Network


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

Hotspot


Dive into the research topics where Yunyi Hong is active.

Publication


Featured researches published by Yunyi Hong.


Biochemical and Biophysical Research Communications | 2012

SIRT2 activity is required for the survival of C6 glioma cells

Xin He; Hui Nie; Yunyi Hong; Caibin Sheng; Weiliang Xia; Weihai Ying

SIRT2 is a tubulin deacetylase, which can play either detrimental or beneficial roles in cell survival under different conditions. While it has been suggested that reduced SIRT2 expression in human gliomas may contribute to development of gliomas, there has been no study that directly determines the effects of decreased SIRT2 activity on the survival of glioma cells. In this study we applied both pharmacological and molecular approaches to determine the roles of SIRT2 in the survival of glioma cells. Our studies, by conducting such assays as flow cytometry-based Annexin V assay and caspase-3 immunostaining, have indicated that decreased SIRT2 activity leads to apoptosis of C6 glioma cells by caspase-3-dependent pathway. Our experiments have further shown that reduced SIRT2 activity produces necrosis of C6 glioma cells. Moreover, our study applying SIRT2 siRNA has also shown that decreased SIRT2 leads to both necrosis and apoptotic changes of C6 glioma cells. Collectively, our study has provided novel evidence indicating that SIRT2 activity plays a key role in maintaining the survival of glioma cells, and that reduced SIRT2 activity can induce both necrosis and caspase-3-dependent apoptosis of C6 glioma cells. These results have also suggested that inhibition of SIRT2 might become a novel therapeutic strategy for gliomas.


Current Drug Targets | 2012

NAD+ metabolism and NAD(+)-dependent enzymes: promising therapeutic targets for neurological diseases.

Yingxin Ma; Heyu Chen; Xin He; Hui Nie; Yunyi Hong; Caibin Sheng; Qing Wang; Weiliang Xia; Weihai Ying

Numerous studies have indicated that four interacting factors, including oxidative stress, mitochondrial alterations, calcium dyshomeostasis and inflammation, play crucial pathological roles in multiple major neurological diseases, including stroke, Alzheimers disease (AD) and Parkinsons disease (PD). Increasing evidence has also indicated that NAD(+) plays important roles in not only mitochondrial functions and energy metabolism, but also calcium homeostasis and inflammation. The key NAD(+)-consuming enzyme--poly(ADP-ribose) polymerase-1 (PARP-1) and sirtuins--have also been shown to play important roles in cell death and aging, which are two key factors in the pathology of multiple major age-dependent neurological diseases: PARP-1 plays critical roles in both inflammation and oxidative stress-induced cell death; and sirtuins also mediate the process of aging, cell death and inflammation. Thus, it is conceivable that increasing evidence has suggested that NAD(+) metabolism and NAD(+)-dependent enzymes are promising targets for treating a number of neurological illnesses. For examples, the key NAD(+)-dependent enzymes SIRT1 and SIRT2 have been indicated to strongly affect the pathological changes of PD and AD; PARP-1 inhibition can profoundly reduce the brain injury in the animal models of multiple neurological diseases; and administration of either NAD(+) or nicotinamide can also decrease ischemic brain damage. Future studies are necessary to further investigate the roles of NAD+ metabolism and NAD⁺-dependent enzymes in neurological diseases, which may expose novel targets for treating the debilitating illnesses.


Neuroreport | 2014

SIRT2 mediates oxidative stress-induced apoptosis of differentiated PC12 cells.

Hui Nie; Yunyi Hong; Xiaofei Lu; Jie Zhang; Heyu Chen; Yexin Li; Weihai Ying

Sirtuin 2 (SIRT2) is a member of the sirtuin family. Previous studies have suggested that SIRT2 mediates the cell death in models of Parkinson’s disease and Huntington’s disease. However, the role of SIRT2 in oxidative stress-induced cell death has remained unclear. In this study, we investigated the roles of SIRT2 in oxidative stress-induced cell death using differentiated PC12 cells as a cell model. We found that H2O2 induced a significant increase in the SIRT2 level in the cells. Both SIRT2 silencing and the SIRT2 inhibitor AGK2 significantly decreased H2O2-induced apoptosis, partially by inhibiting caspase-3 activation. We further found that silencing of SIRT2 led to decreased reactive oxygen species levels in the H2O2-treated cells. Collectively, our observations have suggested that SIRT2 plays a significant role in oxidative stress-induced cell death.


Current Medicinal Chemistry | 2015

NAD + /NADH Metabolism and NAD + -Dependent Enzymes in Cell Death and Ischemic Brain Injury: Current Advances and Therapeutic Implications

Yingxin Ma; Hui Nie; Heyu Chen; Jiefu Li; Yunyi Hong; Ban Wang; Caixia Wang; Jie Zhang; Wei Cao; Mingchao Zhang; Yongning Xu; Xianting Ding; Shan Kai Yin; Xinkai Qu; Weihai Ying

NAD(+) and NADH play crucial roles in a variety of biological processes including energy metabolism, mitochondrial functions, and gene expression. Multiple studies have indicated that NAD(+) administration can profoundly decrease oxidative cell death as well as ischemic and traumatic brain injury, suggesting NAD(+) metabolism as a promising therapeutic target for cerebral ischemia and head injury. Cumulating evidence has suggested that NAD(+) can produce its protective effects by multiple mechanisms, including preventing mitochondrial alterations, enhancing energy metabolism, preventing virtually all forms of cell death including apoptosis, necrosis and autophagy, inhibiting inflammation, directly increasing antioxidation capacity of cells and tissues, and activating SIRT1. Increasing evidence has also suggested that NADH metabolism is a potential therapeutic target for treating several neurological disorders. A number of studies have further indicated that multiple NAD(+)-dependent enzymes such as sirtuins, polymerase(ADP-ribose) polymerases (PARPs) and CD38 mediate cell death and multiple biological processes. In this article, an overview of the recent findings regarding the roles of NAD(+)/NADH and NAD(+)-dependent enzymes in cell death and ischemic brain injury is provided. These findings have collectively indicated that NAD(+)/NADH and NAD(+)-dependent enzymes play fundamental roles in oxidative stress-induced cell death and ischemic brain injury, which may become promising therapeutic targets for brain ischemia and multiple other neurological disorders.


Neuroscience Letters | 2014

NAD+ treatment prevents rotenone-induced apoptosis and necrosis of differentiated PC12 cells

Yunyi Hong; Hui Nie; Danhong Wu; Xunbin Wei; Xianting Ding; Weihai Ying

Nicotinamide adenine dinucleotide (NAD(+)) plays critical roles in not only energy metabolism and mitochondrial functions, but also calcium homeostasis and immunological functions. It has been reported that NAD(+) administration can reduce ischemic brain damage. However, the mechanisms underlying the protective effects remain unclear. Because mitochondrial impairments play a key role in the cell death in cerebral ischemia, in this study we tested our hypothesis that NAD(+) can decrease mitochondrial damage-induced cell death using differentiated PC12 cells as a cellular model. We found that NAD(+) can decrease both early-stage and late-stage apoptosis, as well as necrosis of rotenone-treated PC12 cells, as assessed by FACS-based Annexin V/AAD assay. We also found that NAD(+) treatment can restore the intracellular NAD(+) levels of the rotenone-treated cells. Moreover, NAD(+) treatment can prevent rotenone-induced mitochondria depolarization. In summary, our study has provided first direct evidence that NAD(+) treatment can prevent rotenone-induced apoptosis and necrosis. Our study has also indicated that NAD(+) treatment can prevent mitochondrial damage-induced cell death, which may at least partially result from its protective effects on rotenone-induced mitochondrial depolarization. Because both mitochondrial damage and apoptosis play key roles in multiple neurological disorders, our study has highlighted the therapeutic potential of NAD(+) for brain ischemia and other neurological diseases.


FEBS Letters | 2016

SIRT2 mediates NADH‐induced increases in Nrf2, GCL, and glutathione by modulating Akt phosphorylation in PC12 cells

Wei Cao; Yunyi Hong; Heyu Chen; Fan Wu; Xunbin Wei; Weihai Ying

SIRT2 plays important roles in multiple biological processes. It is unclear whether SIRT2 affects antioxidant capacity by modulating Nrf2, a key transcription factor for multiple antioxidant genes. By studying NADH‐treated differentiated PC12 cells, we found that NADH induced a significant increase in the nuclear Nrf2, which was prevented by both SIRT2 siRNA and SIRT2 inhibitor, AGK2. SIRT2 siRNA also blocked the NADH‐induced increases in glutamate cysteine ligase (GCL) and glutathione. Moreover, SIRT2 siRNA and AGK2 blocked NADH‐induced Akt phosphorylation, and inhibition of Akt phosphorylation prevented NADH‐induced increases in the nuclear Nrf2 and glutathione. Collectively, our study shows that SIRT2 regulates nuclear Nrf2 levels by modulating Akt phosphorylation, thus modulating the levels of GCL and total glutathione.


Neurochemical Research | 2015

NAD+ treatment can prevent rotenone-induced increases in DNA damage, Bax levels and nuclear translocation of apoptosis-inducing factor in differentiated PC12 cells.

Yunyi Hong; Hui Nie; Xunbin Wei; Shen Fu; Weihai Ying

Nicotinamide adenine dinucleotide (NAD+) plays critical roles in energy metabolism, mitochondrial functions, calcium homeostasis and immunological functions. Our previous studies have found that NAD+ administration can profoundly decrease ischemic brain injury and traumatic brain injury. Our recent study has also provided first direct evidence indicating that NAD+ treatment can decrease cellular apoptosis, while the mechanisms underlying this protective effect remain unclear. In our current study, we determined the effects of NAD+ treatment on several major factors in apoptosis and necrosis, including levels of Bax and nuclear translocation of apoptosis-inducing factor (AIF), as well as levels of DNA double-strand breaks (DSBs) and intracellular ATP in rotenone-treated differentiated PC12 cells. We found that NAD+ treatment can markedly attenuate the rotenone-induced increases in the levels of Bax and nuclear translocation of AIF in the cells. We further found that NAD+ treatment can significantly attenuate the rotenone-induced increase in the levels of DSBs and decrease in the intracellular ATP levels. Collectively, our study has suggested mechanisms underlying the preventive effects of NAD+ on apoptosis, which has highlighted the therapeutic potential of NAD+ for decreasing apoptotic changes in multiple major diseases.


Spermatogenesis | 2015

Antioxidant protects blood-testis barrier against synchrotron radiation X-ray-induced disruption

Tingting Zhang; Tengyuan Liu; Jiaxiang Shao; Caibin Sheng; Yunyi Hong; Weihai Ying; Weiliang Xia

Synchrotron radiation (SR) X-ray has wide biomedical applications including high resolution imaging and brain tumor therapy due to its special properties of high coherence, monochromaticity and high intensity. However, its interaction with biological tissues remains poorly understood. In this study, we used the rat testis as a model to investigate how SR X-ray would induce tissue responses, especially the blood-testis barrier (BTB) because BTB dynamics are critical for spermatogenesis. We irradiated the male gonad with increasing doses of SR X-ray and obtained the testicles 1, 10 and 20 d after the exposures. The testicle weight and seminiferous tubule diameter reduced in a dose- and time-dependent manner. Cryosections of testes were stained with tight junction (TJ) component proteins such as occludin, claudin-11, JAM-A and ZO-1. Morphologically, increasing doses of SR X-ray consistently induced developing germ cell sloughing from the seminiferous tubules, accompanied by shrinkage of the tubules. Interestingly, TJ constituent proteins appeared to be induced by the increasing doses of SR X-ray. Up to 20 d after SR X-ray irradiation, there also appeared to be time-dependent changes on the steady-state level of these protein exhibiting differential patterns at 20-day after exposure, with JAM-A/claudin-11 still being up-regulated whereas occludin/ZO-1 being down-regulated. More importantly, the BTB damage induced by 40 Gy of SR X-ray could be significantly attenuated by antioxidant N-Acetyl-L-Cysteine (NAC) at a dose of 125 mg/kg. Taken together, our studies characterized the changes of TJ component proteins after SR X-ray irradiation, illustrating the possible protective effects of antioxidant NAC to BTB integrity.


International journal of physiology, pathophysiology and pharmacology | 2011

Silencing of SIRT2 induces cell death and a decrease in the intracellular ATP level of PC12 cells

Hui Nie; Heyu Chen; Jin Han; Yunyi Hong; Yingxin Ma; Weiliang Xia; Weihai Ying


International journal of physiology, pathophysiology and pharmacology | 2012

NAD(+) administration significantly attenuates synchrotron radiation X-ray-induced DNA damage and structural alterations of rodent testes.

Caibin Sheng; Heyu Chen; Ban Wang; Tengyuan Liu; Yunyi Hong; Jiaxiang Shao; Xin He; Yingxin Ma; Hui Nie; Na Liu; Weiliang Xia; Weihai Ying

Collaboration


Dive into the Yunyi Hong's collaboration.

Top Co-Authors

Avatar

Weihai Ying

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Heyu Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Hui Nie

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Weiliang Xia

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Caibin Sheng

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jiaxiang Shao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jie Zhang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Ban Wang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Caixia Wang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xianting Ding

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge