Network


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

Hotspot


Dive into the research topics where Rui Ning is active.

Publication


Featured researches published by Rui Ning.


CNS Neuroscience & Therapeutics | 2012

Fluoxetine Induces Hepatic Lipid Accumulation Via Both Promotion of the SREBP1c-Related Lipogenesis and Reduction of Lipolysis in Primary Mouse Hepatocytes

Xuemin Feng; Jing Xiong; Hao Qin; Wei Liu; Ruini Chen; Wei Shang; Rui Ning; Gang Hu; Jian Yang

In this study, we investigated the peripheral mechanisms underlying the metabolic side effects of fluoxetine (FLX) by focusing on hepatic lipid metabolism.


Molecular Carcinogenesis | 2016

Aspafilioside B Induces G2/M Cell Cycle Arrest and Apoptosis by Up-Regulating H-Ras and N-Ras via ERK and p38 MAPK Signaling Pathways in Human Hepatoma HepG2 Cells

Wei Liu; Rui Ning; Ruini Chen; Xue-Feng Huang; Qinsheng Dai; Jinhua Hu; Yu-Wen Wang; Lili Wu; Jing Xiong; Gang Hu; Qinglong Guo; Jian Yang; Hao Wang

We recently establish that aspafilioside B, a steroidal saponin extracted from Asparagus filicinus, is an active cytotoxic component. However, its antitumor activity is till unknown. In this study, the anticancer effect of aspafilioside B against HCC cells and the underlying mechanisms were investigated. Our results showed that aspafilioside B inhibited the growth and proliferation of HCC cell lines. Further study revealed that aspafilioside B could significantly induce G2 phase cell cycle arrest and apoptosis, accompanying the accumulation of reactive oxygen species (ROS), but blocking ROS generation with N‐acetyl‐l‐cysteine (NAC) could not prevent G2/M arrest and apoptosis. Additionally, treatment with aspafilioside B induced phosphorylation of extracellular signal‐regulated kinase (ERK) and p38 MAP kinase. Moreover, both ERK inhibitor PD98059 and p38 inhibitor SB203580 almost abolished the G2/M phase arrest and apoptosis induced by aspafilioside B, and reversed the expression of cell cycle‐ and apoptosis‐related proteins. We also found that aspafilioside B treatment increased both Ras and Raf activation, and transfection of cells with H‐Ras and N‐Ras shRNA almost attenuated aspafilioside B‐induced G2 phase arrest and apoptosis as well as the ERK and p38 activation. Finally, in vivo, aspafilioside B suppressed tumor growth in mouse xenograft models, and the mechanism was the same as in vitro study. Collectively, these findings indicated that aspafilioside B may up‐regulate H‐Ras and N‐Ras, causing c‐Raf phosphorylation, and lead to ERK and p38 activation, which consequently induced the G2 phase arrest and apoptosis. This study provides the evidence that aspafilioside B is a promising therapeutic agent against HCC.


Biochemical Pharmacology | 2012

DEC1 binding to the proximal promoter of CYP3A4 ascribes to the downregulation of CYP3A4 expression by IL-6 in primary human hepatocytes

Zhao Mao; Xiaofei Luan; Gang Cao; Wei Liu; Jing Xiong; Gang Hu; Ruini Chen; Rui Ning; Wei Shang; Jian Yang; Bingfang Yan

In this study, we provided molecular evidences that interleukin-6 (IL-6) contributed to the decreased capacity of oxidative biotransformation in human liver by suppressing the expression of cytochrome P450 3A4 (CYP3A4). After human hepatocytes were treated with IL-6, differentially expressed in chondrocytes 1 (DEC1) expression rapidly increased, and subsequently, the CYP3A4 expression decreased continuously. Furthermore, the repression of CYP3A4 by IL-6 occurred after the increase of DEC1 in primary human hepatocytes. In HepG2 cells, knockdown of DEC1 increased the CYP3A4 expression and its enzymatic activity. In addition, it partially abolished the decreased CYP3A4 expression as well as its enzymatic activity induced by IL-6. Consistent with this, overexpression of DEC1 markedly reduced the CYP3A4 promoter activity and the CYP3A4 expression as well as its enzymatic activity. Using sequential truncation and site directed mutagenesis of CYP3A4 proximal promoter with DEC1 construct, we showed that DEC1 specifically bound to CCCTGC sequence in the proximal promoter of CYP3A4, which was validated by EMSA and ChIP assay. These findings suggest that the repression of CYP3A4 by IL-6 is achieved through increasing the DEC1 expression in human hepatocytes, the increased DEC1 binds to the CCCTGC sequence in the promoter of CYP3A4 to form CCCTGC-DEC1 complex, and the complex downregulates the CYP3A4 expression and its enzymatic activity.


Life Sciences | 2014

Glucose dominates the regulation of carboxylesterases induced by lipopolysaccharide or interleukin-6 in primary mouse hepatocytes.

Jing Xiong; Wei Shang; Lili Wu; Ruini Chen; Wei Liu; Rui Ning; Gang Hu; Jian Yang

AIMS Altered drug disposition has been associated with inflammation and diabetes, leading to the alteration of drug efficacy and toxicity. Carboxylesterases are major hydrolytic enzymes in the liver, catalyzing the hydrolytic biotransformation of numerous therapeutic agents. Therefore, how glucose affects the regulation of carboxylesterases by interleukin-6 (IL-6) and lipopolysaccharide (LPS) were investigated. MAIN METHODS Primary mouse hepatocytes were cultured. Protein levels were measured by Western blot or enzyme linked immunosorbent assay (ELISA), while confocal laser scanning microscope and flow cytometry were used to confirm the activation of pregnane X receptor (PXR). Carboxylesterase activity was evaluated by enzymatic and toxicological assays. KEY FINDINGS Elevated glucose (11 or 25 mM) significantly increased carboxylesterase expression compared to 5.6 mM glucose. Carboxylesterase expression and activity were inhibited by LPS or IL-6 in 25 mM glucose, but stimulated in 5.6 mM glucose. The altered expression of carboxylesterases was not consistent with the activation of nuclear factor kappa B (NFκB) but repeatedly with the expression and activation of pregnane X receptor (PXR). The altered activation of PXR was further evidenced by the differential subcellular translocation and the expression of its target gene multidrug resistance 1 (MDR1). It implies that PXR, instead of inflammatory signaling, mediates the regulation of carboxylesterases by inflammatory mediators in different glucose concentrations. SIGNIFICANCE The findings contribute to clarify the regulation of carboxylesterases by inflammatory mediators, and indicate that carboxylesterase-involved drug metabolism and drug-drug interactions in diabetes should be reevaluated according to the intensity of inflammatory reactions and hyperglycemia.


Biochemical Pharmacology | 2017

Icariin protects against glucocorticoid induced osteoporosis, increases the expression of the bone enhancer DEC1 and modulates the PI3K/Akt/GSK3β/β-catenin integrated signaling pathway

Jinhua Hu; Zhao Mao; Shuangcheng He; Yuanran Zhan; Rui Ning; Wei Liu; Bingfang Yan; Jian Yang

Graphical abstract Figure. No Caption available. Abstract Osteoporosis is a serious public health concern worldwide. Herba epimedii has been used for centuries and even thousands of years to treat osteoporotic conditions. Icariin, a flavonol glycoside, is one of the major active ingredients. In this study, we have shown that icariin protected against glucocorticoid‐induced osteoporotic changes in SaoS‐2 cells and mice. We have also shown that dexamethasone (a glucocorticoid) suppressed and icariin induced DEC1, a structurally distinct helix‐loop‐helix protein. DEC1 overexpression promoted whereas DEC1 knockdown decreased osteogenic activity. Likewise, DEC1 overexpression and knockdown inversely regulated the expression of &bgr;‐catenin and PIK3CA, an essential player in the Wnt/&bgr;‐catenin and PI3K/Akt signaling pathways, respectively. Interestingly, DKK1, an inhibitor of Wnt/&bgr;‐catenin signaling inhibitor, and LY294002, an inhibitor of PI3K/Akt signaling, abolished the induction of DEC1 by icariin. It is established that these two pathways are interconnected by the phosphorylation status of GSK3&bgr;. Dexamethasone decreased but icariin increased GSK3&bgr; phosphorylation. Finally, DEC1 deficient mice developed osteoporotic phenotypes. Taken together, it is concluded that DEC1 likely supports the action of icariin against glucocorticoid induced osteoporosis with an involvement of the PI3K/Akt/GSK3&bgr;/&bgr;‐catenin integrated signaling pathway.


Xenobiotica | 2016

Fluoxetine reduces CES1, CES2, and CYP3A4 expression through decreasing PXR and increasing DEC1 in HepG2 cells.

Wei Shang; Jie Liu; Ruini Chen; Rui Ning; Jing Xiong; Wei Liu; Zhao Mao; Gang Hu; Jian Yang

Abstract 1. This study investigated the mechanisms of the decreases of carboxylesterases (CES) and cytochrome P4503A4 (CYP3A4) and the enzymatic activities induced by fluoxetine (FLX) in HepG2 cells. We found that FLX decreased the carboxylesterase 1 (CES1) and carboxylesterase 2 (CES2) expression and the hydrolytic activity. 2. FLX decreased the pregnane X receptor (PXR) expression which regulated the target genes such as CYP3A4, whereas increased the differentiated embryonic chondrocyte-expressed gene 1 (DEC1) expression. 3. FLX repressed the PXR at transcriptional level. 4. Overexpression of PXR alone increased the expression of CES1, CES2, and CYP3A4 and attenuated the decreases of CES1, CES2, and CYP3A4 induced by FLX. On the contrary, knockdown of PXR alone decreased the expression of CES1, CES2, and CYP3A4 and almost abolished the decreases of CES1, CES2, and CYP3A4 induced by FLX. 5. Knockdown of DEC1 alone increased the expression of PXR and CYP3A4 and almost abolished the decreases of CES1, CES2, and CYP3A4 induced by FLX. 6. Taken together, the decreases of CES and CYP3A4 expression and enzymatic activities induced by FLX are through decreasing PXR and increasing DEC1 in HepG2 cells.


Xenobiotica | 2015

Decreased carboxylesterases expression and hydrolytic activity in type 2 diabetic mice through Akt/mTOR/HIF-1α/Stra13 pathway

Ruini Chen; Yu-Wen Wang; Rui Ning; Jinhua Hu; Wei Liu; Jing Xiong; Lili Wu; Jie Liu; Gang Hu; Jian Yang

Abstract 1. This study investigated the alteration of carboxylesterases in type 2 diabetes. We found that the carboxylesterase 1d (Ces1d) and carboxylesterase 1e (Ces1e) expression and the capacity of hydrolytic activity of liver and intestine decreased, whereas the Akt/mTOR/HIF-1α/ Stra13 (DEC1) signaling was activated in T2D mice. Consistently, high insulin could give rise to the same results in the high-glucose DMEM condition, which mimicked T2D, in primary mouse hepatocytes. 2. Perifosine or rapamycin almost abolished the decrease of the Ces1d and Ces1e expression and the hydrolytic activity induced by the insulin in the primary mouse hepatocytes. 3. The responsiveness of human hepatoma (HepG2) cells to high insulin in high-glucose condition was similar to that of primary mouse hepatocytes in terms of the altered expression of carboxylesterases. 4. The knockdown of HIF-1α or DEC1 with shRNA construct abrogated the decrease of the CES1 and CES2 expression induced by the insulin in high glucose condition in HepG2 cells. 5. Taken together, the decreased carboxylesterases expression and hydrolytic activity in T2D mice are through the Akt/mTOR/HIF-1α/Stra13 (DEC1) pathway.


Xenobiotica | 2016

Gambogic acid potentiates clopidogrel-induced apoptosis and attenuates irinotecan-induced apoptosis through down-regulating human carboxylesterase 1 and -2

Rui Ning; Xiao-Ping Wang; Yunran Zhan; Qi Qi; Xue-Feng Huang; Gang Hu; Qinglong Guo; Wei Liu; Jian Yang

Abstract 1. In this study, we report that gambogic acid (GA), a promising anticancer agent, potentiates clopidogrel-induced apoptosis and attenuates CPT-11-induced apoptosis by down-regulating human carboxylesterase (CES) 1 and -2 via ERK and p38 MAPK pathway activation, which provides a molecular explanation linking the effect of drug combination directly to the decreased capacity of hydrolytic biotransformation. 2. The expression levels of CES1 and CES2 decreased significantly in a concentration- and time-dependent manner in response to GA in Huh7 and HepG2 cells; hydrolytic activity was also reduced. 3. The results showed that pretreatment with GA potentiated clopidogrel-induced apoptosis by down-regulating CES1. Moreover, the GA-mediated repression of CES2 attenuated CPT-11-induced apoptosis. 4. Furthermore, the ERK and p38 MAPK pathways were involved in the GA-mediated down-regulation of CES1 and CES2. 5. Taken together, our data suggest that GA is a potent repressor of CES1 and CES2 and that combination with GA will affect the metabolism of drugs containing ester bonds.


Evidence-based Complementary and Alternative Medicine | 2013

Curcumin Protects against 1-Methyl-4-phenylpyridinium Ion- and Lipopolysaccharide-Induced Cytotoxicities in the Mouse Mesencephalic Astrocyte via Inhibiting the Cytochrome P450 2E1

Haiyan Gui; Ruini Chen; Yan Peng; Jinhua Hu; Zhao Mao; Rui Ning; Wei Shang; Wei Liu; Jing Xiong; Gang Hu; Jian Yang

Curcumin is extracted from the rhizomes of the ginger family plant Curcuma longa L., which has a good protection for liver, kidney, and immune system. However, there is little information about its contribution in protection of astrocytes recently. The present study was undertaken to elucidate the protective effect of curcumin, an herbal antioxidant, on 1-methyl-4-phenylpyridinium ion- (MPP+-) and lipopolysaccharide- (LPS-) induced cytotoxicities, as well as the underlying mechanisms by using primary mouse mesencephalic astrocytes. The results showed that curcumin protected the mesencephalic astrocytes from MPP+- and LPS-induced toxicities along with reducing reactive oxygen species (P < 0.05) and maleic dialdehyde (P < 0.05) sufficiently. Moreover, curcumin significantly inhibited the cytochrome P450 2E1 (CYP2E1) expression (P < 0.01 at mRNA level, P < 0.05 at protein level) and its activity (P < 0.05) sufficiently induced by MPP+ and LPS in the mouse mesencephalic astrocytes. And curcumin as well as diallyl sulphide, a CYP2E1 positive inhibitor, ameliorated MPP+- and LPS-induced mouse mesencephalic astrocytes damage. Accordingly, curcumin protects against MPP+- and LPS-induced cytotoxicities in the mouse mesencephalic astrocyte via inhibiting the CYP2E1 expression and activity.


Frontiers in Pharmacology | 2017

Interleukin-6 Induces DEC1, Promotes DEC1 Interaction with RXRα and Suppresses the Expression of PXR, CAR and Their Target Genes

Rui Ning; Yunran Zhan; Shuangcheng He; Jinhua Hu; Zhu Zhu; Gang Hu; Bingfang Yan; Jian Yang; Wei Liu

Inflammatory burden is a primary cellular event in many liver diseases, and the overall capacity of drug elimination is decreased. PXR (pregnane X receptor) and CAR (constitutive androstane receptor) are two master regulators of genes encoding drug-metabolizing enzymes and transporters. DEC1 (differentiated embryonic chondrocyte-expressed gene 1) is a ligand-independent transcription factor and reportedly is induced by many inflammatory cytokines including IL-6. In this study, we used primary hepatocytes (human and mouse) as well as HepG2 cell line and demonstrated that IL-6 increased DEC1 expression and decreased the expressions of PXR, CAR, and their target genes. Overexpression of DEC1 had similar effect as IL-6 on the expression of these genes, and knockdown of DEC1 reversed their downregulation by IL-6. Interestingly, neither IL-6 nor DEC1 altered the expression of RXRα, a common dimerization partner for many nuclear receptors including PXR and CAR. Instead, DEC1 was found to interact with RXRα and IL-6 enhanced the interaction. These results conclude that DEC1 uses diverse mechanisms of action and supports IL-6 downregulation of drug-elimination genes and their regulators.

Collaboration


Dive into the Rui Ning's collaboration.

Top Co-Authors

Avatar

Jian Yang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Liu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Gang Hu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Ruini Chen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Xiong

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinhua Hu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Shang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhao Mao

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jie Liu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Lili Wu

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge