Network


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

Hotspot


Dive into the research topics where Emmanuel Thomas is active.

Publication


Featured researches published by Emmanuel Thomas.


Gastroenterology | 2012

HCV Infection Induces a Unique Hepatic Innate Immune Response Associated With Robust Production of Type III Interferons

Emmanuel Thomas; Veronica D. Gonzalez; Qisheng Li; Ankit A. Modi; Weiping Chen; Mazen Noureddin; Yaron Rotman; T. Jake Liang

BACKGROUND & AIMS Polymorphisms in the IL28B gene have been associated with clearance of hepatitis C virus (HCV), indicating a role for type III interferons (IFNs) in HCV infection. Little is known about the function of type III IFNs in intrinsic antiviral innate immunity. METHODS We used in vivo and in vitro models to characterize the role of the type III IFNs in HCV infection and analyzed gene expression in liver biopsy samples from HCV-infected chimpanzees and patients. Messenger RNA and protein expression were studied in HCV-infected hepatoma cell lines and primary human hepatocytes. RESULTS HCV infection of primary human hepatocytes induced production of chemokines and type III IFNs, including interleukin (IL)-28, and led to expression of IFN-stimulated genes (ISGs). Chimpanzees infected with HCV showed rapid induction of hepatic type III IFN, associated with up-regulation of ISGs and minimal induction of type I IFNs. In liver biopsy specimens from HCV-infected patients, hepatic expression of IL-28 correlated with levels of ISGs but not of type I IFNs. HCV infection produced extensive changes with gene expression in addition to ISGs in primary human hepatocytes. The induction of type III IFNs is regulated by IFN regulatory factor 3 and nuclear factor κB. Type III IFNs up-regulate ISGs with a different kinetic profile than type 1 IFNs and induce a distinct set of genes, which might account for their functional differences. CONCLUSIONS HCV infection results predominantly in induction of type III IFNs in livers of humans and chimpanzees; the level of induction correlates with hepatic levels of ISGs. These findings might account for the association among IL-28, level of ISGs, and recovery from HCV infection and provide a therapeutic strategy for patients who do not respond to IFN therapy.


Hepatology | 2011

Ribavirin potentiates interferon action by augmenting interferon-stimulated gene induction in hepatitis C virus cell culture models.

Emmanuel Thomas; Jordan J. Feld; Qisheng Li; Zongyi Hu; Michael W. Fried; T. Jake Liang

The combination of pegylated interferon (PEG‐IFN) and ribavirin is the standard treatment for chronic hepatitis C. Our recent clinical study suggests that ribavirin augments the induction of interferon‐stimulated genes (ISGs) in patients treated for hepatitis C virus (HCV) infection. In order to further characterize the mechanisms of action of ribavirin, we examined the effect of ribavirin treatment on ISG induction in cell culture. In addition, the effect of ribavirin on infectious HCV cell culture systems was studied. Similar to interferon (IFN)‐α, ribavirin potently inhibits JFH‐1 infection of Huh7.5.1 cells in a dose‐dependent manner, which spans the physiological concentration of ribavirin in vivo. Microarray analysis and subsequent quantitative polymerase chain reaction assays demonstrated that ribavirin treatment resulted in the induction of a distinct set of ISGs. These ISGs, including IFN regulatory factors 7 and 9, are known to play an important role in anti‐HCV responses. When ribavirin is used in conjunction with IFN‐α, induction of specific ISGs is synergistic when compared with either drug applied separately. Direct up‐regulation of these antiviral genes by ribavirin is mediated by a novel mechanism different from those associated with IFN signaling and intracellular double‐stranded RNA sensing pathways such as RIG‐I and MDA5. RNA interference studies excluded the activation of the Toll‐like receptor and nuclear factor κB pathways in the action of ribavirin. Conclusion: Our study suggests that ribavirin, acting by way of a novel innate mechanism, potentiates the anti‐HCV effect of IFN. Understanding the mechanism of action of ribavirin would be valuable in identifying novel antivirals (HEPATOLOGY 2011.)


Hepatology | 2013

Association of IL28B genotype with fibrosis progression and clinical outcomes in patients with chronic hepatitis C: A longitudinal analysis

Mazen Noureddin; Elizabeth C. Wright; Harvey J. Alter; Shauna Clark; Emmanuel Thomas; Richard Chen; Xiongce Zhao; Cathy Conry-Cantilena; David E. Kleiner; T. Jake Liang; Marc G. Ghany

Interleukin (IL)28B polymorphisms are associated with spontaneous clearance of hepatitis C virus (HCV) infection and response to therapy. Whether IL28B genotype affects fibrosis progression or clinical outcome is unclear. Our aim was to study the relationship between IL28B genotype and both histological and clinical outcomes in patients with chronic hepatitis C (CHC). Hepatic fibrosis was scored using the Ishak (0‐6) scale; progression was defined as a 2‐point increase in Ishak score between biopsies. Multiple logistic and Cox regressions were used to identify variables associated with fibrosis progression. In all, 1,483 patients were included in a baseline cross‐sectional analysis, from which 276 were eligible for a paired biopsy analysis (median time between biopsies 4 years), and 400 for a clinical outcome analysis. At baseline biopsy, patients with IL28B CC genotype had significantly higher portal inflammation (2.4 versus 2.2) and alanine aminotransferase (ALT) levels (133 versus 105 U/L; P < 0.05 for all). In the paired biopsy analysis, there was no difference in the frequency of fibrosis progression between patients with IL28B CC and non‐CC genotypes (17% versus 23%). In logistic regression, only higher baseline alkaline phosphatase, lower platelets, and greater hepatic steatosis were associated with fibrosis progression. Patients with IL28B CC were twice as likely to develop adverse clinical outcomes compared to non‐CC (32% versus 16%; P = 0.007). Conclusion: IL28B CC genotype was associated with greater hepatic necroinflammation, higher ALT, and worse clinical outcomes in CHC patients. This suggests that IL28B CC is associated with a state of enhanced immunity that, on the one hand, can promote viral clearance, but alternately can increase necroinflammation and hepatic decompensation without enhancing fibrosis progression. (Hepatology 2013;58:1548–1557)


PLOS Pathogens | 2014

Integrative functional genomics of hepatitis C virus infection identifies host dependencies in complete viral replication cycle.

Qisheng Li; Yong–Yuan Zhang; Stephan Chiu; Zongyi Hu; Keng Hsin Lan; Helen Cha; Catherine Sodroski; Fang Zhang; Ching Sheng Hsu; Emmanuel Thomas; T. Jake Liang

Recent functional genomics studies including genome-wide small interfering RNA (siRNA) screens demonstrated that hepatitis C virus (HCV) exploits an extensive network of host factors for productive infection and propagation. How these co-opted host functions interact with various steps of HCV replication cycle and exert pro- or antiviral effects on HCV infection remains largely undefined. Here we present an unbiased and systematic strategy to functionally interrogate HCV host dependencies uncovered from our previous infectious HCV (HCVcc) siRNA screen. Applying functional genomics approaches and various in vitro HCV model systems, including HCV pseudoparticles (HCVpp), single-cycle infectious particles (HCVsc), subgenomic replicons, and HCV cell culture systems (HCVcc), we identified and characterized novel host factors or pathways required for each individual step of the HCV replication cycle. Particularly, we uncovered multiple HCV entry factors, including E-cadherin, choline kinase α, NADPH oxidase CYBA, Rho GTPase RAC1 and SMAD family member 6. We also demonstrated that guanine nucleotide binding protein GNB2L1, E2 ubiquitin-conjugating enzyme UBE2J1, and 39 other host factors are required for HCV RNA replication, while the deubiquitinating enzyme USP11 and multiple other cellular genes are specifically involved in HCV IRES-mediated translation. Families of antiviral factors that target HCV replication or translation were also identified. In addition, various virologic assays validated that 66 host factors are involved in HCV assembly or secretion. These genes included insulin-degrading enzyme (IDE), a proviral factor, and N-Myc down regulated Gene 1 (NDRG1), an antiviral factor. Bioinformatics meta-analyses of our results integrated with literature mining of previously published HCV host factors allows the construction of an extensive roadmap of cellular networks and pathways involved in the complete HCV replication cycle. This comprehensive study of HCV host dependencies yields novel insights into viral infection, pathogenesis and potential therapeutic targets.


Gut | 2014

Effect of ribavirin on viral kinetics and liver gene expression in chronic hepatitis C

Yaron Rotman; Mazen Noureddin; Jordan J. Feld; Jeremie Guedj; Michael Witthaus; Hwalih Han; Yoon J. Park; Su–Hyung Park; Theo Heller; Marc G. Ghany; Edward Doo; Christopher Koh; Adil A. Abdalla; Naveen Gara; Souvik Sarkar; Emmanuel Thomas; Golo Ahlenstiel; Birgit Edlich; Rachel Titerence; Leah J. Hogdal; Barbara Rehermann; Harel Dahari; Alan S. Perelson; Jay H. Hoofnagle; T. Jake Liang

Objective Ribavirin improves treatment response to pegylated-interferon (PEG-IFN) in chronic hepatitis C but the mechanism remains controversial. We studied correlates of response and mechanism of action of ribavirin in treatment of hepatitis C. Design 70 treatment-naive patients were randomised to 4 weeks of ribavirin (1000–1200 mg/d) or none, followed by PEG-IFNα-2a and ribavirin at standard doses and durations. Patients were also randomised to a liver biopsy 24 h before or 6 h after starting PEG-IFN. Hepatic gene expression was assessed by microarray and interferon-stimulated gene (ISG) expression quantified by nCounter platform. Temporal changes in ISG expression were assessed by qPCR in peripheral-blood mononuclear cells (PBMC) and by serum levels of IP-10. Results After 4 weeks of ribavirin monotherapy, hepatitis C virus (HCV) levels decreased by 0.5±0.5 log10 (p=0.009 vs controls) and ALT by 33% (p<0.001). Ribavirin pretreatment, while modestly augmenting ISG induction by PEG-IFN, did not modify the virological response to subsequent PEG-IFN and ribavirin treatment. However, biochemical, but not virological, response to ribavirin monotherapy predicted response to subsequent combination treatment (rapid virological response, 71% in biochemical responders vs 22% non-responders, p=0.01; early virological response, 100% vs 68%, p=0.03; sustained virological response 83% vs 41%, p=0.053). Ribavirin monotherapy lowered serum IP-10 levels but had no effect on ISG expression in PBMC. Conclusions Ribavirin is a weak antiviral but its clinical effect seems to be mediated by a separate, indirect mechanism, which may act to reset IFN-responsiveness in HCV-infected liver.


Gastroenterology | 2011

S-adenosyl methionine improves early viral responses and interferon-stimulated gene induction in hepatitis C nonresponders

Jordan J. Feld; Apurva A. Modi; Ramy El–Diwany; Yaron Rotman; Emmanuel Thomas; Golo Ahlenstiel; Rachel Titerence; Christopher Koh; Vera Cherepanov; Theo Heller; Marc G. Ghany; Yoon Park; Jay H. Hoofnagle; T. Jake Liang

BACKGROUND & AIMS Less than half of patients infected with hepatitis C virus (HCV) achieve sustained viral clearance after pegylated interferon (peginterferon) and ribavirin therapy. S-adenosyl methionine (SAMe) improves interferon signaling in cell culture. We assessed the effect of SAMe on the kinetics of the early antiviral response and interferon signaling in nonresponders to previous antiviral therapy and investigated the mechanisms involved. METHODS Nonresponders with HCV genotype 1 were given peginterferon alfa-2a and ribavirin for 2 weeks (course A, baseline/control). After 1 month, patients received SAMe (1600 mg daily) for 2 weeks and then peginterferon and ribavirin for 48 weeks (course B; completed by 21 of 24 patients). Viral kinetics and interferon-stimulated gene (ISG) expression in peripheral blood mononuclear cells (PBMCs) were compared between courses. RESULTS The decrease in HCV RNA from 0 to 48 hours (phase 1) was similar with and without SAMe. However, the second phase slope of viral decline was improved with SAMe (course A, 0.11 ± 0.04 log(10) IU/mL/wk; course B, 0.27 ± 0.06; P = .009); 11 patients (53%) achieved an early virological response, and 10 (48%) had undetectable HCV RNA by week 24. Induction of ISGs in PBMCs was significantly greater during course B. In cultured cells, SAMe increased induction of ISGs and the antiviral effects of interferon by increasing STAT1 methylation, possibly affecting STAT1-DNA binding. CONCLUSIONS The addition of SAMe to peginterferon and ribavirin improves the early viral kinetics and increases ISG induction in nonresponders to previous therapy. SAMe might be a useful adjunct to peginterferon-based therapies in chronic HCV infection.


Genes and Immunity | 2015

Hepatic expression levels of interferons and interferon-stimulated genes in patients with chronic hepatitis C: A phenotype–genotype correlation study

M. Noureddin; Yaron Rotman; Fang Zhang; H. Park; Barbara Rehermann; Emmanuel Thomas; T. J. Liang

IFNL4 is linked to hepatitis C virus treatment response and type III interferons (IFNs). We studied the functional associations among hepatic expressions of IFNs and IFN-stimulated genes (ISGs), and treatment response to peginterferon and ribavirin. Type I IFNs (IFNA1, IFNB1), type II (IFNG), type III (IFNL1, IFNL2/3), IFNL4 and ISG hepatic expressions were measured by qPCR from in 65 chronic hepatitis C (CHC) patients whose IFNL4-associated rs368234815 and IFNL3-associated rs12989760 genotype were determined. There was a robust correlation of hepatic expression within type I and type III IFNs and between type III IFNs and IFNL4 but no correlation between other IFN types. Expression of ISGs correlated with type III IFNs and IFNL4 but not with type I IFNs. Levels of ISGs and IFNL2/3 mRNAs were lower in IFNL3 rs12979860 CC patients compared with non-CC patients, and in treatment responders, compared with nonresponders. IFNL4-ΔG genotype was associated with high ISG levels and nonresponse. Hepatic levels of ISGs in CHC are associated with IFNL2/3 and IFNL4 expression, suggesting that IFNLs, not other types of IFNs, drive ISG expression. Hepatic IFNL2/3 expression is functionally linked to IFNL4 and IFNL3 polymorphisms, potentially explaining the tight association among ISG expression and treatment response.


Gastroenterología y Hepatología | 2014

Effects of statins on the risk of hepatocellular carcinoma.

Pejman G. Mansourian; Masato Yoneda; M. Krishna Rao; Fernando Martínez; Emmanuel Thomas; Eugene R. Schiff


Cancer Research | 2018

Abstract 5539: An integrated multidisciplinary approach for prediction of HCC development in patients with liver disease utilizing genomics, elastography, imaging and circulating tumor cells

Emmanuel Thomas; Eugene R. Schiff; Deukwoo Kwon; Beatrice L. Madrazo


Gastroenterología y Hepatología | 2015

Genomics in the Early Detection of Cholangiocarcinoma.

Emmanuel Thomas

Collaboration


Dive into the Emmanuel Thomas's collaboration.

Top Co-Authors

Avatar

T. Jake Liang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yaron Rotman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jordan J. Feld

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mazen Noureddin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qisheng Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc G. Ghany

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ankit A. Modi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christopher Koh

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel Titerence

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge