Network


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

Hotspot


Dive into the research topics where Donna Yu is active.

Publication


Featured researches published by Donna Yu.


Hepatology | 2008

Farnesoid X receptor antagonizes nuclear factor κB in hepatic inflammatory response

Yan-Dong Wang; Wei-Dong Chen; Meihua Wang; Donna Yu; Barry M. Forman; Wendong Huang

The farnesoid X receptor (FXR) is a nuclear receptor that plays key roles in hepatoprotection by maintaining the homeostasis of liver metabolism. FXR null mice display strong hepatic inflammation and develop spontaneous liver tumors. In this report, we demonstrate that FXR is a negative modulator of nuclear factor κB (NF‐κB)–mediated hepatic inflammation. Activation of FXR by its agonist ligands inhibited the expression of inflammatory mediators in response to NF‐κB activation in both HepG2 cells and primary hepatocytes cultured in vitro. In vivo, compared with wild‐type controls, FXR−/− mice displayed elevated messenger RNA (mRNA) levels of inducible nitric oxide synthase (iNOS), cyclooxygenase‐2 (COX‐2), interferon‐inducible protein 10, and interferon‐γ in response to lipopolysaccharide (LPS). Examination of FXR−/− livers showed massive necroses and inflammation after treatment with LPS at a dose that does not induce significant liver damage or inflammation in wild‐type mice. Moreover, transfection of a constitutively active FXR expression construct repressed the iNOS, COX‐2, interferon‐inducible protein 10 and interferon‐γ mRNA levels induced by LPS administration. FXR activation had no negative effects on NF‐κB‐activated antiapoptotic genes, suggesting that FXR selectively inhibits the NF‐κB‐mediated hepatic inflammatory response but maintains or even enhances the cell survival response. On the other hand, NF‐κB activation suppressed FXR‐mediated gene expression both in vitro and in vivo, indicating a negative crosstalk between the FXR and NF‐κB signaling pathways. Our findings reveal that FXR is a negative mediator of hepatic inflammation, which may contribute to the critical roles of FXR in hepatoprotection and suppression of hepatocarcinogenesis. (HEPATOLOGY 2008;48:1632–1643.)


Hepatology | 2011

The G-Protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) in mice†‡§

Yan-Dong Wang; Wei-Dong Chen; Donna Yu; Barry M. Forman; Wendong Huang

Gpbar1 (TGR5), a membrane‐bound bile acid receptor, is well known for its roles in regulation of energy homeostasis and glucose metabolism. TGR5 also displays strong attenuation of macrophage reactivity in vitro, but the physiological roles of TGR5 in inflammatory response, and its mechanism, is unknown. Here, we demonstrate that TGR5 is a negative modulator of nuclear factor kappa light‐chain enhancer of activated B cells (NF‐κB)‐mediated inflammation. TGR5 activation suppresses the phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B‐cells inhibitor, alpha (IκBα), the translocation of p65, NF‐κB DNA‐binding activity, and its transcription activity. Furthermore, TGR5 activation enhances the interaction of IκBα and β‐arrestin2. Suppression of NF‐κB transcription activity and its target gene expression by TGR5 agonist are specifically abolished by the expression of anti‐β‐arrestin2 small interfering RNA. These results show that TGR5 suppresses the NF‐κB pathway by mediation of the interaction between IκBα and β‐arrestin2. In a lipopolysaccharide (LPS)‐induced inflammation model, TGR5−/− mice show more severe liver necroses and inflammation, compared with wild‐type (WT) mice. Activation of TGR5 by its agonist ligand inhibits the expression of inflammatory mediators in response to NF‐κB activation induced by LPS in WT, but not TGR5−/−, mouse liver. Conclusion: These findings identify TGR5 as a negative mediator of inflammation that may serve as an attractive therapeutic tool for immune and inflammatory liver diseases. (HEPATOLOGY 2011;)


Hepatology | 2009

Farnesoid X receptor alleviates age‐related proliferation defects in regenerating mouse livers by activating forkhead box m1b transcription

Wei-Dong Chen; Yan-Dong Wang; Lisheng Zhang; Steven Shiah; Meihua Wang; Fan Yang; Donna Yu; Barry M. Forman; Wendong Huang

Elucidating the mechanism of liver regeneration could lead to life‐saving therapy for a large number of patients, especially elderly patients, after segmental liver transplantation or resection of liver tumors. The forkhead box m1b (Foxm1b) transcription factor is required for normal liver regeneration. Here we report that Foxm1b is the first direct farnesoid X receptor (FXR) target gene known to be involved in cell cycle regulation and that aging regenerating livers have delayed activation of FXR, which results in defective induction of Foxm1b and thereby contributes to defective liver regeneration. An inverted repeat 0 (IR‐0) FXR response element, acting as an enhancer in intron 3 of the Foxm1b gene, was identified by a combination of transcriptional reporter, electrophoretic mobility shift, and chromatin immunoprecipitation assays. Diminished FXR binding to the IR‐0 element was found in aging regenerating livers. FXR activation by a novel ligand in aging livers induced Foxm1b expression and elevated hepatocyte DNA replication to about 70% of the levels found in young regenerating livers, which were specifically suppressed by hepatic expression of anti‐Foxm1b short hairpin RNA. Conclusion: Our results have revealed Foxm1b as the first known direct FXR target gene involved in cell cycle regulation and have demonstrated that defective activation of FXR could be an intrinsic defect in aging regenerating livers. Activation of FXR alone is largely able to alleviate age‐related liver regeneration defects. These findings highlight FXR as a potential target of drug design for promoting liver regeneration in older subjects. (HEPATOLOGY 2010.)


Hepatology | 2016

Vertical sleeve gastrectomy activates GPBAR-1/TGR5 to sustain weight loss, improve fatty liver, and remit insulin resistance in mice.

Lili Ding; Kyle M. Sousa; Lihua Jin; Bingning Dong; Byung-Wook Kim; Ricardo Ramirez; Zhenzhou Xiao; Ying Gu; Qiaoling Yang; Jie Wang; Donna Yu; Alessio Pigazzi; Dustin E. Schones; Li Yang; David D. Moore; Zhengtao Wang; Wendong Huang

Vertical sleeve gastrectomy (VSG) is one of the most commonly performed clinical bariatric surgeries used for the remission of obesity and diabetes. However, the precise molecular mechanism by which VSG exerts its beneficial effects remains elusive. We report that the membrane‐bound G protein‐coupled bile acid receptor, GPBAR‐1 (also known as TGR5), is required to mediate the effects of anti‐obesity, anti‐hyperglycemia, and improvements of fatty liver of VSG in mice. In the absence of TGR5, the beneficial metabolic effects of VSG in mice are lost. Moreover, we found that the expression of TGR5 increased significantly after VSG, and VSG alters both BA levels and composition in mice, resulting in enhancement of TGR5 signaling in the ileum and brown adipose tissues, concomitant with improved glucose control and increased energy expenditure. Conclusion: Our study elucidates a novel underlying mechanism by which VSG achieves its postoperative therapeutic effects through enhanced TGR5 signaling. (Hepatology 2016;64:760‐773)


PLOS ONE | 2014

GPBAR1/TGR5 Mediates Bile Acid-Induced Cytokine Expression in Murine Kupffer Cells

Guiyu Lou; Xiaoxiao Ma; Xianghui Fu; Zhipeng Meng; Wenyu Zhang; Yan-Dong Wang; Carl Van Ness; Donna Yu; Rongzhen Xu; Wendong Huang

GPBAR1/TGR5 is a novel plasma membrane-bound G protein–coupled bile acid (BA) receptor. BAs are known to induce the expression of inflammatory cytokines in the liver with unknown mechanism. Here we show that without other external stimuli, TGR5 activation alone induced the expression of interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) in murine macrophage cell line RAW264.7 or murine Kupffer cells. The TGR5-mediated increase of pro-inflammatory cytokine expression was suppressed by JNK inhibition. Moreover, the induced pro-inflammatory cytokine expression in mouse liver by 1% cholic acid (CA) diet was blunted in JNK−/− mice. TGR5 activation by its ligands enhanced the phosphorylation levels, DNA-binding and trans-activities of c-Jun and ATF2 transcription factors. Finally, the induced pro-inflammatory cytokine expression in Kupffer cells by TGR5 activation correlated with the suppression of Cholesterol 7α-hydroxylase (Cyp7a1) expression in murine hepatocytes. These results suggest that TGR5 mediates the BA-induced pro-inflammatory cytokine production in murine Kupffer cells through JNK-dependent pathway. This novel role of TGR5 may correlate to the suppression of Cyp7a1 expression in hepatocytes and contribute to the delicate BA feedback regulation.


Steroids | 2012

An improved synthesis of 6α-ethylchenodeoxycholic acid (6ECDCA), a potent and selective agonist for the Farnesoid X Receptor (FXR).

Donna Yu; Daniell L. Mattern; Barry M. Forman

The active, potent, and selective Farnesoid X Receptor (FXR) agonist 6α-ethylchenodeoxycholic acid (6ECDCA) has been synthesized in improved yield compared to the published methodologies. The synthesis employed selective oxidation of one of the two hydroxyls of the readily-available starting material chenodeoxycholic acid (CDCA) as a key step. After protection of the remaining hydroxyl, LDA/HMPA/EtI/PPTS provided an efficient deprotonation/ethylation/deprotection sequence. The two synthetic improvements that allow a productive yield are the use of PCC in the oxidation step, and the use of HMPA/ethyl iodide in the stereoselective alkylation step. This synthesis offers an economical and efficient strategy which provides a simple and cost-effective procedure for potential large-scale production of this promising FXR agonist, which is a research tool and potential drug substance of current interest.


Molecular Endocrinology | 2012

FXR Protects Lung from Lipopolysaccharide-Induced Acute Injury

Lisheng Zhang; Tao Li; Donna Yu; Barry M. Forman; Wendong Huang

Acute lung injury and its more severe form, acute respiratory distress syndrome, are characterized by an acute inflammatory response in the airspaces and lung parenchyma. The nuclear receptor farnesoid X receptor (FXR) is expressed in pulmonary artery endothelial cells. Here, we report a protective role of FXR in a lipopolysaccharide-induced mouse model of acute lung injury. Upon intratracheal injection of lipopolysaccharide, FXR-/- mice showed higher lung endothelial permeability, released more bronchoalveolar lavage cells to the alveoli, and developed acute pneumonia. Cell adhesion molecules were expressed at higher levels in FXR-/- mice as compared with control mice. Furthermore, lung regeneration was much slower in FXR-/- mice. In vitro experiments showed that FXR activation blocked TNFα-induced expression of P-selectin but stimulated proliferation of lung microvascular endothelial cells through up-regulation of Foxm1b. In addition, expression of a constitutively active FXR repressed the expression of proinflammatory genes and improved lung permeability and lung regeneration in FXR-/- mice. This study demonstrates a critical role of FXR in suppressing the inflammatory response in lung and promoting lung repair after injury.


Hepatology | 2013

Deficiency of G-protein-coupled bile acid receptor Gpbar1 (TGR5) enhances chemically induced liver carcinogenesis.

Wei-Dong Chen; Donna Yu; Barry M. Forman; Wendong Huang; Yan-Dong Wang

Gpbar1 (TGR5), a membrane‐bound bile acid receptor, is well known for its roles in regulation of energy homeostasis and glucose metabolism. TGR5 activation also inhibits nuclear factor κB (NF‐κB)‐mediated inflammation. Here we show that TGR5 deficiency enhances chemically induced liver carcinogenesis, and that TGR5 is a negative regulator of signal transducer and activator of transcription 3 (STAT3) signaling. Mice lacking TGR5 were much more susceptible to diethylnitrosamine (DEN)‐induced acute liver injury and liver carcinogenesis than wildtype (WT) mice. Consistent with the increasing incidence of liver cancer in TGR5−/− mice, hepatocyte death, compensatory proliferation, and gene expression of certain inflammatory cytokines and matrix metalloproteinases were more sensitive to DEN induction in the absence of TGR5 signaling. In vitro, TGR5 activation greatly inhibited proliferation and migration of human liver cancer cells. We then found that TGR5 activation strongly suppressed STAT3 signaling in vitro and in vivo. Furthermore, we observed that TGR5 antagonizes the STAT3 pathway through suppressing STAT3 phosphorylation, its transcription activity, and DNA binding activity, which suggests that TGR5 antagonizes liver tumorigenesis at least in part by inhibiting STAT3 signaling. Conclusion: These findings identify TGR5 as a novel liver tumor suppressor that may serve as an attractive therapeutic tool for human liver cancer. (HEPATOLOGY 2013;)


Bioorganic & Medicinal Chemistry | 2014

Identification of Trisubstituted-pyrazol Carboxamide Analogs as Novel and Potent Antagonists of Farnesoid X Receptor

Donna Yu; Wenwei Lin; Barry M. Forman; Taosheng Chen

Farnesoid X receptor (FXR, NRIH4) plays a major role in the control of cholesterol metabolism. This suggests that antagonizing the transcriptional activity of FXR is a potential means to treat cholestasis and related metabolic disorders. Here we describe the synthesis, biological evaluation, and structure-activity relationship (SAR) studies of trisubstituted-pyrazol carboxamides as novel and potent FXR antagonists. One of these novel FXR antagonists, 4j has an IC50 of 7.5 nM in an FXR binding assay and 468.5 nM in a cell-based FXR antagonistic assay. Compound 4j has no detectable FXR agonistic activity or cytotoxicity. Notably, 4j is the most potent FXR antagonist identified to date; it has a promising in vitro profile and could serve as an excellent chemical tool to elucidate the biological function of FXR.


Bioorganic & Medicinal Chemistry | 2013

Development of time resolved fluorescence resonance energy transfer-based assay for FXR antagonist discovery.

Donna Yu; Wenwei Lin; Taosheng Chen; Barry M. Forman

FXR (farnesoid X receptor, NRIH4), a nuclear receptor, plays a major role in the control of cholesterol metabolism. FXR ligands have been investigated in preclinical studies for targeted therapy against metabolic diseases, but have shown limitations. Therefore, there is a need for new agonist or antagonist ligands of FXR, both for potential clinical applications, as well as to further elucidate its biological functions. Here we describe the use of the X-ray crystal structure of FXR complexed with the potent small molecule agonist GW4064 to design and synthesize a novel fluorescent, high-affinity probe (DY246) for time resolved fluorescence resonance energy transfer (TR-FRET) assays. We then used the TR-FRET assay for high throughput screening of a library of over 5000 bioactive compounds. From this library, we identified 13 compounds that act as putative FXR transcriptional antagonists.

Collaboration


Dive into the Donna Yu's collaboration.

Top Co-Authors

Avatar

Barry M. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wendong Huang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yan-Dong Wang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wei-Dong Chen

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jia Su

Beijing University of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongzhi Li

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisheng Zhang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Meihua Wang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Taosheng Chen

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge