Network


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

Hotspot


Dive into the research topics where Emily C. Gurley is active.

Publication


Featured researches published by Emily C. Gurley.


Hepatology | 2008

Prevention of free fatty acid–induced hepatic lipotoxicity by 18β‐glycyrrhetinic acid through lysosomal and mitochondrial pathways

Xudong Wu; Luyong Zhang; Emily C. Gurley; Elaine Studer; Jing Shang; Tao Wang; Cuifen Wang; Ming Yan; Zhenzhou Jiang; Phillip B. Hylemon; Arun J. Sanyal; William M. Pandak; Huiping Zhou

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease and affects millions of people worldwide. Despite the increasing prevalence of NAFLD, the exact molecular/cellular mechanisms remain obscure and effective therapeutic strategies are still limited. It is well‐accepted that free fatty acid (FFA)‐induced lipotoxicity plays a pivotal role in the pathogenesis of NAFLD. Inhibition of FFA‐associated hepatic toxicity represents a potential therapeutic strategy. Glycyrrhizin (GL), the major bioactive component of licorice root extract, has a variety of pharmacological properties including anti‐inflammatory, antioxidant, and immune‐modulating activities. GL has been used to treat hepatitis to reduce liver inflammation and hepatic injury; however, the mechanism underlying the antihepatic injury property of GL is still poorly understood. In this report, we provide evidence that 18 β‐glycyrrhetinic acid (GA), the biologically active metabolite of GL, prevented FFA‐induced lipid accumulation and cell apoptosis in in vitro HepG2 (human liver cell line) NAFLD models. GA also prevented high fat diet (HFD)‐induced hepatic lipotoxicity and liver injury in in vivo rat NAFLD models. GA was found to stabilize lysosomal membranes, inhibit cathepsin B expression and enzyme activity, inhibit mitochondrial cytochrome c release, and reduce FFA‐induced oxidative stress. These characteristics may represent major cellular mechanisms, which account for its protective effects on FFA/HFD‐induced hepatic lipotoxicity. Conclusion: GA significantly reduced FFA/HFD‐induced hepatic lipotoxicity by stabilizing the integrity of lysosomes and mitochondria and inhibiting cathepsin B expression and enzyme activity. (HEPATOLOGY 2008.)


PLOS ONE | 2014

Flavonoid apigenin inhibits lipopolysaccharide-induced inflammatory response through multiple mechanisms in macrophages.

Xiaoxuan Zhang; Guangji Wang; Emily C. Gurley; Huiping Zhou

Background Apigenin is a non-toxic natural flavonoid that is abundantly present in common fruits and vegetables. It has been reported that apigenin has various beneficial health effects such as anti-inflammation and chemoprevention. Multiple studies have shown that inflammation is an important risk factor for atherosclerosis, diabetes, sepsis, various liver diseases, and other metabolic diseases. Although it has been long realized that apigenin has anti-inflammatory activities, the underlying functional mechanisms are still not fully understood. Methodology and Principal Findings In the present study, we examined the effect of apigenin on LPS-induced inflammatory response and further elucidated the potential underlying mechanisms in human THP-1-induced macrophages and mouse J774A.1 macrophages. By using the PrimePCR array, we were able to identify the major target genes regulated by apigenin in LPS-mediated immune response. The results indicated that apigenin significantly inhibited LPS-induced production of pro-inflammatory cytokines, such as IL-6, IL-1β, and TNF-α through modulating multiple intracellular signaling pathways in macrophages. Apigenin inhibited LPS-induced IL-1β production by inhibiting caspase-1 activation through the disruption of the NLRP3 inflammasome assembly. Apigenin also prevented LPS-induced IL-6 and IL-1β production by reducing the mRNA stability via inhibiting ERK1/2 activation. In addition, apigenin significantly inhibited TNF-α and IL-1β-induced activation of NF-κB. Conclusion and Significance Apigenin Inhibits LPS-induced Inflammatory Response through multiple mechanisms in macrophages. These results provided important scientific evidences for the potential application of apigenin as a therapeutic agent for inflammatory diseases.


Bioorganic & Medicinal Chemistry Letters | 2008

Synthesis and anti-inflammatory activities of mono-carbonyl analogues of curcumin

Guang Liang; Xiaokun Li; Li Chen; Shulin Yang; Xudong Wu; Elaine Studer; Emily C. Gurley; Phillip B. Hylemon; Faqing Ye; Yueru Li; Huiping Zhou

Curcumin has been extensively studied for its anti-inflammatory activities. However, its potential beneficial effects on various disease preventions and treatments are limited by its unstable structure. The beta-diketone moiety renders curcumin to be rapidly metabolized by aldo-keto reductase in liver. In the present study, a series of curcumin analogues with more stable chemical structures were synthesized and several compounds showed an enhanced ability to inhibit lipopolysaccharide (LPS)-induced TNF-alpha and IL-6 synthesis in macrophages.


Gastroenterology | 2010

HIV Protease Inhibitors Induce Endoplasmic Reticulum Stress and Disrupt Barrier Integrity in Intestinal Epithelial Cells

Xudong Wu; Lixin Sun; Weibin Zha; Elaine Studer; Emily C. Gurley; Li Chen; Xuan Wang; Phillip B. Hylemon; William M. Pandak; Arun J. Sanyal; Luyong Zhang; Guangji Wang; Jie Chen; Jian Ying Wang; Huiping Zhou

BACKGROUND & AIMS Human immunodeficiency virus (HIV) protease inhibitor (PI)-induced adverse effects have become a serious clinical problem. In addition to their metabolic and cardiovascular complications, these drugs also frequently cause severe gastrointestinal disorders, including mucosal erosions, epithelial barrier dysfunction, and diarrhea. However, the exact mechanisms underlying gastrointestinal adverse effects of HIV PIs remain unknown. This study investigated whether HIV PIs disrupt intestinal epithelial barrier integrity by activating endoplasmic reticulum (ER) stress. METHODS The most commonly used HIV PIs (lopinavir, ritonavir, and amprenavir) were used; their effects on ER stress activation and epithelial paracellular permeability were examined in vitro as well as in vivo using wild-type and CHOP(-)/(-) mice. RESULTS Treatment with lopinavir and ritonavir, but not amprenavir, induced ER stress, as indicated by a decrease in secreted alkaline phosphatase activities and an increase in the unfolded protein response. This activated ER stress partially impaired the epithelial barrier integrity by promoting intestinal epithelial cell apoptosis. CHOP silencing by specific small hairpin RNA prevented lopinavir- and ritonavir-induced barrier dysfunction in cultured intestinal epithelial cells, whereas CHOP(-)/(-) mice exhibited decreased mucosal injury after exposure to lopinavir and ritonavir. CONCLUSIONS HIV PIs induce ER stress and activate the unfolded protein response in intestinal epithelial cells, thus resulting in disruption of the epithelial barrier integrity.


Journal of Cellular and Molecular Medicine | 2009

Inhibition of LPS-induced production of inflammatory factors in the macrophages by mono-carbonyl analogues of curcumin

Guang Liang; Huiping Zhou; Emily C. Gurley; Biao Feng; Li Chen; Jian Xiao; Shulin Yang; Xiaokun Li

Curcumin (diferuloylmethane) is an orange–yellow compound from turmeric (Curcuma longa), a spice found in curry powder. Traditionally known for its anti‐inflammatory effects, curcumin has established itself in the last two decades to be a potent immunomodulatory agent that can regulate the activation of a variety of immunocytes and the expression of inflammatory factors. Considering that the β‐diketone moiety of curcumin may result in its instability and poor metabolic property, we previously designed a series of mono‐carbonyl analogues of curcumin with enhanced stability by deleting this moiety. These compounds demonstrate improved pharmacokinetic profiles both in vitro and in vivo. In this study, we reported a total of 44 mono‐carbonyl analogues, which have been evaluated for the inhibitory activities against LPS‐induced TNF‐α and IL‐6 release in the macrophages. Based on the screening results of these analogues, five active compounds A01, A03, A13, B18 and C22 were investigated to inhibit TNF‐α and IL‐6 release in a dose‐dependent manner, three of which further demonstrated inhibitory effects on LPS‐induced TNF‐α, IL‐1β, IL‐6, MCP‐1, COX‐2, PGES, iNOS and p65 NF‐κB mRNA production. The results indicated that these mono‐carbonyl analogues may possess anti‐inflammatory activities similar to curcumin despite the absence of the β‐diketone. These mono‐carbonyl analogues may be a favourable alternative for the development of curcumin‐based anti‐inflammatory drugs both pharmacokinetically and pharmacologically. We further examined the biological properties of A13, the only hydrosoluble analogue when combined with hydrochloric acid. The results showed a dose‐dependent inhibition of LPS‐induced cytokine production. These data further indicated that compound A13 may be explored as a promising anti‐inflammatory molecule.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2014

Colonic inflammation and secondary bile acids in alcoholic cirrhosis

Genta Kakiyama; Phillip B. Hylemon; Huiping Zhou; William M. Pandak; Douglas M. Heuman; Dae Joong Kang; Hajime Takei; Hiroshi Nittono; Jason M. Ridlon; Michael Fuchs; Emily C. Gurley; Yun F. Wang; Runping Liu; Arun J. Sanyal; Patrick M. Gillevet; Jasmohan S. Bajaj

Alcohol abuse with/without cirrhosis is associated with an impaired gut barrier and inflammation. Gut microbiota can transform primary bile acids (BA) to secondary BAs, which can adversely impact the gut barrier. The purpose of this study was to define the effect of active alcohol intake on fecal BA levels and ileal and colonic inflammation in cirrhosis. Five age-matched groups {two noncirrhotic (control and drinkers) and three cirrhotic [nondrinkers/nonalcoholics (NAlc), abstinent alcoholic for >3 mo (AbsAlc), currently drinking (CurrAlc)]} were included. Fecal and serum BA analysis, serum endotoxin, and stool microbiota using pyrosequencing were performed. A subgroup of controls, NAlc, and CurrAlc underwent ileal and sigmoid colonic biopsies on which mRNA expression of TNF-α, IL-1β, IL-6, and cyclooxygenase-2 (Cox-2) were performed. One hundred three patients (19 healthy, 6 noncirrhotic drinkers, 10 CurrAlc, 38 AbsAlc, and 30 NAlc, age 56 yr, median MELD: 10.5) were included. Five each of healthy, CurrAlc, and NAlc underwent ileal/colonic biopsies. Endotoxin, serum-conjugated DCA and stool total BAs, and secondary-to-primary BA ratios were highest in current drinkers. On biopsies, a significantly higher mRNA expression of TNF-α, IL-1β, IL-6, and Cox-2 in colon but not ileum was seen in CurrAlc compared with NAlc and controls. Active alcohol use in cirrhosis is associated with a significant increase in the secondary BA formation compared with abstinent alcoholic cirrhotics and nonalcoholic cirrhotics. This increase in secondary BAs is associated with a significant increase in expression of inflammatory cytokines in colonic mucosa but not ileal mucosa, which may contribute to alcohol-induced gut barrier injury.


Hepatology | 2017

The role of sphingosine 1-phosphate receptor 2 in bile-acid–induced cholangiocyte proliferation and cholestasis-induced liver injury in mice

Yongqing Wang; Hiroaki Aoki; Jing Yang; Kesong Peng; Runping Liu; Xiaojiaoyang Li; Xiaoyan Qiang; Lixin Sun; Emily C. Gurley; Guanhua Lai; Luyong Zhang; Guang Liang; Masayuki Nagahashi; Kazuaki Takabe; William M. Pandak; Phillip B. Hylemon; Huiping Zhou

Bile duct obstruction is a potent stimulus for cholangiocyte proliferation, especially for large cholangiocytes. Our previous studies reported that conjugated bile acids (CBAs) activate the protein kinase B (AKT) and extracellular signal‐regulated kinase 1 and 2 (ERK1/2) signaling pathways through sphingosine 1‐phosphate receptor (S1PR) 2 in hepatocytes and cholangiocarcinoma cells. It also has been reported that taurocholate (TCA) promotes large cholangiocyte proliferation and protects cholangiocytes from bile duct ligation (BDL)‐induced apoptosis. However, the role of S1PR2 in bile‐acid–mediated cholangiocyte proliferation and cholestatic liver injury has not been elucidated. Here, we report that S1PR2 is the predominant S1PR expressed in cholangiocytes. Both TCA‐ and sphingosine‐1‐phosphate (S1P)‐induced activation of ERK1/2 and AKT were inhibited by JTE‐013, a specific antagonist of S1PR2, in cholangiocytes. In addition, TCA‐ and S1P‐induced cell proliferation and migration were inhibited by JTE‐013 and a specific short hairpin RNA of S1PR2, as well as chemical inhibitors of ERK1/2 and AKT in mouse cholangiocytes. In BDL mice, expression of S1PR2 was up‐regulated in whole liver and cholangiocytes. S1PR2 deficiency significantly reduced BDL‐induced cholangiocyte proliferation and cholestatic injury, as indicated by significant reductions in inflammation and liver fibrosis in S1PR2 knockout mice. Treatment of BDL mice with JTE‐013 significantly reduced total bile acid levels in serum and cholestatic liver injury. Conclusion: This study suggests that CBA‐induced activation of S1PR2‐mediated signaling pathways plays a critical role in obstructive cholestasis and may represent a novel therapeutic target for cholestatic liver diseases. (Hepatology 2017;65:2005‐2018).


Hepatology | 2017

The role of long noncoding RNA H19 in gender disparity of cholestatic liver injury in multidrug resistance 2 gene knockout mice

Xiaojiaoyang Li; Runping Liu; Jing Yang; Lixin Sun; Luyong Zhang; Zhenzhou Jiang; Puneet Puri; Emily C. Gurley; Guan-Hua Lai; Yuping Tang; Zhiming Huang; William M. Pandak; Phillip B. Hylemon; Huiping Zhou

The multidrug resistance 2 knockout (Mdr2–/–) mouse is a well‐established model of cholestatic cholangiopathies. Female Mdr2–/– mice develop more severe hepatobiliary damage than male Mdr2–/– mice, which is correlated with a higher proportion of taurocholate in the bile. Although estrogen has been identified as an important player in intrahepatic cholestasis, the underlying molecular mechanisms of gender‐based disparity of cholestatic injury remain unclear. The long noncoding RNA H19 is an imprinted, maternally expressed, and estrogen‐targeted gene, which is significantly induced in human fibrotic/cirrhotic liver and bile duct–ligated mouse liver. However, whether aberrant expression of H19 accounts for gender‐based disparity of cholestatic injury in Mdr2–/– mice remains unknown. The current study demonstrated that H19 was markedly induced (∼200‐fold) in the livers of female Mdr2–/– mice at advanced stages of cholestasis (100 days old) but not in age‐matched male Mdr2–/– mice. During the early stages of cholestasis, H19 expression was minimal. We further determined that hepatic H19 was mainly expressed in cholangiocytes, not hepatocytes. Both taurocholate and estrogen significantly activated the extracellular signal–regulated kinase 1/2 signaling pathway and induced H19 expression in cholangiocytes. Knocking down H19 not only significantly reduced taurocholate/estrogen‐induced expression of fibrotic genes and sphingosine 1‐phosphate receptor 2 in cholangiocytes but also markedly reduced cholestatic injury in female Mdr2–/– mice. Furthermore, expression of small heterodimer partner was substantially inhibited at advanced stages of liver fibrosis, which was reversed by H19 short hairpin RNA in female Mdr2–/– mice. Similar findings were obtained in human primary sclerosing cholangitis liver samples. Conclusion: H19 plays a critical role in the disease progression of cholestasis and represents a key factor that causes the gender disparity of cholestatic liver injury in Mdr2–/– mice. (Hepatology 2017;66:869–884).


Hepatology | 2013

The role of CCAAT enhancer-binding protein homologous protein in human immunodeficiency virus protease-inhibitor-induced hepatic lipotoxicity in mice.

Yun F. Wang; Luyong Zhang; Xudong Wu; Emily C. Gurley; Elaine Kennedy; Phillip B. Hylemon; William M. Pandak; Arun J. Sanyal; Huiping Zhou

Human immunodeficiency virus (HIV) protease inhibitors (HIV PIs) are the core components of highly active antiretroviral therapy, which has been successfully used in the treatment of HIV‐1 infection in the past two decades. However, benefits of HIV PIs are compromised by clinically important adverse effects, such as dyslipidemia, insulin resistance, and cardiovascular complications. We have previously shown that activation of endoplasmic reticulum (ER) stress plays a critical role in HIV PI–induced dys‐regulation of hepatic lipid metabolism. HIV PI–induced hepatic lipotoxicity is closely linked to the up‐regulation of CCAAT enhancer binding protein homologous protein (CHOP) in hepatocytes. To further investigate whether CHOP is responsible for HIV PI–induced hepatic lipotoxicity, C57BL/6J wild‐type (WT) or CHOP knockout (CHOP−/−) mice or the corresponding primary mouse hepatocytes were used in this study. Both in vitro and in vivo studies indicated that HIV PIs (ritonavir and lopinavir) significantly increased hepatic lipid accumulation in WT mice. In contrast, CHOP−/− mice showed a significant reduction in hepatic triglyceride accumulation and liver injury, as evidenced by hematoxylin and eosin and Oil Red O staining. Real‐time reverse‐transcriptase polymerase chain reaction and immunoblotting data showed that in the absence of CHOP, HIV PI–induced expression of stress‐related proteins and lipogenic genes were dramatically reduced. Furthermore, tumor necrosis factor alpha and interleukin‐6 levels in serum and liver were significantly lower in HIV PI–treated CHOP−/− mice, compared to HIV PI–treated WT mice. Conclusion: Taken together, these data suggest that CHOP is an important molecular link of ER stress, inflammation, and hepatic lipotoxicity, and that increased expression of CHOP represents a critical factor underlying events leading to hepatic injury. (HEPATOLOGY 2013)


Journal of Pharmacology and Experimental Therapeutics | 2010

Prevention of HIV Protease Inhibitor-Induced Dysregulation of Hepatic Lipid Metabolism by Raltegravir via Endoplasmic Reticulum Stress Signaling Pathways

Risheng Cao; Yiqiao Hu; Yun F. Wang; Emily C. Gurley; Elaine Studer; Xuan Wang; Phillip B. Hylemon; William M. Pandak; Arun J. Sanyal; Luyong Zhang; Huiping Zhou

Hyperlipidemia associated with the HIV protease inhibitor (PI), the major component of highly active antiretroviral treatment (HAART) for HIV infection, has stimulated interest in developing new agents that minimize these side effects in the clinic. HIV integrase inhibitor is a new class of anti-HIV agents. Raltegravir is a first-in-its-class oral integrase inhibitor and has potent inhibitory activity against HIV-1 strains that are resistant to other antiretroviral regimens. Our previous studies have demonstrated that HIV PI-induced endoplasmic reticulum (ER) stress links to dysregulation of lipid metabolism. However, little information is available as to whether raltegravir would have similar effects as the HIV PIs. In this study, we examined the effect of raltegravir on lipid metabolism both in primary rat hepatocytes and in in vivo mouse models, and we further determined whether the combination of raltegravir with existing HIV PIs would potentially exacerbate or prevent the previously observed development of dyslipidemia. The results indicated that raltegravir did not induce ER stress or disrupt lipid metabolism either in vitro or in vivo. However, HIV PI-induced ER stress and lipid accumulation were significantly inhibited by raltegravir both in in vitro primary rat hepatocytes and in in vivo mouse liver. High-performance liquid chromatography analysis further demonstrated that raltegravir did not affect the uptake and metabolism of HIV PIs in hepatocytes. Thus, raltegravir has less hepatic toxicity and could prevent HIV PI-induced dysregulation of lipid metabolism by inhibiting ER stress. These results suggest that incorporation of this HIV integrase inhibitor may reduce the side effects associated with current HAART.

Collaboration


Dive into the Emily C. Gurley's collaboration.

Top Co-Authors

Avatar

Phillip B. Hylemon

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Huiping Zhou

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

William M. Pandak

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Elaine Studer

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Xudong Wu

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Arun J. Sanyal

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Li Chen

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Risheng Cao

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Weibin Zha

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Xuan Wang

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge