Network


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

Hotspot


Dive into the research topics where Dongdong Lu is active.

Publication


Featured researches published by Dongdong Lu.


Hepatology | 2013

Epigenetic regulation of MicroRNA‐122 by peroxisome proliferator activated receptor‐gamma and hepatitis b virus X protein in hepatocellular carcinoma cells

Kyoung-Sub Song; Chang Han; Jinqiang Zhang; Dongdong Lu; Srikanta Dash; Mark A. Feitelson; Kyu Lim; Tong Wu

MicroRNA‐122 (miR‐122), a pivotal liver‐specific miRNA, has been implicated in several liver diseases including hepatocellular carcinoma (HCC) and hepatitis C and B viral infection. This study aimed to explore epigenetic regulation of miR‐122 in human HCC cells and to examine the effect of hepatitis C virus (HCV) and hepatitis B virus (HBV). We performed microRNA microarray analysis and identified miR‐122 as the most up‐regulated miRNA (6‐fold) in human HCC cells treated with 5′aza‐2′deoxycytidine (5‐Aza‐CdR, DNA methylation inhibitor) and 4‐phenylbutyric acid (PBA, histone deacetylation inhibitor). Real‐time polymerase chain reaction (PCR) analysis verified significant up‐regulation of miR‐122 by 5′aza and PBA in HCC cells, and to a lesser extent in primary hepatocytes. Peroxisome proliferator activated receptor‐gamma (PPARγ) and retinoid X receptor alpha (RXRα) complex was found to be associated with the DR1 and DR2 consensus site in the miR‐122 gene promoter which enhanced miR‐122 gene transcription. 5‐Aza‐CdR and PBA treatment increased the association of PPARγ/RXRα, but decreased the association of its corepressors (N‐CoR and SMRT), with the miR‐122 DR1 and DR2 motifs. The aforementioned DNA‐protein complex also contains SUV39H1, an H3K9 histone methyl transferase, which down‐regulates miR‐122 expression. Conclusions: These findings establish a novel role of the PPARγ binding complex for epigenetic regulation of miR‐122 in human HCC cells. Moreover, we show that hepatitis B virus X protein binds PPARγ and inhibits the transcription of miR‐122, whereas hepatitis C viral particles exhibited no significant effect; these findings provide mechanistic insight into reduction of miR‐122 in patients with HBV but not with HCV infection. (Hepatology 2013;58:1681–1692)


Oncogene | 2012

Microsomal prostaglandin E synthase-1 promotes hepatocarcinogenesis through activation of a novel EGR1/β-catenin signaling axis

Dongdong Lu; Chang Han; Tong Wu

Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme that couples with cyclooxygenase-2 (COX-2) for the production of PGE2. Although COX-2 is known to mediate the growth and progression of several human cancers including hepatocellular carcinoma (HCC), the role of mPGES-1 in hepatocarcinogenesis is not well established. This study provides novel evidence for a key role of mPGES-1 in HCC growth and progression. Forced overexpression of mPGES-1 in two HCC cell lines (Hep3B and Huh7) increased tumor cell growth, clonogenic formation, migration and invasion, whereas knockdown of mPGES-1 inhibited these parameters, in vitro. In a mouse tumor xenograft model, mPGES-1-overexpressed cells formed palpable tumors at earlier time points and developed larger tumors when compared with the control (P<0.01); in contrast, mPGES-1 knockdown delayed tumor development and reduced tumor size (P<0.01). Mechanistically, mPGES-1-induced HCC cell proliferation, invasion and migration involve PGE2 production and activation of early growth response 1 (EGR1) and β-catenin. Specifically, mPGES-1-derived PGE2 induces the formation of EGR1-β-catenin complex, which interacts with T-cell factor 4/lymphoid enhancer factor 1 transcription factors and activates the expression of β-catenin downstream genes. Our findings depict a novel crosstalk between mPGES-1/PGE2 and EGR1/β-catenin signaling that is critical for hepatocarcinogenesis.


Oncogene | 2014

15-PGDH inhibits hepatocellular carcinoma growth through 15-keto-PGE2/PPARγ-mediated activation of p21WAF1/Cip1.

Dongdong Lu; Chang Han; Tong Wu

15-Hydroxyprostaglandin dehydrogenase (15-PGDH) is a key enzyme in prostaglandin (PG) metabolism. This study provides important evidence for inhibition of hepatocellular carcinoma (HCC) growth by 15-PGDH through the 15-keto-prostaglandin E2 (15-keto-PGE2)/peroxisome proliferator-activated receptor-γ (PPARγ)/p21WAF1/Cip1 signaling pathway. Forced overexpression of 15-PGDH inhibited HCC cell growth in vitro, whereas knockdown of 15-PGDH enhanced tumor growth parameters. In a tumor xenograft model in severe combined immunodeficiency mice, inoculation of human HCC cells (Huh7) with overexpression of 15-PGDH led to significant inhibition of tumor growth, whereas knockdown of 15-PGDH enhanced tumor growth. In a separate tumor xenograft model in which mouse HCC cells (Hepa1-6) were inoculated into syngeneic C57BL/6 mice, intratumoral injection of adenovirus vector expressing 15-PGDH (pAd-15-PGDH) significantly inhibited xenograft tumor growth. The antitumor effect of 15-PGDH is mediated through its enzymatic product, 15-keto-PGE2, which serves as an endogenous PPARγ ligand. Activation of PPARγ by 15-PGDH-derived 15-keto-PGE2 enhanced the association of PPARγ with the p21WAF1/Cip1 promoter and increased p21 expression and association with cyclin-dependent kinase 2 (CDK2), CDK4 and proliferating cell nuclear antigen. Depletion of p21 by short hairpin RNA reversed 15-PGDH-induced inhibition of HCC cell growth; overexpression of p21 prevented 15-PGDH knockdown-induced tumor cell growth. These results show a key 15-PGDH/15-keto-PGE2-mediated activation of PPARγ and p21WAF1/Cip1 signaling cascade that regulates hepatocarcinogenesis and tumor progression.


Journal of Biological Chemistry | 2013

15-Hydroxyprostaglandin Dehydrogenase-derived 15-Keto-prostaglandin E2 Inhibits Cholangiocarcinoma Cell Growth through Interaction with Peroxisome Proliferator-activated Receptor-γ, SMAD2/3, and TAP63 Proteins

Dongdong Lu; Chang Han; Tong Wu

Background: 15-PGDH catalyzes PGE2 oxidation to form 15-keto-PGE2. Results: 15-PGDH-derived 15-keto-PGE2 is a PPAR-γ ligand that causes Smad2/3 association with TGFBRI/SARA and induces formation of pSmad2/3-TAP63-p53 ternary complex. Conclusion: 15-PGDH-mediated 15-keto-PGE2 signaling cascade interacts with PPAR-γ, Smad2/3, and TAP63. Significance: Induction of 15-PGDH expression or administration of 15-keto-PGE2 may represent a promising anti-cancer therapeutic strategy. Prostaglandin E2 (PGE2) is a potent lipid mediator that plays a key role in inflammation and carcinogenesis. NAD+-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH) catalyzes the oxidation of the 15(S)-hydroxyl group of PGE2, which leads to PGE2 biotransformation. In this study, we showed that the 15-PGDH-derived 15-keto-PGE2 is an endogenous peroxisome proliferator-activated receptor-γ (PPAR-γ) ligand that causes PPAR-γ dissociation from Smad2/3, allowing Smad2/3 association with the TGF-β receptor I and Smad anchor for receptor activation and subsequent Smad2/3 phosphorylation and transcription activation in human cholangiocarcinoma cells. The 15-PGDH/15-keto-PGE2-induced Smad2/3 phosphorylation resulted in the formation of the pSmad2/3-TAP63-p53 ternary complex and their binding to the TAP63 promoter, inducing TAP63 autotranscription. The role of TAP63 in 15-PGDH/15-keto-PGE2-induced inhibition of tumor growth was further supported by the observation that knockdown of TAP63 prevented 15-PGDH-induced inhibition of tumor cell proliferation, colony formation, and migration. These findings disclose a novel 15-PGDH-mediated 15-keto-PGE2 signaling cascade that interacts with PPAR-γ, Smad2/3, and TAP63.


Cancer Research | 2012

Abstract 1179: 15-PGDH inhibits cholangiocarcinogenesis through the induction of TAp63

Dongdong Lu; Chang Han; Tong Wu

NAD+-linked 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is responsible for the catabolism of prostaglandins in a variety of cell types such as inflammation an dtumorigenesis. In this study we showed that forced overexpression of 15-PGDH in human cholangiocarcinoma cells (CCLP1) suppressed tumor cell growth, invasion and clonogenic formation capacity. In contrast, 15-PGDH knockdown in CCLP1 cells enhanced tumor cell growth, invasion and clonogenic formation. Flow cytometry analysis revealed that 15-PGDH overexpression induced cell cycle arrest at G1/S checkpoint. When implanted into SCID mice, 15-PGDH overexpressed CCLP1 cells formed smaller xenograft tumors, whereas 15-PGDH-depleted cells formed larger xenograft tumors when compared with each of the corresponding controls (p Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1179. doi:1538-7445.AM2012-1179


Cancer Research | 2011

Abstract 247: A novel mPGES-1/EGR1/beta-catenin signaling axis in hepatocarcinogenesis

Dongdong Lu; Chang Han; Tong Wu

Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme that couples with cyclooxygenase-2 (COX-2) for the production of PGE 2 . Although COX-2-derived PGE 2 is known to participate in the development and progression of several human cancers including hepatocellular carcinoma (HCC), the role of mPGES-1 in carcinogenesis remains poorly understood. This study provides novel evidence for an important role of mPGES-1 in human HCC. Forced overexpression of mPGES-1 in two HCC cell lines (Hep3B and Huh7) promoted tumor cell growth and increased clonogenic formation capacity, in vitro. Conversely, mPGES-1 knockdown inhibited tumor cell growth and prevented clonogenic formation. Overexpression of mPGES-1 in Hep3B and Huh7 cells also increased cell invasion and migration/repair capacity, whereas knockdown of mPGES-1 reduced these parameters. Flowcytometric analysis revealed that mPGES-1 knockdown caused cell cycle arrest at G1/S checkpoint. In SCID mouse tumor xenograft model, mPGES-1 overexpressed cells formed palpable tumors at earlier time points and developed larger tumor nodules when compared to the control cells (p Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 247. doi:10.1158/1538-7445.AM2011-247


American Journal of Pathology | 2014

miR-185 Inhibits Hepatocellular Carcinoma Growth by Targeting the DNMT1/PTEN/Akt Pathway

Ximena V. Qadir; Chang Han; Dongdong Lu; Jinqiang Zhang; Tong Wu


American Journal of Pathology | 2014

miR-17-92 cluster promotes cholangiocarcinoma growth: evidence for PTEN as downstream target and IL-6/Stat3 as upstream activator.

Hanqing Zhu; Chang Han; Dongdong Lu; Tong Wu


American Journal of Pathology | 2014

Regular articleTumorigenesis and neoplastic progressionmiR-17-92 Cluster Promotes Cholangiocarcinoma Growth: Evidence for PTEN as Downstream Target and IL-6/Stat3 as Upstream Activator

Hanqing Zhu; Chang Han; Dongdong Lu; Tong Wu


Gastroenterology | 2011

Microsomal Prostaglandin E Synthase-1 Inhibits PTEN and Promotes Experimental Cholangiocarcinogenesis and Tumor Progression

Dongdong Lu; Chang Han; Tong Wu

Collaboration


Dive into the Dongdong Lu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyu Lim

Chungnam National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge