Network


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

Hotspot


Dive into the research topics where Handong Wang is active.

Publication


Featured researches published by Handong Wang.


Free Radical Biology and Medicine | 2014

Melatonin stimulates antioxidant enzymes and reduces oxidative stress in experimental traumatic brain injury: the Nrf2–ARE signaling pathway as a potential mechanism

Ke Ding; Handong Wang; Jianguo Xu; Tao Li; Li Zhang; Yu Ding; Lin Zhu; Mengliang Zhou

UNLABELLED The goal of this study was to evaluate the potential involvement of melatonin in the activation of the nuclear factor erythroid 2-related factor 2 and antioxidant-responsive element (Nrf2-ARE) signaling pathway and the modulation of antioxidant enzyme activity in an experimental model of traumatic brain injury (TBI). In experiment 1, ICR mice were divided into four groups: sham group, TBI group, TBI + vehicle group, and TBI + melatonin group (n = 38 per group). Melatonin (10mg/kg) was administered via an intraperitoneal (ip) injection at 0, 1, 2, 3, and 4h post-TBI. In experiment 2, Nrf2 wild-type (Nrf2(+/+) group) and Nrf2-knockout (Nrf2(-/-) group) mice received a TBI insult followed by melatonin administration (10mg/kg, ip) at the corresponding time points (n = 35 per group). The administration of melatonin after TBI significantly ameliorated the effects of the brain injury, such as oxidative stress, brain edema, and cortical neuronal degeneration. Melatonin markedly promoted the translocation of Nrf2 protein from the cytoplasm to the nucleus; increased the expression of Nrf2-ARE pathway-related downstream factors, including heme oxygenase-1 and NAD(P)H quinone oxidoreductase 1; and prevented the decline of antioxidant enzyme activities, including superoxide dismutase and glutathione peroxidase. Furthermore, knockout of Nrf2 partly reversed the neuroprotection of melatonin after TBI. In conclusion, melatonin administration may increase the activity of antioxidant enzymes and attenuate brain injury in a TBI model, potentially via mediation of the Nrf2-ARE pathway.


Mediators of Inflammation | 2011

Beneficial Effects of Ethyl Pyruvate through Inhibiting High-Mobility Group Box 1 Expression and TLR4/NF-κB Pathway after Traumatic Brain Injury in the Rat

Xingfen Su; Handong Wang; Jinbing Zhao; Hao Pan; Lei Mao

Ethyl pyruvate (EP) has demonstrated neuroprotective effects against acute brain injury through its anti-inflammatory action. The nuclear protein high-mobility group box 1 (HMGB1) can activate inflammatory pathways when released from dying cells. This study was designed to investigate the protective effects of EP against secondary brain injury in rats after Traumatic Brain Injury (TBI). Adult male rats were randomly divided into three groups: (1) Sham + vehicle group, (2) TBI + vehicle group, and (3) TBI + EP group (n = 30 per group). Right parietal cortical contusion was made by using a weight-dropping TBI method. In TBI + EP group, EP was administered intraperitoneally at a dosage of 75 mg/kg at 5 min, 1 and 6 h after TBI. Brain samples were harvested at 24 h after TBI. We found that EP treatment markedly inhibited the expressions of HMGB1 and TLR4, NF-κB DNA binding activity and inflammatory mediators, such as IL-1β, TNF-α and IL-6. Also, EP treatment significantly ameliorated beam walking performance, brain edema, and cortical apoptotic cell death. These results suggest that the protective effects of EP may be mediated by the reduction of HMGB1/TLR4/NF-κB-mediated inflammatory response in the injured rat brain.


Free Radical Biology and Medicine | 2014

Luteolin provides neuroprotection in models of traumatic brain injury via the Nrf2–ARE pathway

Jianguo Xu; Handong Wang; Ke Ding; Li Zhang; Chunxi Wang; Tao Li; Wuting Wei; Xinyu Lu

UNLABELLED Luteolin has recently been proven to exert neuroprotection in a variety of neurological diseases; however, its roles and the underlying mechanisms in traumatic brain injury are not fully understood. The present study was aimed to investigate the neuroprotective effects of luteolin in models of traumatic brain injury (TBI) and the possible role of the Nrf2-ARE pathway in the putative neuroprotection. A modified Marmarou׳s weight-drop model in mice and the scratch model in mice primary cultured neurons were used to induce TBI. We determined that luteolin significantly ameliorated secondary brain injury induced by TBI, including neurological deficits, brain water content, and neuronal apoptosis. Furthermore, the level of malondialdehyde (MDA) and the activity of glutathione peroxidase (GPx) were restored in the group with luteolin treatment. in vitro studies showed that luteolin administration lowered the intracellular reactive oxygen species (ROS) level and increased the neuron survival. Moreover, luteolin enhanced the translocation of Nrf2 to the nucleus both in vivo and in vitro, which was proved by the results of Western blot, immunohistochemistry, and electrophoretic mobility shift assay (EMSA). Subsequently upregulation of the expression of the downstream factors such as heme oxygenase 1 (HO1) and NAD(P)H quinone oxidoreductase 1 (NQO1) was also examined. However, luteolin treatment failed to provide neuroprotection after TBI in Nrf2(-/-) mice. Taken together, these in vivo and in vitro data demonstrated that luteolin provided neuroprotective effects in the models of TBI, possibly through the activation of the Nrf2-ARE pathway.


Toxicology Letters | 2014

Inhibition of cathepsin S induces autophagy and apoptosis in human glioblastoma cell lines through ROS-mediated PI3K/AKT/mTOR/p70S6K and JNK signaling pathways.

Li Zhang; Handong Wang; Jianguo Xu; Jianhong Zhu; Ke Ding

Cathepsin S is a lysosomal cysteine protease that is overexpressed in various cancer models and plays important role in tumorigenesis, however the mechanisms are unclear. In the present study, we found that inhibition of cathepsin S induced autophagy and mitochondrial apoptosis in human glioblastoma cells. Blockade of autophagy by either a chemical inhibitor or RNA interference attenuated cathespin S inhibition-induced apoptosis. Furthermore, autophagy and apoptosis induction was dependent on the suppression of phosphatidylinositide 3-kinases/protein kinase B/mammalian target of rapamycin/p70S6 kinase (PI3K/AKT/mTOR/p70S6K) signaling pathway and activation of c-Jun N-terminal kinase (JNK) signaling pathway. In addition, reactive oxygen species (ROS) served as an upstream of PI3K/AKT/mTOR/p70S6K and JNK signaling pathways. In conclusion, the current study revealed that cathepsin S played an important role in the regulation of autophagy and apoptosis in human glioblastoma cells.


Mediators of Inflammation | 2010

Disruption of Nrf2 enhances the upregulation of nuclear factor-kappaB activity, tumor necrosis factor-α, and matrix metalloproteinase-9 after spinal cord injury in mice.

Lei Mao; Handong Wang; Liang Qiao; Xiaoliang Wang

Matrix metalloproteinase-9 (MMP-9) plays an important role in the acute periods of spinal cord injury (SCI), and its expression is related to the inflammation which could cause the disruption of the blood-spinal barrier (BBB). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor that plays a crucial role in cytoprotection against inflammation. The present study investigated the role of Nrf2 in upregulating of nuclear factor kappa B (NF-κB) activity, tumor necrosis factor-α (TNF-α), and MMP-9 after SCI. Wild-type Nrf2 (+/+) and Nrf2-deficient (Nrf (−/−)) mice were subjected to an SCI model induced by the application of vascular clips (force of 10 g) to the dura after a three-level T8-T10 laminectomy. We detected the wet/dry weight ratio of impaired spinal cord tissue, the activation of NF-κB, the mRNA and protein levels of TNF-α and MMP-9, and the enzyme activity of MMP-9. Nrf2 (−/−) mice were demonstrated to have more spinal cord edema, NF-κB activation, TNF-α production, and MMP-9 expression after SCI compared with the wild-type controls. The results suggest that Nrf2 may play an important role in limiting the upregulation of NF-κB activity, TNF-α, and MMP-9 in spinal cord after SCI.


Neurochemistry International | 2014

Melatonin reduced microglial activation and alleviated neuroinflammation induced neuron degeneration in experimental traumatic brain injury: Possible involvement of mTOR pathway

Ke Ding; Handong Wang; Jianguo Xu; Xinyu Lu; Li Zhang; Lin Zhu

This study was designed to detect the modulation manner of melatonin on microglial activation and explore herein possible involvement of mammalian target of rapamycin (mTOR) pathway following traumatic brain injury (TBI). ICR mice were divided into four groups: sham group, TBI group, TBI+sal group and TBI+Melatonin group. A weight-drop model was employed to cause TBI. Neurological severity score (NSS) tests were performed to measure behavioral outcomes. Nissl staining was conducted to observe the neuronal degeneration and wet-to-dry weight ratio indicated brain water content. Immunofluorescence was designed to investigate microglial activation. Enzyme-linked immunosorbent assay (ELISA) was employed to evaluate proinflammatory cytokine levels (interleukin-beta (IL-1β), tumor necrosis factor-alpha (TNF-α)). Western blotting was engaged to analyze the protein content of mammalian target of rapamycin (mTOR), p70 ribosomal S6 kinase (p70S6K) and S6 ribosomal protein (S6RP). Melatonin administration was associated with markedly restrained microglial activation, decreased release of proinflammatory cytokines and increased the number of surviving neurons at the site of peri-contusion. Meanwhile, melatonin administration resulted in dephosphorylated mTOR pathway. In conclusion, this study presents a new insight into the mechanisms responsible for the anti-neuroinflammation of melatonin, with possible involvement of mTOR pathway.


International Journal of Cancer | 2014

Knockdown of Nrf2 suppresses glioblastoma angiogenesis by inhibiting hypoxia‐induced activation of HIF‐1α

Xiangjun Ji; Handong Wang; Jianhong Zhu; Lin Zhu; Hao Pan; Wei Li; Yuan Zhou; Zi-Xiang Cong; Feng Yan; Suihua Chen

Concerns were increasingly raised that several types of cancers overexpressed the nuclear factor erythroid 2‐related factor 2 (Nrf2), which contributed strikingly to cancer biological capabilities and chemoresistance. However, the role of Nrf2 in the tumor vascular biology had yet to be mechanistically determined. Here, we investigated the involvement of Nrf2 in glioblastoma (GB) angiogenesis in hypoxia. First, we detected the overexpression of Nrf2 and correlated its protein level with microvessel density (MVD) in human GB tissues. Then, we established the stable RNAi‐mediated Nrf2‐knockdown cells and mimicked hypoxic condition in vitro. The knockdown of Nrf2 inhibited cell proliferation in vitro and suppressed tumor growth in mouse xenografts with a concomitant reduction in VEGF expression and MVD. Similar antiangiogenic effects were documented in endothelial tube formation assays. The downregulation of Nrf2 in glioma cells led to much lower accumulation of HIF‐1α protein and limited expression of VEGF and other HIF‐1α target genes in mimicking hypoxia. Mechanistic investigations suggested that HIF‐1α degradation during hypoxia could be attributed to reduced mitochondrial O2 consumption in Nrf2‐inhibited cells. It can be concluded that Nrf2, with its capacity for affecting the protein level of HIF‐1α expression, has good reasons to be considered as a critical transcription factor for controlling glioma angiogenesis.


Mediators of Inflammation | 2012

The Absence of Nrf2 Enhances NF-κB-Dependent Inflammation following Scratch Injury in Mouse Primary Cultured Astrocytes

Hao Pan; Handong Wang; Xiaoliang Wang; Lin Zhu; Lei Mao

It has been proved that Nrf2 depletion enhances inflammatory process through activation of NF-κB in the brain after TBI, but little is known about the relationship between Nrf2 and NF-κB in astrocytes after TBI. Hence, we used primary cultured astrocytes from either Nrf2 wildtype or knockout mice to study the influence of Nrf2 on the activation of NF-κB and expression of proinflammatory cytokines in a model of TBI in vitro. Primary cultured astrocytes were scratched to mimic the traumatic injury in vitro. Then the DNA-binding activity of NF-κB was evaluated by EMSA. The mRNA and protein levels of TNF-α, IL-1β, IL-6, and MMP9 were also evaluated. Gelatin zymography was performed to detect the activity of MMP9. The activity of NF-κB and expression of proinflammatory cytokines mentioned above were upregulated at 24 h after scratch. The expression and activity of MMP9 were also elevated. And such tendency was much more prominent in Nrf2 KO astrocytes than that in WT astrocytes. These results suggest that the absence of Nrf2 may induce more aggressive inflammation through activation of NF-κB and downstream proinflammatory cytokines in astrocytes.


World Neurosurgery | 2013

The role of Nrf2 in migration and invasion of human glioma cell U251.

Hao Pan; Handong Wang; Lin Zhu; Lei Mao; Liang Qiao; Xingfen Su

OBJECTIVE NF-E2-related factor 2 (Nrf2) is a transcription factor that is related to tumor cell multidrug resistance and proliferation. Here we studied the involvement of Nrf2 in the migration and invasion of human U251 glioma cells. METHODS Two kinds of plasmid, that is, pEGFP-Nrf2 and Si-Nrf2, were constructed and transfected to upregulate or downregulate the expression of Nrf2 in U251 glioma cell line. Blank vectors or random siRNA plasmid were used as negative control. Cells treated with lipofectamine only were set up as blank control. Protein and mRNA level of Nrf2 and matrix metalloproteinase 9 (MMP9) were investigated by reverse transcriptase-polymerase chain reaction and western blot after transfection. Wound healing assay and transwell assay were used to study migration and invasion of U251 after transfection. Gelatin zymography was performed to reveal the change of MMP9 activity after transfection. RESULTS The mRNA and protein level of Nrf2 was upregulated in U251-pEGFP-Nrf2 while downregulated in U251-Si-Nrf2 48 hours after transfection. In the wound healing assay, there were more cells in group pEGFP-Nrf2 crossing the scratch line than in group Si-Nrf2. Furthermore, in transwell migration and invasion assay, there were more cells in group pEGFP-Nrf2 penetrating the membranes than in group Si-Nrf2. Then we investigated the change of MMP9 activity, mRNA, and protein levels after transfection. The results suggested that upregulation of Nrf2 led to an increase in MMP9 expression and activity whereas downregulation of Nrf2 led to a decrease in MMP9 expression and activity. CONCLUSION Nrf2 is involved in migration and invasion of U251 cells, which may be related to MMP9.


Mediators of Inflammation | 2013

Inhibition of Cathepsin S Produces Neuroprotective Effects after Traumatic Brain Injury in Mice

Jianguo Xu; Handong Wang; Ke Ding; Xinyu Lu; Tao Li; Jiawei Wang; Chunxi Wang; Jian Wang

Cathepsin S (CatS) is a cysteine protease normally present in lysosomes. It has long been regarded as an enzyme that is primarily involved in general protein degradation. More recently, mounting evidence has shown that it is involved in Alzheimer disease, seizures, age-related inflammatory processes, and neuropathic pain. In this study, we investigated the time course of CatS protein and mRNA expression and the cellular distribution of CatS in a mouse model of traumatic brain injury (TBI). To clarify the roles of CatS in TBI, we injected the mice intraventricularly with LHVS, a nonbrain penetrant, irreversible CatS inhibitor, and examined the effect on inflammation and neurobehavioral function. We found that expression of CatS was increased as early as 1 h after TBI at both protein and mRNA levels. The increased expression was detected in microglia and neurons. Inhibition of CatS significantly reduced the level of TBI-induced inflammatory factors in brain tissue and alleviated brain edema. Additionally, administration of LHVS led to a decrease in neuronal degeneration and improved neurobehavioral function. These results imply that CatS is involved in the secondary injury after TBI and provide a new perspective for preventing secondary injury after TBI.

Collaboration


Dive into the Handong Wang'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge