Network


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

Hotspot


Dive into the research topics where Lirui Wang is active.

Publication


Featured researches published by Lirui Wang.


Hepatology | 2013

Deficiency of intestinal mucin-2 ameliorates experimental alcoholic liver disease in mice.

Phillipp Hartmann; Peng Chen; Hui J. Wang; Lirui Wang; Declan F. McCole; Katharina Brandl; Peter Stärkel; Clara Belzer; Claus Hellerbrand; Hidekazu Tsukamoto; Samuel B. Ho; Bernd Schnabl

The intestinal mucus layer protects the epithelium from noxious agents, viruses, and pathogenic bacteria present in the gastrointestinal tract. It is composed of mucins, predominantly mucin (Muc) 2, secreted by goblet cells of the intestine. Experimental alcoholic liver disease requires translocation of bacterial products across the intestinal barrier into the systemic circulation, which induces an inflammatory response in the liver and contributes to steatohepatitis. We investigated the roles of the intestinal mucus layer, and in particular Muc2, in development of experimental alcohol‐associated liver disease in mice. We studied experimental alcohol‐induced liver disease, induced by the Tsukamoto‐French method (which involves continuous intragastric feeding of an isocaloric diet or alcohol) in wild‐type and Muc2−/− mice. Muc2−/− mice showed less alcohol‐induced liver injury and steatosis than developed in wild‐type mice. Most notably, Muc2−/− mice had significantly lower plasma levels of lipopolysaccharide than wild‐type mice after alcohol feeding. In contrast to wild‐type mice, Muc2−/− mice were protected from alcohol‐associated microbiome changes that are dependent on intestinal mucins. The antimicrobial proteins regenerating islet‐derived 3 beta and gamma were expressed at significantly higher levels in the jejunum of Muc2−/− mice fed the isocaloric diet or alcohol compared with wild‐type mice. Consequently, Muc2−/− mice showed increased killing of commensal bacteria and prevented intestinal bacterial overgrowth. Conclusion: Muc2−/− mice are protected from intestinal bacterial overgrowth and dysbiosis in response to alcohol feeding. Subsequently, lower amounts of bacterial products such as endotoxin translocate into the systemic circulation, decreasing liver disease. (HEPATOLOGY 2013;)


FEBS Journal | 2006

Biochemical characterization and inhibitor discovery of shikimate dehydrogenase from Helicobacter pylori

Cong Han; Lirui Wang; Kunqian Yu; Lili Chen; Lihong Hu; Kaixian Chen; Hualiang Jiang; Xu Shen

Shikimate dehydrogenase (SDH) is the fourth enzyme involved in the shikimate pathway. It catalyzes the NADPH‐dependent reduction of 3‐dehydroshikimate to shikimate, and has been developed as a promising target for the discovery of antimicrobial agent. In this report, we identified a new aroE gene encoding SDH from Helicobacter pylori strain SS1. The recombinant H. pylori shikimate dehydrogenase (HpSDH) was cloned, expressed, and purified in Escherichia coli system. The enzymatic characterization of HpSDH demonstrates its activity with kcat of 7.7 s−1 and Km of 0.148 mm toward shikimate, kcat of 7.1 s−1 and Km of 0.182 mm toward NADP, kcat of 5.2 s−1 and Km of 2.9 mm toward NAD. The optimum pH of the enzyme activity is between 8.0 and 9.0, and the optimum temperature is around 60 °C. Using high throughput screening against our laboratory chemical library, five compounds, curcumin (1), 3‐(2‐naphthyloxy)‐4‐oxo‐2‐(trifluoromethyl)‐4H‐chromen‐7‐yl 3‐chlorobenzoate (2), butyl 2‐{[3‐(2‐naphthyloxy)‐4‐oxo‐2‐(trifluoromethyl)‐4H‐chromen‐7‐yl]oxy}propanoate (3), 2‐({2‐[(2‐{[2‐(2,3‐dimethylanilino)‐2‐oxoethyl]sulfanyl}‐1,3‐benzothiazol‐6‐yl)amino]‐2‐oxoethyl}sulfanyl)‐N‐(2‐naphthyl)acetamide (4), and maesaquinone diacetate (5) were discovered as HpSDH inhibitors with IC50 values of 15.4, 3.9, 13.4, 2.9, and 3.5 µm, respectively. Further investigation indicates that compounds 1, 2, 3, and 5 demonstrate noncompetitive inhibition pattern, and compound 4 displays competitive inhibition pattern with respect to shikimate. Compounds 1, 4, and 5 display noncompetitive inhibition mode, and compounds 2 and 3 show competitive inhibition mode with respect to NADP. Antibacterial assays demonstrate that compounds 1, 2, and 5 can inhibit the growth of H. pylori with MIC of 16, 16, and 32 µg·mL−1, respectively. This current work is expected to favor better understanding the features of SDH and provide useful information for the development of novel antibiotics to treat H. pylori‐associated infection.


Cell Host & Microbe | 2016

Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation.

Lirui Wang; Derrick E. Fouts; Peter Stärkel; Phillipp Hartmann; Peng Chen; Cristina Llorente; Jessica DePew; Kelvin Moncera; Samuel B. Ho; David A. Brenner; Lora V. Hooper; Bernd Schnabl

Approximately half of all deaths from liver cirrhosis, the tenth leading cause of mortality in the United States, are related to alcohol use. Chronic alcohol consumption is accompanied by intestinal dysbiosis and bacterial overgrowth, yet little is known about the factors that alter the microbial composition or their contribution to liver disease. We previously associated chronic alcohol consumption with lower intestinal levels of the antimicrobial-regenerating islet-derived (REG)-3 lectins. Here, we demonstrate that intestinal deficiency in REG3B or REG3G increases numbers of mucosa-associated bacteria and enhances bacterial translocation to the mesenteric lymph nodes and liver, promoting the progression of ethanol-induced fatty liver disease toward steatohepatitis. Overexpression of Reg3g in intestinal epithelial cells restricts bacterial colonization of mucosal surfaces, reduces bacterial translocation, and protects mice from alcohol-induced steatohepatitis. Thus, alcohol appears to impair control of the mucosa-associated microbiota, and subsequent breach of the mucosal barrier facilitates progression of alcoholic liver disease.


The FASEB Journal | 2015

Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice

Magdalena Mazagova; Lirui Wang; Andrew T. Anfora; Max Wissmueller; Scott A. Lesley; Yukiko Miyamoto; Lars Eckmann; Wimal Pathmasiri; Susan Sumner; Caroline Westwater; David A. Brenner; Bernd Schnabl

Translocation of bacteria and their products across the intestinal barrier is common in patients with liver disease, and there is evidence that experimental liver fibrosis depends on bacterial translocation. The purpose of our study was to investigate liver fibrosis in conventional and germ‐free (GF) C57BL/6 mice. Chronic liver injury was induced by administration of thioacetamide (TAA) in the drinking water for 21 wk or by repeated intraperitoneal injections of carbon tetrachloride (CCl4). Increased liver fibrosis was observed in GF mice compared with conventional mice. Hepatocytes showed more toxin‐induced oxidative stress and cell death. This was accompanied by increased activation of hepatic stellate cells, but hepatic mediators of inflammation were not significantly different. Similarly, a genetic model using Myd88/Trif‐deficient mice, which lack downstream innate immunity signaling, had more severe fibrosis than wild‐type mice. Isolated Myd88/Trif‐deficient hepatocytes were more susceptible to toxin‐induced cell death in culture. In conclusion, the commensal microbiota prevents fibrosis upon chronic liver injury in mice. This is the first study describing a beneficial role of the commensal microbiota in maintaining liver homeostasis and preventing liver fibrosis.—Mazagova, M., Wang, L., Anfora, A. T., Wissmueller, M., Lesley, S. A., Miyamoto, Y., Eckmann, L., Dhungana, S., Pathmasiri, W., Sumner, S., Westwater, C., Brenner, D. A., Schnabl, B., Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice. FASEB J. 29, 1043–1055 (2015). www.fasebj.org


Journal of Immunological Methods | 2015

Methods to determine intestinal permeability and bacterial translocation during liver disease

Lirui Wang; Cristina Llorente; Phillipp Hartmann; An-Ming Yang; Peng Chen; Bernd Schnabl

Liver disease is often times associated with increased intestinal permeability. A disruption of the gut barrier allows microbial products and viable bacteria to translocate from the intestinal lumen to extraintestinal organs. The majority of the venous blood from the intestinal tract is drained into the portal circulation, which is part of the dual hepatic blood supply. The liver is therefore the first organ in the body to encounter not only absorbed nutrients, but also gut-derived bacteria and pathogen associated molecular patterns (PAMPs). Chronic exposure to increased levels of PAMPs has been linked to disease progression during early stages and to infectious complications during late stages of liver disease (cirrhosis). It is therefore important to assess and monitor gut barrier dysfunction during hepatic disease. We review methods to assess intestinal barrier disruption and discuss advantages and disadvantages. We will in particular focus on methods that we have used to measure increased intestinal permeability and bacterial translocation in experimental liver disease models.


Journal of Clinical Investigation | 2017

Intestinal fungi contribute to development of alcoholic liver disease

An-Ming Yang; Tatsuo Inamine; Katrin Hochrath; Peng Chen; Lirui Wang; Cristina Llorente; Sena Bluemel; Phillipp Hartmann; Jun Xu; Yukinori Koyama; Tatiana Kisseleva; Manolito Torralba; Kelvin Moncera; Karen Beeri; Chien-Sheng Chen; K Freese; C Hellerbrand; Serene M.L. Lee; Hal M. Hoffman; Wajahat Z. Mehal; Guadalupe Garcia-Tsao; Ece Mutlu; Ali Keshavarzian; Gordon D. Brown; Samuel B. Ho; Ramon Bataller; Peter Stärkel; Derrick E. Fouts; Bernd Schnabl

Chronic liver disease with cirrhosis is the 12th leading cause of death in the United States, and alcoholic liver disease accounts for approximately half of all cirrhosis deaths. Chronic alcohol consumption is associated with intestinal bacterial dysbiosis, yet we understand little about the contribution of intestinal fungi, or mycobiota, to alcoholic liver disease. Here we have demonstrated that chronic alcohol administration increases mycobiota populations and translocation of fungal &bgr;-glucan into systemic circulation in mice. Treating mice with antifungal agents reduced intestinal fungal overgrowth, decreased &bgr;-glucan translocation, and ameliorated ethanol-induced liver disease. Using bone marrow chimeric mice, we found that &bgr;-glucan induces liver inflammation via the C-type lectin–like receptor CLEC7A on Kupffer cells and possibly other bone marrow–derived cells. Subsequent increases in IL-1&bgr; expression and secretion contributed to hepatocyte damage and promoted development of ethanol-induced liver disease. We observed that alcohol-dependent patients displayed reduced intestinal fungal diversity and Candida overgrowth. Compared with healthy individuals and patients with non–alcohol-related cirrhosis, alcoholic cirrhosis patients had increased systemic exposure and immune response to mycobiota. Moreover, the levels of extraintestinal exposure and immune response correlated with mortality. Thus, chronic alcohol consumption is associated with an altered mycobiota and translocation of fungal products. Manipulating the intestinal mycobiome might be an effective strategy for attenuating alcohol-related liver disease.


Nature Communications | 2017

Gastric acid suppression promotes alcoholic liver disease by inducing overgrowth of intestinal Enterococcus

Cristina Llorente; Peter Jepsen; Tatsuo Inamine; Lirui Wang; Sena Bluemel; Hui J. Wang; Rohit Loomba; Jasmohan S. Bajaj; Mitchell L. Schubert; Masoumeh Sikaroodi; Patrick M. Gillevet; Jun Xu; Tatiana Kisseleva; Samuel B. Ho; Jessica DePew; Xin Du; Henrik Toft Sørensen; Hendrik Vilstrup; Karen E. Nelson; David A. Brenner; Derrick E. Fouts; Bernd Schnabl

Chronic liver disease is rising in western countries and liver cirrhosis is the 12th leading cause of death worldwide. Simultaneously, use of gastric acid suppressive medications is increasing. Here, we show that proton pump inhibitors promote progression of alcoholic liver disease, non-alcoholic fatty liver disease, and non-alcoholic steatohepatitis in mice by increasing numbers of intestinal Enterococcus spp. Translocating enterococci lead to hepatic inflammation and hepatocyte death. Expansion of intestinal Enterococcus faecalis is sufficient to exacerbate ethanol-induced liver disease in mice. Proton pump inhibitor use increases the risk of developing alcoholic liver disease among alcohol-dependent patients. Reduction of gastric acid secretion therefore appears to promote overgrowth of intestinal Enterococcus, which promotes liver disease, based on data from mouse models and humans. Recent increases in the use of gastric acid-suppressive medications might contribute to the increasing incidence of chronic liver disease.Proton pump inhibitors (PPIs) reduce gastric acid secretion and modulate gut microbiota composition. Here Llorente et al. show that PPIs induce bacterial overgrowth of enterococci, which, in turn, exacerbate ethanol-induced liver disease both in mice and humans.


Journal of Hepatology | 2014

Nod2 deficiency protects mice from cholestatic liver disease by increasing renal excretion of bile acids

Lirui Wang; Phillipp Hartmann; Michael Haimerl; Sai Praneeth R. Bathena; Christopher Sjöwall; Sven Almer; Yazen Alnouti; Alan F. Hofmann; Bernd Schnabl

BACKGROUND & AIMS Chronic liver disease is characterized by fibrosis that may progress to cirrhosis. Nucleotide oligomerization domain 2 (Nod2), a member of the Nod-like receptor (NLR) family of intracellular immune receptors, plays an important role in the defense against bacterial infection through binding to the ligand muramyl dipeptide (MDP). Here, we investigated the role of Nod2 in the development of liver fibrosis. METHODS We studied experimental cholestatic liver disease induced by bile duct ligation or toxic liver disease induced by carbon tetrachloride in wild type and Nod2(-/-) mice. RESULTS Nod2 deficiency protected mice from cholestatic but not toxin-induced liver injury and fibrosis. Most notably, the hepatic bile acid concentration was lower in Nod2(-/-) mice than wild type mice following bile duct ligation for 3 weeks. In contrast to wild type mice, Nod2(-/-) mice had increased urinary excretion of bile acids, including sulfated bile acids, and an upregulation of the bile acid efflux transporters MRP2 and MRP4 in tubular epithelial cells of the kidney. MRP2 and MRP4 were downregulated by IL-1β in a Nod2 dependent fashion. CONCLUSIONS Our findings indicate that Nod2 deficiency protects mice from cholestatic liver injury and fibrosis through enhancing renal excretion of bile acids that in turn contributes to decreased concentration of bile acids in the hepatocyte.


Hepatology | 2018

Modulation of the intestinal bile acid/farnesoid X receptor/fibroblast growth factor 15 axis improves alcoholic liver disease in mice

Phillipp Hartmann; Katrin Hochrath; Angela Horvath; Peng Chen; Caroline T. Seebauer; Cristina Llorente; Lirui Wang; Yazen Alnouti; Derrick E. Fouts; Peter Stärkel; Rohit Loomba; Sally Coulter; Christopher Liddle; Ruth T. Yu; Lei Ling; Stephen J. Rossi; Alex M. DePaoli; Michael Downes; Ronald M. Evans; David A. Brenner; Bernd Schnabl

Alcoholic liver disease (ALD) is associated with changes in the intestinal microbiota. Functional consequences of alcohol‐associated dysbiosis are largely unknown. The aim of this study was to identify a mechanism of how changes in the intestinal microbiota contribute to ALD. Metagenomic sequencing of intestinal contents demonstrated that chronic ethanol feeding in mice is associated with an over‐representation of bacterial genomic DNA encoding choloylglycine hydrolase, which deconjugates bile acids in the intestine. Bile acid analysis confirmed an increased amount of unconjugated bile acids in the small intestine after ethanol administration. Mediated by a lower farnesoid X receptor (FXR) activity in enterocytes, lower fibroblast growth factor (FGF)‐15 protein secretion was associated with increased hepatic cytochrome P450 enzyme (Cyp)‐7a1 protein expression and circulating bile acid levels. Depletion of the commensal microbiota with nonabsorbable antibiotics attenuated hepatic Cyp7a1 expression and reduced ALD in mice, suggesting that increased bile acid synthesis is dependent on gut bacteria. To restore intestinal FXR activity, we used a pharmacological intervention with the intestine‐restricted FXR agonist fexaramine, which protected mice from ethanol‐induced liver injury. Whereas bile acid metabolism was only minimally altered, fexaramine treatment stabilized the gut barrier and significantly modulated hepatic genes involved in lipid metabolism. To link the beneficial metabolic effect to FGF15, a nontumorigenic FGF19 variant—a human FGF15 ortholog—was overexpressed in mice using adeno‐associated viruses. FGF19 treatment showed similarly beneficial metabolic effects and ameliorated alcoholic steatohepatitis. Conclusion: Taken together, alcohol‐associated metagenomic changes result in alterations of bile acid profiles. Targeted interventions improve bile acid–FXR–FGF15 signaling by modulation of hepatic Cyp7a1 and lipid metabolism, and reduce ethanol‐induced liver disease in mice. (Hepatology 2018;67:2150‐2166).


PLOS ONE | 2010

Activation of protein serine/threonine phosphatase PP2Cα efficiently prevents liver fibrosis.

Lirui Wang; Xu Wang; Jing Chen; Zhengyi Yang; Liang Yu; Lihong Hu; Xu Shen

Background Over-activation of TGFβ signaling pathway and uncontrolled cell proliferation of hepatic stellate cells (HSCs) play pivotal roles in liver fibrogenesis, while the protein serine/threonine phosphatase PP2Cα was reported to negatively regulate TGFβ signaling pathway and cell cycle. Our study aimed to investigate the role of PP2Cα in liver fibrogenesis. Methodology/Principal Findings The effects of PP2Cα activation on liver fibrosis were investigated in human HSCs and primary rat HSCs in vitro using western blotting, real-time PCR, nuclear translocation, cell viability and cell cycle analyses. The antifibrogenic effects in carbon tetrachloride (CCl4)- and bile duct ligation (BDL)-induced mice in vivo were assessed using biochemical, histological and immunohistochemical analyses. The results demonstrated that activation of PP2Cα by overexpression or the new discovered small molecular activator NPLC0393 terminated TGFβ-Smad3 and TGFβ-p38 signaling pathways, induced cell cycle arrest in HSCs and decreased α-smooth muscle actin (α-SMA) expression, collagen deposition and hepatic hydroxyproline (HYP) level in CCl4- and BDL-induced mice. Conclusions/Significance Our findings suggested that PP2Cα activation might be an attractive new strategy for treating liver fibrosis while the small molecular activator NPLC0393 might represent a lead compound for antifibrogenic drug development. Moreover, our study might provide the first evidence for the role of PP2C family members in the fibrotic disease.

Collaboration


Dive into the Lirui Wang's collaboration.

Top Co-Authors

Avatar

Bernd Schnabl

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel B. Ho

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peng Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Sena Bluemel

University of California

View shared research outputs
Top Co-Authors

Avatar

Tatsuo Inamine

University of California

View shared research outputs
Top Co-Authors

Avatar

Peter Stärkel

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge