Network


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

Hotspot


Dive into the research topics where Wenshi Wang is active.

Publication


Featured researches published by Wenshi Wang.


The Journal of Infectious Diseases | 2017

Hepatitis E virus infects neurons and brains.

Xinying Zhou; Fen Huang; Lei Xu; Zhanmin Lin; Femke M.S. de Vrij; Ane C. Ayo-Martin; Mark van der Kroeg; Manzhi Zhao; Yuebang Yin; Wenshi Wang; Wanlu Cao; Yijin Wang; Steven A. Kushner; Jean Marie Peron; Laurent Alric; Robert A. de Man; Bart C. Jacobs; Jeroen J.J. van Eijk; Eleonora Aronica; Dave Sprengers; Herold J. Metselaar; Chris I. De Zeeuw; Harry R. Dalton; Nassim Kamar; Maikel P. Peppelenbosch; Qiuwei Pan

Hepatitis E virus (HEV), as a hepatotropic virus, is supposed to exclusively infect the liver and only cause hepatitis. However, a broad range of extrahepatic manifestations (in particular, idiopathic neurological disorders) have been recently reported in association with its infection. In this study, we have demonstrated that various human neural cell lines (embryonic stem cell-derived neural lineage cells) induced pluripotent stem cell-derived human neurons and primary mouse neurons are highly susceptible to HEV infection. Treatment with interferon-α or ribavirin, the off-label antiviral drugs for chronic hepatitis E, exerted potent antiviral activities against HEV infection in neural cells. More importantly, in mice and monkey peripherally inoculated with HEV particles, viral RNA and protein were detected in brain tissues. Finally, patients with HEV-associated neurological disorders shed the virus into cerebrospinal fluid, indicating a direct infection of their nervous system. Thus, HEV is neurotropic in vitro, and in mice, monkeys, and possibly humans. These results challenge the dogma of HEV as a pure hepatotropic virus and suggest that HEV infection should be considered in the differential diagnosis of idiopathic neurological disorders.


The FASEB Journal | 2016

IFN regulatory factor 1 restricts hepatitis E virus replication by activating STAT1 to induce antiviral IFN-stimulated genes

Lei Xu; Xinying Zhou; Wenshi Wang; Yijin Wang; Yuebang Yin; Luc J. W. van der Laan; Dave Sprengers; Herold J. Metselaar; Maikel P. Peppelenbosch; Qiuwei Pan

IFN regulatory factor 1 (IRF1) is one of the most important IFN‐stimulated genes (ISGs) in cellular antiviral immunity. Although hepatitis E virus (HEV) is a leading cause of acute hepatitis worldwide, how ISGs counteract HEV infection is largely unknown. This study was conducted to investigate the effect of IRF1 on HEV replication. Multiple cell lines were used in 2 models that harbor HEV. In different HEV cell culture systems, IRF1 effectively inhibited HEV replication. IRF1 did not trigger IFN production, and chromatin immunoprecipitation sequencing data analysis revealed that IRF1 bound to the promoter region of signal transducers and activators of transcription 1 (STAT1). Functional assay confirmed that IRF1 could drive the transcription of STAT1, resulting in elevation of total and phosphorylated STAT1 proteins and further activating the transcription of a panel of downstream antiviral ISGs. By pharmacological inhibitors and RNAi‐mediated gene‐silencing approaches, we revealed that antiviral function of IRF1 is dependent on the JAK‐STAT cascade. Furthermore, induction of ISGs and the anti‐HEV effect of IRF1 overlapped that of IFNα, but was potentiated by ribavirin. We demonstrated that IRF1 effectively inhibits HEV replication through the activation of the JAK‐STAT pathway, and the subsequent transcription of antiviral ISGs, but independent of IFN production.—Xu, L., Zhou, X., Wang, W., Wang, Y., Yin, Y., van der Laan, L. J. W., Sprengers, D., Metselaar, H. J., Peppelenbosch, M. P., Pan, Q. IFN regulatory factor 1 restricts hepatitis E virus replication by activating STAT1 to induce antiviral IFN‐stimulated genes. FASEB J. 30, 3352–3367 (2016). www.fasebj.org


Scientific Reports | 2016

Convergent Transcription of Interferon-stimulated Genes by TNF-α and IFN-α Augments Antiviral Activity against HCV and HEV

Wenshi Wang; Lei Xu; Johannes H. Brandsma; Yijin Wang; Mohamad S. Hakim; Xinying Zhou; Yuebang Yin; Gwenny M. Fuhler; Luc J. W. van der Laan; C. Janneke van der Woude; Dave Sprengers; Herold J. Metselaar; Ron Smits; Raymond A. Poot; Maikel P. Peppelenbosch; Qiuwei Pan

IFN-α has been used for decades to treat chronic hepatitis B and C, and as an off-label treatment for some cases of hepatitis E virus (HEV) infection. TNF-α is another important cytokine involved in inflammatory disease, which can interact with interferon signaling. Because interferon-stimulated genes (ISGs) are the ultimate antiviral effectors of the interferon signaling, this study aimed to understand the regulation of ISG transcription and the antiviral activity by IFN-α and TNF-α. In this study, treatment of TNF-α inhibited replication of HCV by 71 ± 2.4% and HEV by 41 ± 4.9%. Interestingly, TNF-α induced the expression of a panel of antiviral ISGs (2-11 fold). Blocking the TNF-α signaling by Humira abrogated ISG induction and its antiviral activity. Chip-seq data analysis and mutagenesis assay further revealed that the NF-κB protein complex, a key downstream element of TNF-α signaling, directly binds to the ISRE motif in the ISG promoters and thereby drives their transcription. This process is independent of interferons and JAK-STAT cascade. Importantly, when combined with IFN-α, TNF-α works cooperatively on ISG induction, explaining their additive antiviral effects. Thus, our study reveals a novel mechanism of convergent transcription of ISGs by TNF-α and IFN-α, which augments their antiviral activity against HCV and HEV.


Liver International | 2017

The global burden of hepatitis E outbreaks: a systematic review.

Mohamad S. Hakim; Wenshi Wang; Wichor M. Bramer; Jiawei Geng; Fen Huang; Robert A. de Man; Maikel P. Peppelenbosch; Qiuwei Pan

Hepatitis E virus (HEV) is responsible for repeated water‐borne outbreaks since the past century, representing an emerging issue in public health. However, the global burden of HEV outbreak has not been comprehensively described. We performed a systematic review of confirmed HEV outbreaks based on published literatures. HEV outbreaks have mainly been reported from Asian and African countries, and only a few from European and American countries. India represents a country with the highest number of reported HEV outbreaks. HEV genotypes 1 and 2 were responsible for most of the large outbreaks in developing countries. During the outbreaks in developing countries, a significantly higher case fatality rate was observed in pregnant women. In fact, outbreaks have occurred both in open and closed populations. The control measures mainly depend upon improvement of sanitation and hygiene. This study highlights that HEV outbreak is not new, yet it is a continuous global health problem.


Trends in Microbiology | 2017

Transcriptional Regulation of Antiviral Interferon-Stimulated Genes

Wenshi Wang; Lei Xu; Junhong Su; Maikel P. Peppelenbosch; Qiuwei Pan

Interferon-stimulated genes (ISGs) are a group of gene products that coordinately combat pathogen invasions, in particular viral infections. Transcription of ISGs occurs rapidly upon pathogen invasion, and this is classically provoked via activation of the Janus kinase/signal transducer and activator of transcription (JAK–STAT) pathway, mainly by interferons (IFNs). However, a plethora of recent studies have reported a variety of non-canonical mechanisms regulating ISG transcription. These new studies are extremely important for understanding the quantitative and temporal differences in ISG transcription under specific circumstances. Because these canonical and non-canonical regulatory mechanisms are essential for defining the nature of host defense and associated detrimental proinflammatory effects, we comprehensively review the state of this rapidly evolving field and the clinical implications of recently acquired knowledge in this respect.


Hepatology | 2017

RIG‐I is a key antiviral interferon‐stimulated gene against hepatitis E virus regardless of interferon production

Lei Xu; Wenshi Wang; Yunlong Li; Xinying Zhou; Yuebang Yin; Yijin Wang; Robert A. de Man; Luc J. W. van der Laan; Fen Huang; Nassim Kamar; Maikel P. Peppelenbosch; Qiuwei Pan

Interferons (IFNs) are broad antiviral cytokines that exert their function by inducing the transcription of hundreds of IFN‐stimulated genes (ISGs). However, little is known about the antiviral potential of these cellular effectors on hepatitis E virus (HEV) infection, the leading cause of acute hepatitis globally. In this study, we profiled the antiviral potential of a panel of important human ISGs on HEV replication in cell culture models by overexpression of an individual ISG. The mechanism of action of the key anti‐HEV ISG was further studied. We identified retinoic acid–inducible gene I (RIG‐I), melanoma differentiation–associated protein 5, and IFN regulatory factor 1 (IRF1) as the key anti‐HEV ISGs. We found that basal expression of RIG‐I restricts HEV infection. Pharmacological activation of the RIG‐I pathway by its natural ligand 5′‐triphosphate RNA potently inhibits HEV replication. Overexpression of RIG‐I activates the transcription of a wide range of ISGs. RIG‐I also mediates but does not overlap with IFN‐α‐initiated ISG transcription. Although it is classically recognized that RIG‐I exerts antiviral activity through the induction of IFN production by IRF3 and IRF7, we reveal an IFN‐independent antiviral mechanism of RIG‐I in combating HEV infection. We found that activation of RIG‐I stimulates an antiviral response independent of IRF3 and IRF7 and regardless of IFN production. However, it is partially through activation of the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) cascade of IFN signaling. RIG‐I activated two distinct categories of ISGs, one JAK‐STAT‐dependent and the other JAK‐STAT‐independent, which coordinately contribute to the anti‐HEV activity. Conclusion: We identified RIG‐I as an important anti‐HEV ISG that can be pharmacologically activated; activation of RIG‐I stimulates the cellular innate immunity against HEV regardless of IFN production but partially through the JAK‐STAT cascade of IFN signaling. (Hepatology 2017;65:1823‐1839).


Journal of Viral Hepatitis | 2016

Disparity of basal and therapeutically activated interferon signalling in constraining hepatitis E virus infection

Xinying Zhou; Lei Xu; Wenshi Wang; Koichi Watashi; Yijin Wang; Dave Sprengers; Petra de Ruiter; Luc J. W. van der Laan; Herold J. Metselaar; Nassim Kamar; Maikel P. Peppelenbosch; Qiuwei Pan

Hepatitis E virus (HEV) represents one of the foremost causes of acute hepatitis globally. Although there is no proven medication for hepatitis E, pegylated interferon‐α (IFN‐α) has been used as off‐label drug for treating HEV. However, the efficacy and molecular mechanisms of how IFN signalling interacts with HEV remain undefined. As IFN‐α has been approved for treating chronic hepatitis C (HCV) for decades and the role of interferon signalling has been well studied in HCV infection, this study aimed to comprehensively investigate virus–host interactions in HEV infection with focusing on the IFN signalling, in comparison with HCV infection. A comprehensive screen of human cytokines and chemokines revealed that IFN‐α was the sole humoral factor inhibiting HEV replication. IFN‐α treatment exerted a rapid and potent antiviral activity against HCV, whereas it had moderate and delayed anti‐HEV effects in vitro and in patients. Surprisingly, blocking the basal IFN pathway by inhibiting JAK1 to phosphorylate STAT1 has resulted in drastic facilitation of HEV, but not HCV infection. Gene silencing of the key components of JAK‐STAT cascade of the IFN signalling, including JAK1, STAT1 and interferon regulatory factor 9 (IRF9), stimulated HEV infection. In conclusion, compared to HCV, HEV is less sensitive to IFN treatment. In contrast, the basal IFN cascade could effectively restrict HEV infection. This bears significant implications in management of HEV patients and future therapeutic development.


Science Signaling | 2017

Unphosphorylated ISGF3 drives constitutive expression of interferon-stimulated genes to protect against viral infections

Wenshi Wang; Yuebang Yin; Lei Xu; Junhong Su; Fen Huang; Yijin Wang; Patrick P. C. Boor; Kan Chen; Wenhui Wang; Wanlu Cao; Xinying Zhou; Pengyu Liu; Luc J. W. van der Laan; Jaap Kwekkeboom; Maikel P. Peppelenbosch; Qiuwei Pan

A tripartite transcription factor complex mediates an interferon-independent antiviral response. An interferon-independent antiviral defense Virally infected cells produce type I interferons (IFNs), which stimulate the phosphorylation and activation of the STAT1 and STAT2 transcription factors. When combined with the transcriptional regulator IRF9, phosphorylated STAT1 and STAT2 form the ISGF3 complex, which drives the expression of IFN-stimulated genes (ISGs) that are important for antiviral immunity. Wang et al. report the formation of an alternative form of ISGF3, U-ISGF3, which drove the expression of ISGs and protected cells from viral infection in the absence of detection of detectable IFN or IFN signaling. In addition to IRF9, U-ISGF3 contained unphosphorylated STAT1 and STAT2. Together, these data suggest that U-ISGF3 drives constitutive expression of ISGs as part of an IFN-independent, antiviral immune response. Interferon (IFN)–stimulated genes (ISGs) are antiviral effectors that are induced by IFNs through the formation of a tripartite transcription factor ISGF3, which is composed of IRF9 and phosphorylated forms of STAT1 and STAT2. However, we found that IFN-independent ISG expression was detectable in immortalized cell lines, primary intestinal and liver organoids, and liver tissues. The constitutive expression of ISGs was mediated by the unphosphorylated ISGF3 (U-ISGF3) complex, consisting of IRF9 together with unphosphorylated STAT1 and STAT2. Under homeostatic conditions, STAT1, STAT2, and IRF9 were found in the nucleus. Analysis of a chromatin immunoprecipitation sequencing data set revealed that STAT1 specifically bound to the promoters of ISGs even in the absence of IFNs. Knockdown of STAT1, STAT2, or IRF9 by RNA interference led to the decreased expression of various ISGs in Huh7.5 human liver cells, which was confirmed in mouse embryonic fibroblasts (MEFs) from STAT1−/−, STAT2−/−, or IRF9−/− mice. Furthermore, decreased ISG expression was accompanied by increased replication of hepatitis C virus and hepatitis E virus. Conversely, simultaneous overexpression of all ISGF3 components, but not any single factor, induced the expression of ISGs and inhibited viral replication; however, no phosphorylated STAT1 and STAT2 were detected. A phosphorylation-deficient STAT1 mutant was comparable to the wild-type protein in mediating the IFN-independent expression of ISGs and antiviral activity, suggesting that ISGF3 works in a phosphorylation-independent manner. These data suggest that the U-ISGF3 complex is both necessary and sufficient for constitutive ISG expression and antiviral immunity under homeostatic conditions.


Antimicrobial Agents and Chemotherapy | 2016

Cross Talk between Nucleotide Synthesis Pathways with Cellular Immunity in Constraining Hepatitis E Virus Replication

Yijin Wang; Wenshi Wang; Lei Xu; Xinying Zhou; Ehsan Shokrollahi; Krzysztof Felczak; Luc J. W. van der Laan; Krzysztof W. Pankiewicz; Dave Sprengers; Nicolaas J. H. Raat; Herold J. Metselaar; Maikel P. Peppelenbosch; Qiuwei Pan

ABSTRACT Viruses are solely dependent on host cells to propagate; therefore, understanding virus-host interaction is important for antiviral drug development. Since de novo nucleotide biosynthesis is essentially required for both host cell metabolism and viral replication, specific catalytic enzymes of these pathways have been explored as potential antiviral targets. In this study, we investigated the role of different enzymatic cascades of nucleotide biosynthesis in hepatitis E virus (HEV) replication. By profiling various pharmacological inhibitors of nucleotide biosynthesis, we found that targeting the early steps of the purine biosynthesis pathway led to the enhancement of HEV replication, whereas targeting the later step resulted in potent antiviral activity via the depletion of purine nucleotide. Furthermore, the inhibition of the pyrimidine pathway resulted in potent anti-HEV activity. Interestingly, all of these inhibitors with anti-HEV activity concurrently triggered the induction of antiviral interferon-stimulated genes (ISGs). Although ISGs are commonly induced by interferons via the JAK-STAT pathway, their induction by nucleotide synthesis inhibitors is completely independent of this classical mechanism. In conclusion, this study revealed an unconventional novel mechanism of cross talk between nucleotide biosynthesis pathways and cellular antiviral immunity in constraining HEV infection. Targeting particular enzymes in nucleotide biosynthesis represents a viable option for antiviral drug development against HEV. HEV is the most common cause of acute viral hepatitis worldwide and is also associated with chronic hepatitis, especially in immunocompromised patients. Although often an acute and self-limiting infection in the general population, HEV can cause severe morbidity and mortality in certain patients, a problem compounded by the lack of FDA-approved anti-HEV medication available. In this study, we have investigated the role of the nucleotide synthesis pathway in HEV infection and its potential for antiviral drug development. We show that targeting the later but not the early steps of the purine synthesis pathway exerts strong anti-HEV activity. In particular, IMP dehydrogenase (IMPDH) is the most important anti-HEV target of this cascade. Importantly, the clinically used IMPDH inhibitors, including mycophenolic acid and ribavirin, have potent anti-HEV activity. Furthermore, targeting the pyrimidine synthesis pathway also exerts potent antiviral activity against HEV. Interestingly, antiviral effects of nucleotide synthesis pathway inhibitors appear to depend on the medication-induced transcription of antiviral interferon-stimulated genes. Thus, this study reveals an unconventional novel mechanism as to how nucleotide synthesis pathway inhibitors can counteract HEV replication.


Gastroenterology | 2016

Distinct Antiviral Potency of Sofosbuvir Against Hepatitis C and E Viruses.

Wenshi Wang; Mohamad S. Hakim; Vidya P. Nair; Petra E. de Ruiter; Fen Huang; Dave Sprengers; Luc J. W. van der Laan; Maikel P. Peppelenbosch; Milan Surjit; Qiuwei Pan

Readers may submit letters to the editor concerning articles that appeared in Gastroenterology within one month of publication. Detailed guidelines regarding the content are included in the Instructions to Authors. 65 66 67 68 69 Distinct Antiviral Potency of Sofosbuvir against Hepatitis C and E Viruses 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 Dear Editors: Sofosbuvir (SOF), the direct-acting anti–hepatitis C virus (HCV) drug (targeting HCV RdRp; RNA-dependent RNA polymerase), has been recently reported to be a potential anti-hepatitis E virus (HEV) drug candidate. However, some debates emerged whether SOF is a promising drug candidate for treating hepatitis E. Given the important potential clinical implications, this study has assessed comparatively the antiviral efficacy of SOF in both HCV and HEV models. We believe that the anti-HEV and anti-HCV potency of SOF should be assessed comparatively, before proposing its clinical application for treating HEV-infected patients. In our study, the potential anti-HEV effect of SOF was investigated in HEV replication models with concentrations ranging from 0.01 to 10 mmol/L (Supplementary Figure 1A),

Collaboration


Dive into the Wenshi Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiuwei Pan

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Lei Xu

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Yijin Wang

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Yuebang Yin

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Dave Sprengers

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Xinying Zhou

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Mohamad S. Hakim

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Herold J. Metselaar

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge