Network


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

Hotspot


Dive into the research topics where Yahong Zhang is active.

Publication


Featured researches published by Yahong Zhang.


Biochemical and Biophysical Research Communications | 2008

Fas-mediated autophagy requires JNK activation in HeLa cells.

Yahong Zhang; Yingliang Wu; Yan Cheng; Zhenkun Zhao; Shin-ichi Tashiro; Satoshi Onodera; Takashi Ikejima

Fas has been reported to play an important role in apoptosis; however, Fas-mediated autophagy and its mechanism are still unclear. Here, we found that Fas agonistic antibody CH11-induced autophagy in HeLa cells, and inhibition of autophagy by 3-MA increased CH11-induced apoptosis. A Fas antagonistic antibody (UB2) suppressed both CH11-induced autophagy and apoptosis. In addition, the CH11-induced autophagy was blocked by JNK inhibitor (SP600125), but it was not affected by caspase 8 inhibitor (Z-IETD); whereas the CH11-induced apoptosis was increased by SP600125, and it was suppressed by Z-IETD. Further experiments confirmed that JNK was activated by CH11 dose-dependently, and the activation was suppressed when autophagy was blocked by 3-MA. Together, our results suggest that JNK, but not caspase 8, involves in Fas-mediated CH11-induced autophagy in HeLa cells, and this autophagy plays a protective role in CH11-induced cell death.


Archives of Biochemistry and Biophysics | 2009

NF-κb facilitates oridonin-induced apoptosis and autophagy in HT1080 cells through a p53-mediated pathway

Yahong Zhang; Yingliang Wu; Di Wu; Shin-ichi Tashiro; Satoshi Onodera; Takashi Ikejima

In this study, we investigated the molecular mechanisms involving in oridonin-induced apoptosis and autophagy. We found that apoptosis and autophagy were simultaneously induced by oridonin time-dependently in HT1080 cells, and inhibition of autophagy by 3MA decreased oridonin-induced apoptosis, indicating that they act in synergy to mediate cell death. In addition, treatment with oridonin caused an increase in NF-kappaB and p53 activities in a time-dependent manner. Inhibition of NF-kappaB or p53 activation by its specific inhibitor PDTC or pifithrin-alpha respectively, significantly reduced both oridonin-induced apoptosis and autophagy accompanied by the decrease in Beclin 1 and LC3 levels. Further experiments confirmed that oridonin-induced p53 activation was reduced by the NF-kappaB inhibitor whereas the activation of NF-kappaB was not affected by p53 inhibition. Taken together, these results demonstrate that NF-kappaB promotes oridonin-induced apoptotic and autophagic cell death through regulating p53 activation in HT1080 cells.


Biochemical and Biophysical Research Communications | 2009

Involvement of PKC signal pathways in oridonin-induced autophagy in HeLa cells: a protective mechanism against apoptosis.

Yahong Zhang; Yingliang Wu; Shin-ichi Tashiro; Satoshi Onodera; Takashi Ikejima

Our previous studies showed that oridonin could induce both apoptosis and autophagy in HeLa cells, and this autophagy might be a protective mechanism against apoptosis. In this study, the roles of PKC signal pathways in oridonin-induced HeLa cell autophagy and apoptosis were further investigated. We found that inhibition of PKC significantly reduced oridonin-induced autophagy whereas markedly increased apoptosis, while pretreatment with PKC activator caused opposite results. Subsequently, the oridonin-induced autophagy was also suppressed by Raf-1 or JNK inhibition accompanied by the increase of apoptosis, but it was not affected by ERK or p38 inhibition. In addition, oridonin-induced protein levels of Raf-1, JNK and p-JNK were sharply downregulated by PKC inhibitor, and they were enhanced by PKC activator. Taken together, these results demonstrate that PKC enhances oridonin-induced autophagy against apoptosis through regulating its downstream factors Raf-1 and JNK in HeLa cells.


Apoptosis | 2012

3-Nitro-naphthalimide and nitrogen mustard conjugate NNM-25 induces hepatocellular carcinoma apoptosis via PARP-1/p53 pathway

Songqiang Xie; Yahong Zhang; Qian Li; Feng-hua Xu; Jiu-wang Miao; Jin Zhao; Chaojie Wang

Hepatocellular carcinoma (HCC) is one of the main causes of death in cancer. Some naphthalimide derivatives exert high anti-proliferative effects on HCC. In this study, it is confirmed that 3-nitro-naphthalimide and nitrogen mustard conjugate (NNM-25), a novel compound conjugated by NNM-25, displayed more potent therapeutic action on HCC, both in vivo and in vitro, than amonafide, a naphthalimide drug in clinical trials. More importantly, preliminary toxicological evaluation also supported that NNM-25 exhibited less systemic toxicity than amonafide at the therapeutic dose. The antitumor mechanism of conjugates of naphthalimides with nitrogen mustard remains poorly understood up to now. Here, we first reported that apoptosis might be the terminal fate of cancer cells treated with NNM-25. Inhibition of p53 by siRNA resulted in a significant decrease of NNM-25-induced apoptosis, which corroborated that p53 played a vital role in the cell apoptosis triggered by NNM-25. NNM-25 inhibited the PARP-1 activity, AKT phosphorylation, up-regulated the protein expression of p53, Bad, and mTOR as well as down-regulating the protein expression of Bcl-2 and decreasing mitochondrial membrane potential. It also facilitated cytochrome c release from mitochondria to cytoplasm, activated caspase 8, caspase 9, and caspase 3 in HepG2 cells in vitro, as also authenticated in H22 tumor-bearing mice in vivo. Collectively, the conjugation of naphthalimides with nitrogen mustard provides favorable biological activity and thus is a valuable strategy for future drug design in HCC therapy.


Anti-Cancer Drugs | 2013

Antitumor effects and preliminary systemic toxicity of ANISpm in vivo and in vitro.

Ming Li; Qian Li; Yahong Zhang; Zhiyong Tian; Hong-xia Ma; Jin Zhao; Songqiang Xie; Chaojie Wang

Polyamines as a vector to ferry toxic agents have attracted attention, and naphthalimide–polyamine conjugates show potent activity and tumor cell selectivity. The present study was carried out to evaluate the antitumor effects and preliminary systemic toxicity of ANISpm, a novel 3-amino-naphthalimide–spermine conjugate. The polyamine transport system recognition of ANISpm, supported by &agr;-difluoromethylornithine (DFMO)/spermidine (Spd) experiments, is in accordance with its potent cell selectivity between human hepatoma HepG2 cells and normal QSG7701 hepatocyte. The antiproliferative effect is because of ANISpm-induced cell apoptosis, a common characteristic of both naphthalimide and polyamine analogs. Various apoptotic assessment assays have shown that ANISpm can induce apoptosis through the PI3K/Akt signal pathway. The apoptotic signaling cascade involves Akt inactivation, which results in a series of cellular events. The downstream pathway includes Bad dephosphorylation, dissociation of 14-3-3 and Bad, and binding to Bcl-xL, which triggers the disruption of the mitochondrial membrane, release of cytochrome c, and caspases’ cascade activation. Furthermore, the Akt/mTOR signal pathway is also involved in ANISpm-mediated cell-cycle arrest. Additive DFMO or Spd, which only enhances or attenuates ANISpm-mediated cell apoptosis, respectively, does not alter the signal pathway. In addition, preliminary toxicology evaluation showed that ANISpm had no obvious system toxicity at a dose of 2.5 mg/kg, which exerted potent antitumor activity in vivo, especially hematotoxicity. Thus, ANISpm merits further investigation as a potential chemotherapeutic agent against hepatocellular carcinoma.


International Journal of Colorectal Disease | 2012

COX-2-independent induction of apoptosis by celecoxib and polyamine naphthalimide conjugate mediated by polyamine depression in colorectal cancer cell lines

Songqiang Xie; Yahong Zhang; Qian Li; Jianhong Wang; Jinghua Li; Jin Zhao; Chaojie Wang

BackgroundPolyamine metabolism is an intriguing tumor therapeutic target. The present study was designed to assess the synergistic antitumor effects of NPC-16, a novel polyamine naphthalimide conjugate, with celecoxib and to elucidate the mechanism of these effects on human colorectal cancer cells.MethodsCell proliferation was assessed by the MTT assay. Cell apoptosis and mitochondria membrane potential were evaluated by high content screening analysis. Intracellular polyamine content was detected by HPLC. Protein expression was detected by western blot analysis.ResultsThe co-treatment with celecoxib enhanced NPC-16-induced apoptosis in HCT116 (COX-2 no expression), HT29 (COX-2 higher expression) and Caco-2 (COX-2 higher expression) colorectal cancer cells, which was mediated by the elevated NPC-16 uptake via the effect of celecoxib on polyamine metabolism, including the up-regulated spermidine/spermine N1-acetyltransferase (SSAT) activity and reduced intracellular polyamine levels. The presence of celecoxib does not result in obviously different effect on the NPC-16-triggered apoptosis in diverse COX-2 expressed colorectal cell lines, suggesting that COX-2 was not one vital factor in the apoptotic mechanism. Furthermore, this synergistic apoptosis was involved in the PKB/AKT signal pathway, Bcl-2 and caspase family members. Z-VAD-FMK, a cell permeable pan caspase inhibitor, almost completely inhibited celecoxib and NPC-16 co-induced apoptosis, indicating that this apoptosis was caspase dependent.ConclusionsCo-treatment of celecoxib and NPC-16 could induce colorectal cancer cell apoptosis via COX-2-independent and caspase-dependent mechanisms. The combination therapy with these agents might provide a novel therapeutic model for colorectal cancer.


Oncology Reports | 2014

Distinct roles of Akt1 in regulating proliferation, migration and invasion in HepG2 and HCT 116 cells

Liang Chen; Qiao-Hui Kang; Ying Chen; Yahong Zhang; Qian Li; Songqiang Xie; Chaojie Wang

Elucidating the effects of genes involved in tumors may improve therapeutic strategies for human cancer. Recently, several studies discovered that Akt1 plays a dual role in mediating cell proliferation, migration and invasion, depending on the cell type. However, the pathophysiological role of Akt1 in hepatocellular carcinoma (HCC) and colorectal carcinoma cells remains poorly understood. In the present study, we transfected the Akt1-expressing plasmids into the tumor cells that expressed only low levels of Akt1. The migration and invasion abilities were analyzed in 24-well Boyden chambers. The expression of proteins was detected using western blot analysis. Our results demonstrated that overexpression of Akt1 significantly enhanced the proliferation rates and promoted the colony formation in both HepG2 and HCT 116 cells. When treated with wortmannin, the ability to form colonies was significantly attenuated in both cell lines. Of note, enforced expression of Akt1 induced HepG2 cell migration and invasion; by contrast, upregulation of Akt1 expression suppressed the migration and invasion of HCT 116 cells. Subsequent mechanistic investigations revealed that upregulation of Akt1 markedly induced the expression of Bcl-2 and NF-κB in both types of tumor cells. Notably, we observed a similar increase of MMP2, MMP9, HIF1α and VEGF in HCC cells, whereas Akt1 significantly suppressed the expression of these molecules in colorectal carcinoma cells. These data suggest a dual role for Akt1 in tumor cell migration and invasion and highlight the cell type-specific actions of Akt1 kinases in the regulation of cell motility.


Journal of Coordination Chemistry | 2013

Synthesis, crystal structures, and cytotoxicity of 2-benzoylpyridine N(4)-cyclohexylthiosemicarbazone and its zinc(II) and diorganotin(IV) complexes

Min Yang; Yahong Zhang; Ming-Xue Li; Yanli Lu; Nan Zhang

[Zn(L)2] (1) and [(Ph)2Sn(L)(CH3COO)] (2), where HL = 2-benzoylpyridine N(4)-cyclohexylthiosemicarbazone, have been synthesized and characterized. The complexes show different coordination depending on their coordinating preferences. Biological studies carried out in vitro against human leukemia K562 cells show that the diorgantin(IV) complex, 2, has significant cytotoxicity with IC50 = 3.3 ± 0.5 μM.


Synthesis and Reactivity in Inorganic Metal-organic and Nano-metal Chemistry | 2015

Synthesis, Crystal Structures, and Cytotoxicity of Thiophene-2-carbaldehyde N(4)-Cyclohexylthiosemicarbazone and Its Diorganotin(IV) Complex

Yahong Zhang; Nan Zhang; Yanke Li; Yanli Lu; Mingxue Li; Danyun Chen

Thiophene-2-carbaldehyde N(4)-cyclohexylthiosemicarbazone C12H17N3S2 (HL) and diorganotin(IV) complex [Ph2Sn(L)Cl] (1) have been synthesized and characterized by elemental analysis, IR, and single-crystal X-ray diffraction. Complex 1 contains mononuclear neutral molecule composed of one five-coordinated diorganotin(IV) ion, one NS bidentate anionic thiosemicarbazone ligand, one chloridion, and two phenyl group, respectively. In vitro biological studies have indicated that coupling of HL with Ph2Sn(IV) leads to an enhancement of the cytotoxicity of the free ligand. Importantly, complex 1 could distinguish the human liver hepatocellular carcinoma HepG-2 cells from normal hepatocyte QSG7701 cells.


Medicinal Chemistry | 2014

Synthesis and Preliminary Biological Evaluation of Polyamine-aniline Acridines as P-glycoprotein Inhibitors

Jianhong Wang; Pengfei Cheng; Tianwei Luo; Zhaoyi Wang; Yahong Zhang; Jin Zhao

We have synthesized a series of polyamine-based anilinoacridine derivatives. The preliminary biological evaluation indicated that the 9-anilinoacridine-polyamine derivatives had low or insignificant in vitro cytotoxicity against K562 cell line and K562/ADM, the drug-resistant cell line. However, the evaluation for P-gp modulation showed that they held potent P-gp inhibitory ability. Among them, the effect of compound 7c on P-gp was even greater than that of Verapamil, the known P-gp modulator. The results suggest that 9-anilinoacridine-polyamine derivatives can be employed as effective P-gp modulators.

Collaboration


Dive into the Yahong Zhang'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

Takashi Ikejima

Shenyang Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yingliang Wu

Shenyang Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Satoshi Onodera

Showa Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Shin-ichi Tashiro

Showa Pharmaceutical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge