Network


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

Hotspot


Dive into the research topics where Xuemei Chen is active.

Publication


Featured researches published by Xuemei Chen.


Frontiers in Molecular Neuroscience | 2017

MiR-30b Attenuates Neuropathic Pain by Regulating Voltage-Gated Sodium Channel Nav1.3 in Rats

Songxue Su; Jinping Shao; Qingzan Zhao; Xiuhua Ren; Weihua Cai; Lei Li; Qian Bai; Xuemei Chen; Bo Xu; Jian Wang; Jing Cao; Weidong Zang

Nav1.3 is a tetrodotoxin-sensitive isoform among voltage-gated sodium channels that are closely associated with neuropathic pain. It can be up-regulated following nerve injury, but its biological function remains uncertain. MicroRNAs (miRNAs) are endogenous non-coding RNAs that can regulate post-transcriptional gene expression by binding with their target mRNAs. Using Target Scan software, we discovered that SCN3A is the major target of miR-30b, and we then determined whether miR-30b regulated the expression of Nav1.3 by transfecting miR-30b agomir through the stimulation of TNF-α or by transfecting miR-30b antagomir in primary dorsal root ganglion (DRG) neurons. The spinal nerve ligation (SNL) model was used to determine the contribution of miR-30b to neuropathic pain, to evaluate changes in Nav1.3 mRNA and protein expression, and to understand the sensitivity of rats to mechanical and thermal stimuli. Our results showed that miR-30b agomir transfection down-regulated Nav1.3 mRNA stimulated with TNF-α in primary DRG neurons. Moreover, miR-30b overexpression significantly attenuated neuropathic pain induced by SNL, with decreases in the expression of Nav1.3 mRNA and protein both in DRG neurons and spinal cord. Activation of Nav1.3 caused by miR-30b antagomir was identified. These data suggest that miR-30b is involved in the development of neuropathic pain, probably by regulating the expression of Nav1.3, and might be a novel therapeutic target for neuropathic pain. Perspective: This study is the first to explore the important role of miR-30b and Nav1.3 in spinal nerve ligation-induced neuropathic pain, and our evidence may provide new insight for improving therapeutic approaches to pain.


Journal of Molecular Neuroscience | 2017

Small Interfering RNA Targeting Dickkopf-1 Contributes to Neuroprotection After Intracerebral Hemorrhage in Rats.

Zhihua Li; Xi Chen; Xiaoyang Zhang; Xiuhua Ren; Xuemei Chen; Jing Cao; Weidong Zang; Xianzhi Liu; Fuyou Guo

Excessive Dickkopf-1 (Dkk-1) plays a vital role in secondary brain injury following ischemic stroke and psychotic disease. However, it is unclear whether an increased expression of Dkk-1 occurred after intracerebral hemorrhage (ICH). The present study examined the potential role of Dkk-1 after ICH. ICH was induced by a single injection of autologous blood into the basal ganglia of rats. Dkk-1 protein levels in brain tissue and serum were detected by enzyme-linked immunosorbent assay after ICH. Rats were treated with small interfering RNA targeting Dkk-1 (siDkk-1) or vehicle following ICH. Behavioral deficits and brain water content were examined. Blood–brain barrier (BBB) integrity was detected by Evans blue extravasation and observed by transmission electron microscopy. Wnt-1 was evaluated by real-time RT-PCR. The tight junction protein zonula occludens-1 (ZO-1) was investigated by immunohistochemistry and Western blot assays. Serum level of Dkk-1 did not differ between the ICH and sham groups. However, the level of Dkk-1 in brain tissue was significantly increased at 24 and 72xa0h after ICH. BBB disruption and brain edema, as well as neurological deficits, were remarkably ameliorated by administration of siDkk-1. Moreover, siDkk-1 treatment significantly increased the transcription of Wnt-1 mRNA and upregulated the expression of ZO-1. These results provide the first evidence that siDkk-1 treatment is neuroprotective against secondary injury including brain edema and BBB permeability following ICH; the mechanism of neuroprotection may be associated with improvement of BBB integrity.


Journal of Cerebral Blood Flow and Metabolism | 2018

20-HETE synthesis inhibition promotes cerebral protection after intracerebral hemorrhage without inhibiting angiogenesis

Xiaoning Han; Xiaochun Zhao; Xi Lan; Qian Li; Yufeng Gao; Xi Liu; Jieru Wan; Zengjin Yang; Xuemei Chen; Weidong Zang; Austin M. Guo; John R. Falck; Raymond C. Koehler; Jian Wang

20-HETE, an arachidonic acid metabolite synthesized by cytochrome P450 4A, plays an important role in acute brain damage from ischemic stroke or subarachnoid hemorrhage. We tested the hypothesis that 20-HETE inhibition has a protective effect after intracerebral hemorrhage (ICH) and then investigated its effect on angiogenesis. We exposed hippocampal slice cultures to hemoglobin and induced ICH in mouse brains by intrastriatal collagenase injection to investigate the protective effect of 20-HETE synthesis inhibitor N-hydroxy-N′-(4-n-butyl-2-methylphenyl)-formamidine (HET0016). Hemoglobin-induced neuronal death was assessed by propidium iodide after 18u2009h in vitro. Lesion volume, neurologic deficits, cell death, reactive oxygen species (ROS), neuroinflammation, and angiogenesis were evaluated at different time points after ICH. In cultured mouse hippocampal slices, HET0016 attenuated hemoglobin-induced neuronal death and decreased levels of proinflammatory cytokines and ROS. In vivo, HET0016 reduced brain lesion volume and neurologic deficits, and decreased neuronal death, ROS production, gelatinolytic activity, and the inflammatory response at three days after ICH. However, HET0016 did not inhibit angiogenesis, as levels of CD31, VEGF, and VEGFR2 were unchanged on day 28. We conclude that 20-HETE is involved in ICH-induced brain damage. Inhibition of 20-HETE synthesis may provide a viable means to mitigate ICH injury without inhibition of angiogenesis.


Brain Behavior and Immunity | 2018

Changes in motor function, cognition, and emotion-related behavior after right hemispheric intracerebral hemorrhage in various brain regions of mouse

Wei Zhu; Yufeng Gao; Jieru Wan; Xi Lan; Xiaoning Han; Shanshan Zhu; Weidong Zang; Xuemei Chen; Wendy C. Ziai; Daniel F. Hanley; Scott J. Russo; Ricardo E. Jorge; Jian Wang

Intracerebral hemorrhage (ICH) is a detrimental type of stroke. Mouse models of ICH, induced by collagenase or blood infusion, commonly target striatum, but not other brain sites such as ventricular system, cortex, and hippocampus. Few studies have systemically investigated brain damage and neurobehavioral deficits that develop in animal models of ICH in these areas of the right hemisphere. Therefore, we evaluated the brain damage and neurobehavioral dysfunction associated with right hemispheric ICH in ventricle, cortex, hippocampus, and striatum. The ICH model was induced by autologous whole blood or collagenase VII-S (0.075 units in 0.5u202fµl saline) injection. At different time points after ICH induction, mice were assessed for brain tissue damage and neurobehavioral deficits. Sham control mice were used for comparison. We found that ICH location influenced features of brain damage, microglia/macrophage activation, and behavioral deficits. Furthermore, the 24-point neurologic deficit scoring system was most sensitive for evaluating locomotor abnormalities in all four models, especially on days 1, 3, and 7 post-ICH. The wire-hanging test was useful for evaluating locomotor abnormalities in models of striatal, intraventricular, and cortical ICH. The cylinder test identified locomotor abnormalities only in the striatal ICH model. The novel object recognition test was effective for evaluating recognition memory dysfunction in all models except for striatal ICH. The tail suspension test, forced swim test, and sucrose preference test were effective for evaluating emotional abnormality in all four models but did not correlate with severity of brain damage. These results will help to inform future preclinical studies of ICH outcomes.


Translational Stroke Research | 2017

Alpha-7 Nicotinic Receptor Signaling Pathway Participates in the Neurogenesis Induced by ChAT-Positive Neurons in the Subventricular Zone

Jianping Wang; Zhengfang Lu; Xiaojie Fu; Di Zhang; Lie Yu; Nan Li; Yufeng Gao; Xianliang Liu; Chunmao Yin; Junji Ke; Liyuan Li; Mengmeng Zhai; Shiwen Wu; Jiahong Fan; Liang Lv; Junchao Liu; Xuemei Chen; Qing-Wu Yang; Jian Wang

Choline acetyltransferase-positive (ChAT+) neurons within the subventricular zone (SVZ) have been shown to promote neurogenesis after stroke in mice by secreting acetylcholine (ACh); however, the mechanisms remain unclear. Receptors known to bind ACh include the nicotinic ACh receptors (nAChRs), which are present in the SVZ and have been shown to be important for cell proliferation, differentiation, and survival. In this study, we investigated the neurogenic role of the alpha-7 nAChR (α7 nAChR) in a mouse model of middle cerebral artery occlusion (MCAO) by using α7 nAChR inhibitor methyllycaconitine. Mice subjected to MCAO exhibited elevated expression of cytomembrane and nuclear fibroblast growth factor receptor 1 (FGFR1), as well as increased expression of PI3K, pAkt, doublecortin (DCX), polysialylated - neuronal cell adhesion molecule (PSA-NCAM), and mammalian achaete-scute homolog 1 (Mash1). MCAO mice also had more glial fibrillary acidic protein (GFAP)/5-bromo-2′-deoxyuridine (BrdU)-positive cells and DCX-positive cells in the SVZ than did the sham-operated group. Methyllycaconitine treatment increased cytomembrane FGFR1 expression and GFAP/BrdU-positive cells, upregulated the levels of phosphoinositide 3-kinase (PI3K) and phospho-Akt (pAkt), decreased nuclear FGFR1 expression, decreased the number of DCX-positive cells, and reduced the levels of DCX, PSA-NCAM, and Mash1 in the SVZ of MCAO mice compared with levels in vehicle-treated MCAO mice. MCAO mice treated with α7 nAChR agonist PNU-282987 exhibited the opposite effects. Our data show that α7 nAChR may decrease the proliferation of neural stem cells and promote differentiation of existing neural stem cells after stroke. These results identify a new mechanism of SVZ ChAT+ neuron-induced neurogenesis.


Neuroscience | 2017

Effects of crenolanib, a nonselective inhibitor of PDGFR, in a mouse model of transient middle cerebral artery occlusion

Jianping Wang; Xiaojie Fu; Di Zhang; Lie Yu; Zhengfang Lu; Yufeng Gao; Xianliang Liu; Jiang Man; Sijia Li; Nan Li; Menghan Wang; Xi Liu; Xuemei Chen; Weidong Zang; Qing-Wu Yang; Jian Wang

Neurogenesis in the subventricular zone (SVZ) plays a vital role in neurologic recovery after stroke. However, only a small fraction of newly generated neuroblasts from the SVZ will survive long-term. Successful migration and survival of neuroblasts requires angiogenesis, lesion-derived chemo-attractants, and appropriate local microenvironments, which are partly regulated by the platelet-derived growth factor receptor (PDGFR) signaling pathway. In this study, we investigated the effects of PDGFR inhibition in a mouse model of transient middle cerebral artery occlusion (MCAO). We blocked the pathway using a nonselective PDGFR inhibitor, crenolanib, during the acute post-MCAO phase (days 1-3) or during the sub-acute phase (days 7-9). Downregulating the PDGFR signaling pathway with crenolanib from day 1 to day 3 after MCAO significantly decreased the migration of neuroblasts from the SVZ to the peri-infarct region, decreased angiogenesis, and lowered expression of vascular endothelial growth factor, stromal cell-derived factor-1, and monocyte chemotactic protein-1. Downregulation of the PDGFR signaling pathway on days 7-9 with crenolanib significantly increased apoptosis of the neuroblasts that had migrated to the peri-infarct region, increased the number of activated microglia, and decreased the expression of brain-derived neurotrophic factor, neurotrophin-3, and interleukin-10. Crenolanib treatment increased the apoptosis of pericytes and decreased the pericyte/vascular coverage, but had no effects on apoptosis of astrocytes. We conclude that the PDGFR signaling pathway plays a vital role in the SVZ neurogenesis after stroke. It can also affect angiogenesis, lesion-derived chemo-attractants, and the local microenvironment, which are all important to stroke-induced neurogenesis.


Behavioural Brain Research | 2019

Hippo/YAP signaling pathway mitigates blood-brain barrier disruption after cerebral ischemia/reperfusion injury

Pian Gong; Zhan Zhang; Changlin Zou; Qi Tian; Xuemei Chen; Michael Hong; Xi Liu; Qianxue Chen; Zhou Xu; Mingchang Li; Jian Wang

&NA; Ischemia/reperfusion (I/R) injuries commonly lead to breakdown of the blood‐brain barrier (BBB). Restoration of the BBB can relieve neurologic damage caused by I/R injuries. The Hippo/YAP signaling pathway mediates cell proliferation, regulated cell death, and differentiation in various organisms and has been shown to participate in the restoration of the heart after I/R. In this study, we investigated whether the Hippo/YAP pathway plays a role in I/R injury in brain, especially in regard to I/R‐induced BBB breakdown. The results of our study indicate that I/R injury led to an overall decrease in activity of the core proteins, YAP and TAZ, over a 24‐h period. The most dramatic change was observed 1.5 h after reperfusion. In rats that underwent 1.5 h of reperfusion, intraperitoneal injection of YAP agonist dexamethasone activated YAP and TAZ and led to improved neurologic function, smaller brain infarct sizes, increased levels of tight junction proteins, decreased BBB permeability, decreased cerebral edema, and less apoptosis. Our results suggest that YAP exerts neuroprotective effects on the damaged brain that are likely related to restoration of the BBB. HighlightsYAP and TAZ decrease after ischemic/reperfusion injury.Activation of YAP or TAZ mediates protection against blood‐brain barrier disruption.The Hippo/YAP signaling pathway is involved in ischemic/reperfusion injury.


Journal of Neuroinflammation | 2018

Carbon monoxide-releasing molecule-3 protects against ischemic stroke by suppressing neuroinflammation and alleviating blood-brain barrier disruption

Jianping Wang; Di Zhang; Xiaojie Fu; Lie Yu; Zhengfang Lu; Yufeng Gao; Xianliang Liu; Jiang Man; Sijia Li; Nan Li; Xuemei Chen; Michael Hong; Qing-Wu Yang; Jian Wang

BackgroundAt low levels, carbon monoxide (CO) has been shown to have beneficial effects on multiple organs and tissues through its potential anti-inflammatory, anti-apoptotic, and anti-proliferative properties. However, the effect of CO-releasing molecule (CORM)-3, a water-soluble CORM, on ischemic stroke and its mechanism of action are still unclear.MethodsWe investigated the role of CORM-3 in the mouse model of transient middle cerebral artery occlusion (tMCAO). CORM-3 or saline was administered to mice by retro-orbital injection at the time of reperfusion after 1-h tMCAO or at 1xa0h after sham surgery. We assessed infarct volume and brain water content at 24 and 72xa0h after ischemia, blood-brain barrier permeability at 6 and 72xa0h after ischemia, and neurologic deficits on days 1, 3, 7, and 14.ResultsAmong mice that underwent tMCAO, those that received CORM-3 had significantly smaller infarct volume and greater expression of neuronal nuclear antigen (NeuN) and microtubule-associated protein 2 than did saline-treated mice. CORM-3-treated mice had significantly fewer activated microglia in the peri-infarction zone than did control mice and exhibited downregulated expression of ionized calcium-binding adapter molecule (Iba)-1, tumor necrosis factor-α, and interleukin 1β. CORM-3-treated mice had significantly lower brain water content and enhanced neurologic outcomes on days 3, 7, and 14 post-tMCAO. Lastly, CORM-3 treatment reduced Evans blue leakage; increased expression of platelet-derived growth factor receptor-β, tight junction protein ZO-1, and matrix protein laminin; and decreased protein level of matrix metalloproteinase-9.ConclusionCORM-3 treatment at the time of reperfusion reduces ischemia-reperfusion-induced brain injury by suppressing neuroinflammation and alleviating blood-brain barrier disruption. Our data suggest that CORM-3 may provide an effective therapy for ischemic stroke.


Frontiers in Neurology | 2018

Ultrastructural Characteristics of Neuronal Death and White Matter Injury in Mouse Brain Tissues After Intracerebral Hemorrhage: Coexistence of Ferroptosis, Autophagy, and Necrosis

Qian Li; Abigail Weiland; Xuemei Chen; Xi Lan; Xiaoning Han; Frederick Durham; Xi Liu; Jieru Wan; Wendy C. Ziai; Daniel F. Hanley; Jian Wang

Although intracerebral hemorrhage (ICH) is a devastating disease worldwide, the pathologic changes in ultrastructure during the acute and chronic phases of ICH are poorly described. In this study, transmission electron microscopy was used to examine the ultrastructure of ICH-induced pathology. ICH was induced in mice by an intrastriatal injection of collagenase. Pathologic changes were observed in the acute (3 days), subacute (6 days), and chronic (28 days) phases. Compared with sham animals, we observed various types of cell death in the injured striatum during the acute phase of ICH, including necrosis, ferroptosis, and autophagy. Different degrees of axon degeneration in the striatum were seen in the acute phase, and axonal demyelination was observed in the ipsilateral striatum and corpus callosum at late time points. In addition, phagocytes, resident microglia, and infiltrating monocyte-macrophages were present around red blood cells and degenerating neurons and were observed to engulf red blood cells and other debris. Many synapses appeared abnormal or were lost. This systematic analysis of the pathologic changes in ultrastructure after ICH in mice provides information that will be valuable for future ICH pathology studies.


Free Radical Biology and Medicine | 2018

Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways

Xiangsheng Zhang; Qi Wu; Yue Lu; Jieru Wan; Haibin Dai; Xiao-Ming Zhou; Shengyin Lv; Xuemei Chen; Xin Zhang; Chun-Hua Hang; Jian Wang

Abstract Salvianolic acid B (SalB), a natural polyphenolic compound extracted from the herb of Salvia miltiorrhiza, possesses antioxidant and neuroprotective properties and has been shown to be beneficial for diseases that affect vasculature and cognitive function. Here we investigated the protective effects of SalB against subarachnoid hemorrhage (SAH)‐induced oxidative damage, and the involvement of underlying molecular mechanisms. In a rat model of SAH, SalB inhibited SAH‐induced oxidative damage. The reduction in oxidative damage was associated with suppressed reactive oxygen species generation; decreased lipid peroxidation; and increased glutathione peroxidase, glutathione, and superoxide dismutase activities. Concomitant with the suppressed oxidative stress, SalB significantly reduced neurologic impairment, brain edema, and neural cell apoptosis after SAH. Moreover, SalB dramatically induced nuclear factor‐erythroid 2‐related factor 2 (Nrf2) nuclear translocation and increased expression of heme oxygenase‐1 and NADPH: quinine oxidoreductase‐1. In a mouse model of SAH, Nrf2 knockout significantly reversed the antioxidant effects of SalB against SAH. Additionally, SalB activated sirtuin 1 (SIRT1) expression, whereas SIRT1‐specific inhibitor sirtinol pretreatment significantly suppressed SalB‐induced SIRT1 activation and Nrf2 expression. Sirtinol pretreatment also reversed the antioxidant and neuroprotective effects of SalB. In primary cultured cortical neurons, SalB suppressed oxidative damage, alleviated neuronal degeneration, and improved cell viability. These beneficial effects were associated with activation of the SIRT1 and Nrf2 signaling pathway and were reversed by sirtinol treatment. Taken together, these in vivo and in vitro findings suggest that SalB provides protection against SAH‐triggered oxidative damage by upregulating the Nrf2 antioxidant signaling pathway, which may be modulated by SIRT1 activation. Graphical abstract Schematic illustrating the possible mechanisms of salvianolic acid B (SalB) action after subarachnoid hemorrhage (SAH). As illustrated, SAH markedly increases reactive oxygen species (ROS) generation in the extracellular space. After stimulation, nuclear factor‐erythroid 2‐related factor 2 (Nrf2) dissociates from Keap1 and translocates into the nucleus, subsequently inducing the expression of antioxidant enzymes, including heme oxygenase‐1 (HO‐1), NADPH: quinine oxidoreductase‐1 (NQO‐1), and superoxide dismutase (SOD), which are critical for maintaining cellular redox homeostasis against oxidative insults. In addition, in response to ROS accumulation after SAH, sirtuin 1 (SIRT1) activation inhibits FoxO1 and P53 acetylation and increases Nrf2 expression to reduce brain damage after SAH. SalB treatment induces the expression of SIRT1 and Nrf2, which in turn modulate downstream antioxidant enzyme expression, thereby ameliorating brain damage after SAH. In contrast, SIRT1‐specific inhibitor sirtinol reverses the protective effects against SAH by inhibiting SIRT1 as well as Nrf2 activation, eventually aggravating SAH‐induced brain damage. Figure. No Caption available. HighlightsSalvianolic acid B (SalB) reduces oxidative damage after experimental subarachnoid hemorrhage (SAH) in vivo and in vitro.Activation of Nrf2 by SalB is modulated by SIRT1 activation.Nrf2 knockout suppresses the antioxidative effects of SalB against SAH.SalB induces the Nrf2‐ and sirtuin 1 (SIRT1)‐dependent pathways after SAH.

Collaboration


Dive into the Xuemei Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Wang

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Jian Wang

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Jieru Wan

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Di Zhang

Zhengzhou University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lie Yu

Zhengzhou University

View shared research outputs
Top Co-Authors

Avatar

Nan Li

Zhengzhou University

View shared research outputs
Top Co-Authors

Avatar

Qing-Wu Yang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge