Network


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

Hotspot


Dive into the research topics where Jinha Yu is active.

Publication


Featured researches published by Jinha Yu.


Journal of the National Cancer Institute | 2014

Overactivated Neddylation Pathway as a Therapeutic Target in Lung Cancer

Lihui Li; Mingsong Wang; Guangyang Yu; Ping Chen; Hui Li; Dongping Wei; Ji Zhu; Li Xie; Huixun Jia; Jie-Yi Shi; Chunjie Li; Wantong Yao; Yanchun Wang; Qiang Gao; Lak Shin Jeong; Hyuk Woo Lee; Jinha Yu; Fengqing Hu; Ju Mei; Ping Wang; Yiwei Chu; Hui Qi; Meng Yang; Ziming Dong; Yi Sun; Robert M. Hoffman; Lijun Jia

BACKGROUND A number of oncoproteins and tumor suppressors are known to be neddylated, but whether the neddylation pathway is entirely activated in human cancer remains unexplored. METHODS NEDD8-activating enzyme (NAE) (E1) and NEDD8-conjugating enzyme (E2) expression and global-protein neddylation were examined by immunohistochemistry, immunoblotting, and real-time polymerase chain reaction analysis. Cell proliferation, clonogenic survival, migration, and motility in vitro, as well as tumor formation and metastasis in vivo, were determined upon neddylation inhibition by MLN4924, an investigational NEDD8-activating enzyme inhibitor. Survival was analyzed with Kaplan-Meier methods and compared by the log-rank test. All statistical tests were two-sided. RESULTS The entire neddylation pathway, including NEDD8-activating enzyme E1, NEDD8-conjugating enzyme E2, and global-protein neddylation, is overactivated in both lung adenocarcinoma and squamous-cell carcinoma. Compared with lung adenocarcinoma patients with low expression, those with high expression had worse overall survival (NEDD8-activating enzyme E1 subunit 1 [NAE1]: hazard ratio [HR] = 2.07, 95% confidence interval [CI] = 0.95 to 4.52, P = .07; ubiquitin-conjugating enzyme E2M (UBC12): HR = 13.26, 95% CI = 1.77 to 99.35, P = .01; global protein neddylation: HR = 3.74, 95% CI = 1.65 to 8.47, P = .002). Moreover, inhibition of neddylation by the NAE inhibitor MLN4924 statistically significantly suppressed proliferation, survival, migration, and motility of lung cancer cells in vitro and tumor formation and metastasis in vivo. At the molecular level, MLN4924 inactivated Cullin-RING E3 ligases, led to accumulation of tumor-suppressive Cullin-RING E3 ligase substrates and induced phorbol-12-myristate-13-acetate-induced protein 1 (NOXA)-dependent apoptosis or cellular senescence. CONCLUSIONS Our study highlights the overactivated neddylation pathway in lung cancer development and as a promising therapeutic target.


Clinical Cancer Research | 2016

Neddylation Inhibition Activates the Extrinsic Apoptosis Pathway through ATF4-CHOP-DR5 Axis in Human Esophageal Cancer Cells.

Ping Chen; Tao Hu; Yupei Liang; Pei Li; Xiaoyu Chen; Jingyang Zhang; Yangcheng Ma; Qianyun Hao; JinWu Wang; Ping Zhang; Yanmei Zhang; Hu Zhao; Shengli Yang; Jinha Yu; Lak Shin Jeong; Hui Qi; Meng Yang; Robert M. Hoffman; Ziming Dong; Lijun Jia

Purpose: Targeting the protein neddylation pathway has become an attractive anticancer strategy; however, the role of death receptor–mediated extrinsic apoptosis during treatment remained to be determined. Experimental Design: The activation of extrinsic apoptosis and its role in MLN4924 treatment of human esophageal squamous cell carcinoma (ESCC) were evaluated both in vitro and in vivo. The expression of the components of extrinsic apoptotic pathway was determined by immunoblotting analysis and downregulated by siRNA silencing for mechanistic studies. Results: Pharmaceutical or genetic inactivation of neddylation pathway induced death receptor 5 (DR5)–mediated apoptosis and led to the suppression of ESCC in murine models. Mechanistically, neddylation inhibition stabilized activating transcription factor 4 (ATF4), a Cullin-Ring E3 ubiquitin ligases (CRL) substrate. Transcription factor CHOP was subsequently transactivated by ATF4 and further induced the expression of DR5 to activate caspase-8 and induce extrinsic apoptosis. Moreover, the entire neddylation pathway was hyperactivated in ESCC and was negatively associated with patient overall survival. Conclusions: Our findings highlight a critical role of ATF4–CHOP–DR5 axis-mediated extrinsic apoptosis in neddylation-targeted cancer therapy and support the clinical investigation of neddylation inhibitors (e.g., MLN4924) for the treatment of ESCC, a currently treatment-resistant disease with neddylation hyperactivation. Clin Cancer Res; 22(16); 4145–57. ©2016 AACR.


Biochemical and Biophysical Research Communications | 2013

Neddylation pathway regulates the proliferation and survival of macrophages

Lihui Li; Binghong Liu; Tiangeng Dong; Hyuk Woo Lee; Jinha Yu; Yijie Zheng; Haifeng Gao; Yan Zhang; Yiwei Chu; Guangwei Liu; Weixin Niu; Shimin Zheng; Lak Shin Jeong; Lijun Jia

Neddylation is a new type of protein post-translational modification which adds the ubiquitin-like molecule Nedd8 to target proteins. The well-identified targets of neddylation are cullins, which serve as essential components of Cullin-RING E3 ligases (CRL). It is reported that inhibition of neddylation repressed NF-κB-mediated proinflammatory cytokine production in macrophages. However, the role of neddylation in the proliferation and survival of macrophages has not been well defined. Here we report that partial inactivation of the neddylation pathway by a specific Nedd8-activating enzyme E1 (NAE) inhibitor MLN4924 reduced LPS-induced production of the proinflammatory cytokines TNF-α and IL-6 without obvious impairment of cell viability. However, persistent and severe inactivation of neddylation by MLN4924 significantly inhibited cell proliferation by inducing G2 phase cell-cycle arrest and further triggered cell death by inducing apoptosis in RAW264.7 macrophages. Mechanistic analysis revealed that inactivation of neddylation blocked cullin neddylation, inhibited CRL E3 ligase activity, and thus led to the accumulation of CRL substrates, resulting in cell-cycle arrest, DNA damage response and apoptosis. The findings revealed that neddylation serves as an important signaling pathway regulating the proliferation and survival of macrophages.


Journal of Medicinal Chemistry | 2015

Selenoacyclovir and Selenoganciclovir: Discovery of a New Template for Antiviral Agents

Pramod K. Sahu; Tamima Umme; Jinha Yu; Akshata Nayak; Gyudong Kim; Minsoo Noh; Jae-Young Lee; Dae-Duk Kim; Lak Shin Jeong

On the basis of the potent antiviral activity of acyclovir and ganciclovir, selenoacyclovir (2a) and selenoganciclovir (2b) were designed based on bioisoteric rationale and synthesized via the diselenide 7 as the key intermediate. Compound 2a exhibited potent anti-HSV-1 and -2 activities while 2b exerted moderate anti-HCMV activity, indicating that these nucleosides can serve as a novel template for the development of new antiviral agents.


Biomolecules & Therapeutics | 2015

A cannabinoid receptor agonist N-arachidonoyl dopamine inhibits adipocyte differentiation in human mesenchymal stem cells.

Seyeon Ahn; Sodam Yi; Won Jong Seo; Myeong Jung Lee; Young Keun Song; Seung Yong Baek; Jinha Yu; Soo Hyun Hong; Jin-Young Lee; Dong Wook Shin; Lak Shin Jeong; Minsoo Noh

Endocannabinoids can affect multiple cellular targets, such as cannabinoid (CB) receptors, transient receptor potential cation channel, subfamily V, member 1 (TRPV1) and peroxisome proliferator-activated receptor γ (PPARγ). The stimuli to induce adipocyte differentiation in hBM-MSCs increase the gene transcription of the CB1 receptor, TRPV1 and PPARγ. In this study, the effects of three endocannabinoids, N-arachidonoyl ethanolamine (AEA), N-arachidonoyl dopamine (NADA) and 2-arachidonoyl glycerol (2-AG), on adipogenesis in hBM-MSCs were evaluated. The adipocyte differentiation was promoted by AEA whereas inhibited by NADA. No change was observed by the treatment of non-cytotoxic concentrations of 2-AG. The difference between AEA and NADA in the regulation of adipogenesis is associated with their effects on PPARγ transactivation. AEA can directly activate PPARγ. The effect of AEA on PPARγ in hBM-MSCs may prevail over that on the CB1 receptor mediated signal transduction, giving rise to the AEA-induced promotion of adipogenesis. In contrast, NADA had no effect on the PPARγ activity in the PPARγ transactivation assay. The inhibitory effect of NADA on adipogenesis in hBM-MSCs was reversed not by capsazepine, a TRPV1 antagonist, but by rimonabant, a CB1 antagonist/inverse agonist. Rimonabant by itself promoted adipogenesis in hBM-MSCs, which may be interpreted as the result of the inverse agonism of the CB1 receptor. This result suggests that the constantly active CB1 receptor may contribute to suppress the adipocyte differentiation of hBM-MSCs. Therefore, the selective CB1 agonists that are unable to affect cellular PPARγ activity inhibit adipogenesis in hBM-MSCs.


Oncotarget | 2016

Radiosensitization by the investigational NEDD8-activating enzyme inhibitor MLN4924 (pevonedistat) in hormone-resistant prostate cancer cells

Xiaofang Wang; Wenjuan Zhang; Zi Yan; Yupei Liang; Lihui Li; Xiaoli Yu; Yan Feng; Shen Fu; Yanmei Zhang; Hu Zhao; Jinha Yu; Lak Shin Jeong; Xiaomao Guo; Lijun Jia

Salvage radiotherapy (SRT) is the first-line treatment for prostate cancer patients with biochemical recurrence following radical prostatectomy, and new specific radiosensitizers are in urgent need to enhance SRT effect. MLN4924 (also known as Pevonedistat), a specific inhibitor of NEDD8-activating enzyme, has recently entered phase I/II clinical trials in several malignancies. By inhibiting cullin neddylation, MLN4924 inactivates Cullin-RING ligases (CRL), which have been validated as an attractive radiosensitizing target. In our study, we demonstrate that MLN4924 can be used as a potent radiosensitizer in hormone-resistant prostate cancer cells. We found that MLN4924 inhibited cullin neddylation and sensitized prostate cancer cells to irradiation (IR). Mechanistically, MLN4924 enhanced IR-induced G2 cell-cycle arrest, by inducing accumulation of WEE1/p21/p27, three well-known CRL substrates. Importantly, siRNA knockdown of WEE1/p21/p27 partially abrogated MLN4924-induced G2 cell-cycle arrest, indicating a causal role of WEE1/p21/p27 in MLN4924-induced radiosensitization. Further mechanistic studies revealed that induction of DNA damage and apoptosis also contributed to MLN4924 radiosensitization in hormone-resistant prostate cancer cells. Our findings lay the foundation for future application of MLN4924 as a potential radiosensitizer in hormone refractory prostate cancer (HRPC).


Archives of Pharmacal Research | 2015

Synthesis and biological evaluation of 2′-substituted-4′-selenoribofuranosyl pyrimidines as antitumor agents

Varughese Alexander; Jayoung Song; Jinha Yu; Jung Hee Choi; Jin-Hee Kim; Sang Kook Lee; Won Jun Choi; Lak Shin Jeong

The 2′-substituted-4′-selenoribofuranosyl pyrimidines 3a–3j were synthesized from D-ribose and assayed for anticancer activity. The 2′-azido and 2′-fluoro groups with a ribo configuration were introduced by the regioselective opening of the O2,2′-anhydronucleosides with sodium azide and (HF)x-pyridine, respectively. Among the compounds tested, only 2′-fluoro derivative 3j was found to exhibit significant anticancer activity, but was much less potent than the corresponding 2′-arabino analogue 2c. This study will provide medicinal chemists with the insight into the identification of structural requirements for the anticancer activity for the developments of biologically active nucleosides.Graphical Abstract


Biochemical and Biophysical Research Communications | 2017

Targeting neddylation pathway with MLN4924 (Pevonedistat) induces NOXA-dependent apoptosis in renal cell carcinoma

Jiyou Wang; Shiwen Wang; Wenjuan Zhang; Xiaofang Wang; Xiaojun Liu; Liang Liu; Lihui Li; Yupei Liang; Jinha Yu; Lak Shin Jeong; Lijun Jia; Hu Zhao; Yanmei Zhang

Inhibition of protein neddylation pathway has emerged an attractive anticancer strategy in preclinical studies by using Nedd8-activating enzyme (NAE) inhibitor MLN4924 (Pevonedistat). Previous studies have reported the antitumor activity of MLN4924 mediated by its efficacy on apoptosis, autophagy and senescence. However, whether MLN4924 has any effect on renal carcinoma cells (RCC) remains unexplored. Here we reported that MLN4924 specifically inhibited protein neddylation pathway, leading to statistically significantly suppress the proliferation, survival and migration of RCC cells by inducing G2 cell-cycle arrest, followed by apoptosis in a MLN4924 dose-dependent manner. Further mechanistic study revealed that MLN4924-induced apoptosis was mediated by substantial up-regulation of pro-apoptotic NOXA. These findings highlighted the anticancer effects of the neddylation inhibitors (e.g. MLN4924) for the treatment of RCC.


Molecules | 2017

Structure-Activity Relationships of Acyclic Selenopurine Nucleosides as Antiviral Agents

Pramod K. Sahu; Tamima Umme; Jinha Yu; Gyudong Kim; Shuhao Qu; Siddhi D. Naik; Lak Shin Jeong

A series of acyclic selenopurine nucleosides 3a–f and 4a–g were synthesized based on the bioisosteric rationale between oxygen and selenium, and then evaluated for antiviral activity. Among the compounds tested, seleno-acyclovir (4a) exhibited the most potent anti-herpes simplex virus (HSV)-1 (EC50 = 1.47 µM) and HSV-2 (EC50 = 6.34 µM) activities without cytotoxicity up to 100 µM, while 2,6-diaminopurine derivatives 4e–g exhibited significant anti-human cytomegalovirus (HCMV) activity, which is slightly more potent than the guanine derivative 4d, indicating that they might act as prodrugs of seleno-ganciclovir (4d).


Cell Proliferation | 2018

The Nedd8-activating enzyme inhibitor MLN4924 (TAK-924/Pevonedistat) induces apoptosis via c-Myc-Noxa axis in head and neck squamous cell carcinoma.

Wenjuan Zhang; Yupei Liang; Lihui Li; Xiaofang Wang; Zi Yan; Changsheng Dong; Mu Sheng Zeng; Qian Zhong; Xue-Kui Liu; Jinha Yu; Shuyang Sun; Xiaojun Liu; Jihui Kang; Hu Zhao; Lak Shin Jeong; Yanmei Zhang; Lijun Jia

The present study aimed to reveal expression status of the neddylation enzymes in HNSCC and to elucidate the anticancer efficacy and the underlying mechanisms of inhibiting neddylation pathway.

Collaboration


Dive into the Jinha Yu's collaboration.

Top Co-Authors

Avatar

Lak Shin Jeong

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Pramod K. Sahu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Gyudong Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Sang Kook Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hyuk Woo Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jayoung Song

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akshata Nayak

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Minsoo Noh

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge