of the relationship between miR-21, Cdc25a, and viral replication. Overexpression of miR-21 in NPCs and U-251MG cells inhibited viral gene expression, genome replication, and production of infectious progeny, while shRNA-knockdown of miR-21 in U-251MG cells increased viral gene expression. In contrast, overexpression of Cdc25a in U-251MG cells increased viral gene expression and production of infectious progeny and overcame the inhibitory effects of miR-21 overexpression. Three viral gene products—IE1, pp71, and UL26—were shown to inhibit miR-21 expression at the transcriptional level. These results suggest that Cdc25a promotes HCMV replication and elevation of Cdc25a levels after HCMV infection are due in part to HCMV-mediated repression of miR-21. Thus, miR-21 is an intrinsic antiviral factor that is modulated by HCMV infection. This suggests a role for miR-21 downregulation in the neuropathogenesis of HCMV infection of the developing CNS. IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous pathogen and has very high prevalence among population, especially in China, and congenital HCMV infection is a major cause for birth defects. Elucidating virus-host interactions that govern HCMV replication in neuronal cells is critical to understanding the neuropathogenesis of birth defects resulting from congenital infection. In this study, we confirm that HCMV infection downregulates miR-21 but upregulates Cdc25a. Further determined the negative effects of cellular miRNA miR-21 on HCMV replication in neural progenitor/stem cells and U-251MG glioblastoma/astrocytoma cells. More importantly, our results provide the first evidence that miR-21 negatively regulates HCMV replication by targeting Cdc25a, a vital cell cycle regulator. We further found that viral gene products of IE1, pp71, and UL26 play roles in inhibiting miR-21 expression, which in turn causes increases in Cdc25a and benefits HCMV replication. Thus, miR-21 appears to be an intrinsic antiviral factor that represents a potential target for therapeutic intervention.


Journal of Virology | 2013

Later Passages of Neural Progenitor Cells from Neonatal Brain Are More Permissive for Human Cytomegalovirus Infection

Xing Pan; Xiao-Jun Li; Xi-Juan Liu; Hui Yuan; Jiafu Li; Ying-Liang Duan; Han-Qing Ye; Ya-Ru Fu; Guan-Hua Qiao; Cong-Cong Wu; Bo Yang; Xiaohui Tian; Kanghong Hu; Ling-Feng Miao; Xiao-Ling Chen; Jun Zheng; Simon Rayner; Philip H. Schwartz; William J. Britt; Jiang Xu; Min-Hua Luo

ABSTRACT Congenital human cytomegalovirus (HCMV) infection is the most frequent infectious cause of birth defects, primarily neurological disorders. Neural progenitor/stem cells (NPCs) are the major cell type in the subventricular zone and are susceptible to HCMV infection. In culture, the differentiation status of NPCs may change with passage, which in turn may alter susceptibility to virus infection. Previously, only early-passage (i.e., prior to passage 9) NPCs were studied and shown to be permissive to HCMV infection. In this study, NPC cultures derived at different gestational ages were evaluated after short (passages 3 to 6) and extended (passages 11 to 20) in vitro passages for biological and virological parameters (i.e., cell morphology, expression of NPC markers and HCMV receptors, viral entry efficiency, viral gene expression, virus-induced cytopathic effect, and release of infectious progeny). These parameters were not significantly influenced by the gestational age of the source tissues. However, extended-passage cultures showed evidence of initiation of differentiation, increased viral entry, and more efficient production of infectious progeny. These results confirm that NPCs are fully permissive for HCMV infection and that extended-passage NPCs initiate differentiation and are more permissive for HCMV infection. Later-passage NPCs being differentiated and more permissive for HCMV infection suggest that HCMV infection in fetal brain may cause more neural cell loss and give rise to severe neurological disabilities with advancing brain development.


Journal of Virology | 2013

Human Astrocytic Cells Support Persistent Coxsackievirus B3 Infection

Xiaowei Zhang; Zhenhua Zheng; Bo Shu; Xi-Juan Liu; Zhenfeng Zhang; Yan Liu; Bingke Bai; Qinxue Hu; Panyong Mao; Hanzhong Wang

ABSTRACT Enteroviruses can frequently target the human central nervous system to induce a variety of neurological diseases. Although enteroviruses are highly cytolytic, emerging evidence has shown that these viruses can establish persistent infections both in vivo and in vitro. Here, we investigated the susceptibility of three human brain cell lines, CCF-STTG1, T98G, and SK-N-SH, to infection with three enterovirus serotypes: coxsackievirus B3 (CVB3), enterovirus 71, and coxsackievirus A9. Persistent infection was observed in CVB3-infected CCF-STTG1 cells, as evidenced by prolonged detection of infectious virions, viral RNA, and viral antigens. Of note, infected CCF-STTG1 cells expressed the nonfunctional canonical viral receptors coxsackievirus-adenovirus receptor and decay-accelerating factor, while removal of cell surface chondroitin sulfate from CCF-STTG1 cells inhibited the replication of CVB3, suggesting that receptor usage was one of the major limiting factors in CVB3 persistence. In addition, CVB3 curtailed the induction of beta interferon in infected CCF-STTG1 cells, which likely contributed to the initiation of persistence. Furthermore, proinflammatory chemokines and cytokines, such as vascular cell adhesion molecule 1, interleukin-8 (IL-8), and IL-6, were upregulated in CVB3-infected CCF-STTG1 cells and human progenitor-derived astrocytes. Our data together demonstrate the potential of CCF-STTG1 cells to be a novel cell model for studying CVB3-central nervous system interactions, providing the basis toward a better understanding of CVB3-induced chronic neuropathogenesis.


Journal of Virology | 2015

Human Cytomegalovirus Infection Dysregulates the Localization and Stability of NICD1 and Jag1 in Neural Progenitor Cells

Xiao-Jun Li; Xi-Juan Liu; Bo Yang; Ya-Ru Fu; Fei Zhao; Zhang-Zhou Shen; Ling-Feng Miao; Simon Rayner; Stéphane Chavanas; Hua Zhu; William J. Britt; Qiyi Tang; Michael A. McVoy; Min-Hua Luo

ABSTRACT Human cytomegalovirus (HCMV) infection of the developing fetus frequently results in major neural developmental damage. In previous studies, HCMV was shown to downregulate neural progenitor/stem cell (NPC) markers and induce abnormal differentiation. As Notch signaling plays a vital role in the maintenance of stem cell status and is a switch that governs NPC differentiation, the effect of HCMV infection on the Notch signaling pathway in NPCs was investigated. HCMV downregulated mRNA levels of Notch1 and its ligand, Jag1, and reduced protein levels and altered the intracellular localization of Jag1 and the intracellular effector form of Notch1, NICD1. These effects required HCMV gene expression and appeared to be mediated through enhanced proteasomal degradation. Transient expression of the viral tegument proteins of pp71 and UL26 reduced NICD1 and Jag1 protein levels endogenously and exogenously. Given the critical role of Notch signaling in NPC growth and differentiation, these findings reveal important mechanisms by which HCMV disturbs neural cell development in vitro. Similar events in vivo may be associated with HCMV-mediated neuropathogenesis during congenital infection in the fetal brain. IMPORTANCE Congenital human cytomegalovirus (HCMV) infection is the leading cause of birth defects that primarily manifest as neurological disabilities. Neural progenitor cells (NPCs), key players in fetal brain development, are the most susceptible cell type for HCMV infection in the fetal brain. Studies have shown that NPCs are fully permissive for HCMV infection, which causes neural cell loss and premature differentiation, thereby perturbing NPC fate. Elucidation of virus-host interactions that govern NPC proliferation and differentiation is critical to understanding neuropathogenesis. The Notch signaling pathway is critical for maintaining stem cell status and functions as a switch for differentiation of NPCs. Our investigation into the impact of HCMV infection on this pathway revealed that HCMV dysregulates Notch signaling by altering expression of the Notch ligand Jag1, Notch1, and its active effector in NPCs. These results suggest a mechanism for the neuropathogenesis induced by HCMV infection that includes altered NPC differentiation and proliferation.


Journal of Virology | 2017

ORF7 of Varicella-Zoster Virus Is Required for Viral Cytoplasmic Envelopment in Differentiated Neuronal Cells

Hai-Fei Jiang; Wei Wang; Xuan Jiang; Wen-Bo Zeng; Zhang-Zhou Shen; Yi-Ge Song; Hong Yang; Xi-Juan Liu; Xiao Dong; Jing Zhou; Jin-Yan Sun; Fei-Long Yu; Lin Guo; Tong Cheng; Simon Rayner; Fei Zhao; Hua Zhu; Min-Hua Luo

ABSTRACT Although a varicella-zoster virus (VZV) vaccine has been used for many years, the neuropathy caused by VZV infection is still a major health concern. Open reading frame 7 (ORF7) of VZV has been recognized as a neurotropic gene in vivo, but its neurovirulent role remains unclear. In the present study, we investigated the effect of ORF7 deletion on VZV replication cycle at virus entry, genome replication, gene expression, capsid assembly and cytoplasmic envelopment, and transcellular transmission in differentiated neural progenitor cells (dNPCs) and neuroblastoma SH-SY5Y (dSY5Y) cells. Our results demonstrate that the ORF7 protein is a component of the tegument layer of VZV virions. Deleting ORF7 did not affect viral entry, viral genome replication, or the expression of typical viral genes but clearly impacted cytoplasmic envelopment of VZV capsids, resulting in a dramatic increase of envelope-defective particles and a decrease in intact virions. The defect was more severe in differentiated neuronal cells of dNPCs and dSY5Y. ORF7 deletion also impaired transmission of ORF7-deficient virus among the neuronal cells. These results indicate that ORF7 is required for cytoplasmic envelopment of VZV capsids, virus transmission among neuronal cells, and probably the neuropathy induced by VZV infection. IMPORTANCE The neurological damage caused by varicella-zoster virus (VZV) reactivation is commonly manifested as clinical problems. Thus, identifying viral neurovirulent genes and characterizing their functions are important for relieving VZV related neurological complications. ORF7 has been previously identified as a potential neurotropic gene, but its involvement in VZV replication is unclear. In this study, we found that ORF7 is required for VZV cytoplasmic envelopment in differentiated neuronal cells, and the envelopment deficiency caused by ORF7 deletion results in poor dissemination of VZV among neuronal cells. These findings imply that ORF7 plays a role in neuropathy, highlighting a potential strategy to develop a neurovirulence-attenuated vaccine against chickenpox and herpes zoster and providing a new target for intervention of neuropathy induced by VZV.


PLOS Pathogens | 2017

Human cytomegalovirus IE1 downregulates Hes1 in neural progenitor cells as a potential E3 ubiquitin ligase

Xi-Juan Liu; Bo Yang; Sheng-Nan Huang; Cong-Cong Wu; Xiao-Jun Li; Shuang Cheng; Xuan Jiang; Fei Hu; Ying-Zi Ming; Michael Nevels; William J. Britt; Simon Rayner; Qiyi Tang; Wen-Bo Zeng; Fei Zhao; Min-Hua Luo

Congenital human cytomegalovirus (HCMV) infection is the leading cause of neurological disabilities in children worldwide, but the mechanisms underlying these disorders are far from well-defined. HCMV infection has been shown to dysregulate the Notch signaling pathway in human neural progenitor cells (NPCs). As an important downstream effector of Notch signaling, the transcriptional regulator Hairy and Enhancer of Split 1 (Hes1) is essential for governing NPC fate and fetal brain development. In the present study, we report that HCMV infection downregulates Hes1 protein levels in infected NPCs. The HCMV 72-kDa immediate-early 1 protein (IE1) is involved in Hes1 degradation by assembling a ubiquitination complex and promoting Hes1 ubiquitination as a potential E3 ubiquitin ligase, followed by proteasomal degradation of Hes1. Sp100A, an important component of PML nuclear bodies, is identified to be another target of IE1-mediated ubiquitination. A C-terminal acidic region in IE1, spanning amino acids 451 to 475, is required for IE1/Hes1 physical interaction and IE1-mediated Hes1 ubiquitination, but is dispensable for IE1/Sp100A interaction and ubiquitination. Our study suggests a novel mechanism linking downregulation of Hes1 protein to neurodevelopmental disorders caused by HCMV infection. Our findings also complement the current knowledge of herpesviruses by identifying IE1 as the first potential HCMV-encoded E3 ubiquitin ligase.


Journal of Virology | 2018

WDR5 Facilitates Human Cytomegalovirus Replication by Promoting Capsid Nuclear Egress

Bo Yang; Xi-Juan Liu; Yongxuan Yao; Xuan Jiang; Xian-Zhang Wang; Hong Yang; Jin-Yan Sun; Yun Miao; Wei Wang; Zhen-Li Huang; Yanyi Wang; Qiyi Tang; Simon Rayner; William J. Britt; Michael A. McVoy; Min-Hua Luo; Fei Zhao

ABSTRACT WD repeat-containing protein 5 (WDR5) is essential for assembling the VISA-associated complex to induce a type I interferon antiviral response to Sendai virus infection. However, the roles of WDR5 in DNA virus infections are not well described. Here, we report that human cytomegalovirus exploits WDR5 to facilitate capsid nuclear egress. Overexpression of WDR5 in fibroblasts slightly enhanced the infectious virus yield. However, WDR5 knockdown dramatically reduced infectious virus titers with only a small decrease in viral genome replication or gene expression. Further investigation of late steps of viral replication found that WDR5 knockdown significantly impaired formation of the viral nuclear egress complex and induced substantially fewer infoldings of the inner nuclear membrane. In addition, fewer capsids were associated with these infoldings, and there were fewer capsids in the cytoplasm. Restoration of WDR5 partially reversed these effects. These results suggest that WDR5 knockdown impairs the nuclear egress of capsids, which in turn decreases virus titers. These findings reveal an important role for a host factor whose function(s) is usurped by a viral pathogen to promote efficient replication. Thus, WDR5 represents an interesting regulatory mechanism and a potential antiviral target. IMPORTANCE Human cytomegalovirus (HCMV) has a large (∼235-kb) genome with over 170 open reading frames and exploits numerous cellular factors to facilitate its replication. HCMV infection increases protein levels of WD repeat-containing protein 5 (WDR5) during infection, overexpression of WDR5 enhances viral replication, and knockdown of WDR5 dramatically attenuates viral replication. Our results indicate that WDR5 promotes the nuclear egress of viral capsids, the depletion of WDR5 resulting in a significant decrease in production of infectious virions. This is the first report that WDR5 favors HCMV, a DNA virus, replication and highlights a novel target for antiviral therapy.


Virologica Sinica | 2017

Human cytomegalovirus infection dysregulates neural progenitor cell fate by disrupting Hes1 rhythm and down-regulating its expression

Xi-Juan Liu; Xuan Jiang; Sheng-Nan Huang; Jin-Yan Sun; Fei Zhao; Wen-Bo Zeng; Min-Hua Luo

Human cytomegalovirus (HCMV) infection is a leading cause of birth defects, primarily affecting the central nervous system and causing its maldevelopment. As the essential downstream effector of Notch signaling pathway, Hes1, and its dynamic expression, plays an essential role on maintaining neural progenitor /stem cells (NPCs) cell fate and fetal brain development. In the present study, we reported the first observation of Hes1 oscillatory expression in human NPCs, with an approximately 1.5 hour periodicity and a Hes1 protein half-life of about 17 (17.6 ± 0.2) minutes. HCMV infection disrupts the Hes1 rhythm and down-regulates its expression. Furthermore, we discovered that depleting Hes1 protein disturbed NPCs cell fate by suppressing NPCs proliferation and neurosphere formation, and driving NPCs abnormal differentiation. These results suggested a novel mechanism linking disruption of Hes1 rhythm and down-regulation of Hes1 expression to neurodevelopmental disorders caused by congenital HCMV infection.


Biologia Plantarum | 2018

Differences in responses of moderately salt-tolerant and salt-sensitive tree species to heterogeneous salinity

Xiaohui Feng; Ping An; Kai Guo; Xianglong Li; Xi-Juan Liu

Growth responses of the moderately salt-tolerant velvet ash (Fraxinus velutina) and salt-sensitive poplar (Populus × euramericana) were investigated under heterogeneous root zone salinity. The salinity treatments imposed on the two root zones (lower-higher) were 137-137 (uniform), 103-171, 68-205, 34-239, and 0-273 mM NaCl for velvet ash, and 51-51 (uniform), 34-68, 17-85, and 0-103 mM NaCl for poplar. The leaf gas exchange of the plants was measured one month after these treatments were implemented, and the plants were sampled 75 d after treatment to measure other physiological parameters. Net photosynthetic rate, transpiration rate, total biomass, and fine root compensatory growth increased as the difference in salinity between the two root zones (i.e., salinity heterogeneity) increased in velvet ash. These parameters showed no significant difference among the treatments in poplar. The leaf Na+ content was lower under heterogeneous salinity than under uniform salinity in both tested species. The leaf proline content in velvet ash decreased under heterogeneous salinity compared to that under uniform salinity, whereas that of poplar increased. The soluble sugar content of velvet ash leaves increased under heterogeneous salinity, whereas no changes were observed in poplar. The increased fine root biomass in the lower salinity zone promoted velvet ash growth by decreasing the leaf Na+ and Cl- content under heterogeneous salinity. The poplar’s undifferentiated root distribution and gas exchange in response to the heterogeneous salinity were attributed to its salt sensitivity.


Virology | 2017

Infected T98G glioblastoma cells support human cytomegalovirus reactivation from latency

Shuang Cheng; Xuan Jiang; Bo Yang; Le Wen; Fei Zhao; Wen-Bo Zeng; Xi-Juan Liu; Xiao Dong; Jin-Yan Sun; Ying-Zi Ming; Hua Zhu; Simon Rayner; Qiyi Tang; Elizabeth A. Fortunato; Min-Hua Luo

T98G cells have been shown to support long-term human cytomegalovirus (HCMV) genome maintenance without infectious virus release. However, it remains unclear whether these viral genomes could be reactivated. To address this question, a recombinant HCMV (rHCMV) containing a GFP gene was used to infect T98G cells, and the infected cells absent of infectious virus production were designated T98G-LrV. Upon dibutyryl cAMP plus IBMX (cAMP/IBMX) treatment, a serial of phenomena were observed, including GFP signal increase, viral genome replication, lytic genes expression and infectious viruses release, indicating the reactivation of HCMV in T98G-LrV cells from a latent status. Mechanistically, HCMV reactivation in the T98G-LrV cells induced by cAMP/IBMX was associated with the PKA-CREB signaling pathway. These results demonstrate that HCMV was latent in T98G-LrV cells and could be reactivated. The T98G-LrV cells represent an effective model for investigating the mechanisms of HCMV reactivation from latency in the context of neural cells.

Collaboration


Dive into the Xi-Juan Liu's collaboration.

Top Co-Authors

Avatar

Min-Hua Luo

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Bo Yang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Simon Rayner

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

William J. Britt

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Fei Zhao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Xiao-Jun Li

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Xuan Jiang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Cong-Cong Wu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Qiyi Tang

Ponce Health Sciences University

View shared research outputs
Top Co-Authors

Avatar

Han-Qing Ye

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge