Network


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

Hotspot


Dive into the research topics where Kuiwu Wu is active.

Publication


Featured researches published by Kuiwu Wu.


Stem Cell Research | 2013

miR-210 suppresses BNIP3 to protect against the apoptosis of neural progenitor cells.

Fei Wang; Lei Xiong; Xin Huang; Tong Zhao; Li-Ying Wu; Zhao-hui Liu; Xuefeng Ding; Shuhong Liu; Yan Wu; Yong-Qi Zhao; Kuiwu Wu; Lingling Zhu; Ming Fan

MiR-210 is a hypoxia-inducible factor (HIF)-1 target gene and is the most consistently and predominantly upregulated miRNA in response to hypoxia in various cancer cell lines. Our recent study shows that hypoxia increased miR-210 expression in neural progenitor cells (NPCs) in a time-dependent manner. However, the role of miR-210 in NPCs remains unknown. Following the identification of the miR-210 putative target genes, we demonstrated that the Bcl-2 adenovirus E1B 19kDa-interacting protein 3 (BNIP3), which is regulated by HIF-1 and activates cell death, is regulated by miR-210 in NPCs under hypoxia. Moreover, the over-expression of miR-210 decreased apoptosis in NPCs, and the inhibition of miR-210 expression remarkably increased the number of TUNEL-positive NPCs by 30% in response to hypoxia. Importantly, miR-210 mimics reduced both BNIP3 protein expression and the translocation of AIF into the nucleus, which reduced cell death, whereas miR-210 inhibitors reversed this process, leading to cell death during hypoxia. Taken together, we report a novel feedback loop of BNIP3 regulation in NPCs under hypoxia. HIF-1 is activated under hypoxia and then induces the expression of both BNIP3 and miR-210. The upregulation of miR-210 then directly suppresses BNIP3 expression to maintain the survival of NPCs under hypoxia. This negative feedback regulation might partially contribute to protection against hypoxia-induced cell death via the inhibition of AIF nuclear translocation.


FEBS Journal | 2012

DNA demethylation regulates the expression of miR‐210 in neural progenitor cells subjected to hypoxia

Lei Xiong; Fei Wang; Xin Huang; Zhao-hui Liu; Tong Zhao; Li-Ying Wu; Kuiwu Wu; Xuefeng Ding; Shuhong Liu; Yan Wu; Yong-Qi Zhao; Lingling Zhu; Ming Fan

Several studies have identified a set of hypoxia‐regulated microRNAs, among which is miR‐210, whose expression is highly induced by hypoxia in various cancer cell lines. Recent studies have highlighted the importance of miR‐210 and its transcriptional regulation by the transcription factor hypoxia‐inducible factor‐1 (HIF‐1). We report here that the expression of miR‐210 was highly induced in neural progenitor cells (NPCs) subjected to hypoxia. Specifically, treating hypoxic NPCs with the DNA demethylating agent 5‐aza‐2′‐deoxycytidine significantly increased the expression of miR‐210, even under normoxia; however, the activity of hypoxia‐inducible factor‐1 was unaffected. Further analysis of the miR‐210 sequence revealed that it is embedded in a CpG island. Bisulfite sequencing of the miR‐210 CpG island from NPCs grown under hypoxic conditions showed 24% CpG methylation in NPCs exposed to 20% O2, 18% in NPCs exposed to 3% O2, and 12% in NPCs exposed to 0.3% O2. In addition, the activity of DNA methyltransferases (DNMTs) in NPCs decreased after exposure to hypoxia. Specifically, the expression of DNMT3b decreased significantly after exposure to 0.3% O2. Thus, these results demonstrate that DNA demethylation regulates miR‐210 expression in NPCs under both normoxia and hypoxia.


Molecular Medicine | 2015

Methylene Blue Reduces Acute Cerebral Ischemic Injury via the Induction of Mitophagy.

Yao Di; Tong Zhao; Xin Huang; Kuiwu Wu; Shuhong Liu; Yong-Qi Zhao; Ming Fan; Li-Ying Wu; Lingling Zhu

The treatment of stroke is limited by a short therapeutic window and a lack of effective clinical drugs. Methylene blue (MB) has been used in laboratories and clinics since the 1890s. Few studies have reported the neuroprotective role of MB in cerebral ischemia-reperfusion injury. However, whether and how MB protects against acute cerebral ischemia (ACI) injury was unclear. In this study, we investigated the effect of MB on this injury and revealed that MB protected against ACI injury by augmenting mitophagy. Using a rat middle cerebral artery occlusion (MCAO) model, we demonstrated that MB improved neurological function and reduced the infarct volume and necrosis after ACI injury. These improvements depended on the effect of MB on mitochondrial structure and function. ACI caused the disorder and disintegration of mitochondrial structure, while MB ameliorated the destruction of mitochondria. In addition, mitophagy was inhibited at 24 h after stroke and MB augmented mitophagy. In an oxygen-glucose deprivation (OGD) model in vitro, we further revealed that the elevation of mitochondrial membrane potential (MMP) by MB under OGD conditions mediated the augmented mitophagy. In contrast, exacerbating the decline of MMP during OGD abolished the MB-induced activation of mitophagy. Taken together, MB promotes mitophagy by maintaining the MMP at a relatively high level, which contributes to a decrease in necrosis and an improvement in neurological function, thereby protecting against ACI injury.


Molecular and Cellular Neuroscience | 2011

CHL1 negatively regulates the proliferation and neuronal differentiation of neural progenitor cells through activation of the ERK1/2 MAPK pathway.

Xin Huang; Lingling Zhu; Tong Zhao; Li-Ying Wu; Kuiwu Wu; Melitta Schachner; Zhi-Cheng Xiao; Ming Fan

Neural recognition molecules of the immunoglobulin superfamily play important roles in the development and regeneration of nervous system. Close Homologue of L1 (CHL1) is a member of the L1 family of recognition molecules which are expressed during neuronal development, suggesting a potential role in neural progenitor cells (NPCs). Here, we investigated the role of CHL1 in the proliferation and differentiation of NPCs both in vivo and in vitro, and the possible mechanism involved. The number of BrdU-positive cells in the subventricular zone (SVZ) significantly increased in CHL1-/- mice compared with CHL1+/+ mice. Moreover, there were more Tuj1-positive cells in the cortical plate region in CHL1-/- mice than in CHL1+/+ controls. To further examine the function of CHL1 in the proliferation and differentiation of NPCs, NPCs from CHL1-/- mice versus littermate wild-type mice were isolated and cultured in vitro. NPCs derived from CHL1-/- mice showed increased proliferation and self-renewal ability compared with CHL1+/+ mice. In the course of differentiation, CHL1 deficiency enhanced neuronal differentiation in the absence of growth factors. Furthermore, CHL1 deficiency on the proliferation of NPCs is accompanied by means of enhanced activation of ERK1/2 mitogen-activated protein kinase (MAPK) and the inhibitor of ERK1/2 MAPK eliminates the effect of CHL1 deficiency on the proliferation of NPCs. Our results first describe the negative modulation of the proliferation and neuronal differentiation of NPCs by CHL1/ERK1/2 MAPK signaling.


Free Radical Research | 2015

5-HMF prevents against oxidative injury via APE/Ref-1

Jian-Hong Zhang; Yao Di; Li-Ying Wu; Tong Zhao; Xin Huang; Xuefeng Ding; Kuiwu Wu; Ming Fan; Lingling Zhu

Abstract Oxidative injury is involved in many diseases, including ischemic and neurodegenerative diseases. Antioxidant drugs can be used to relieve the oxidative injury caused by these diseases; however, there are very few antioxidant drugs available for clinical use. In this study, we found that 5-(hydroxymethyl)-2-furfural (5-HMF) protects against the oxidative damage induced by cerebral ischemia in rats or by hydrogen peroxide (H2O2) in PC12 cells. We demonstrated that 5-HMF performs this function via apurinic/apyrimidinic endonuclease/redox factor-1 (APE/Ref-1). APE/Ref-1 is a multifunctional protein involved in oxidative DNA damage repair through the base excision repair (BER) pathway and in the regulation of the DNA-binding activity of several transcription factors. The current study focused on the role of APE/Ref-1 in the antioxidative properties of 5-HMF. The results show that 5-HMF inhibited the reduction of APE/Ref-1 protein level caused by cerebral ischemia–reperfusion injury in rats or H2O2 treatment in PC12 cells. Treatment with an APE/Ref-1 inhibitor blocked 5-HMF-induced protection, suggesting that APE/Ref-1s DNA repair function contributes to antioxidation. In conclusion, this study suggests that APE/Ref-1 may be a potential target for antioxidant drugs.


Neurobiology of Disease | 2014

Notch1 mediates postnatal neurogenesis in hippocampus enhanced by intermittent hypoxia.

Kuan Zhang; Tong Zhao; Xin Huang; Li-Ying Wu; Kuiwu Wu; Lingling Zhu; Ming Fan

Notch1 is a transcription factor on the membrane and regulates various stages of neurogenesis. Recently, studies have shown that in vitro neurogenesis is enhanced by hypoxia, and there is cross-coupling between Notch and hypoxia signaling pathways in vitro. However, to date, no data have reported whether Notch1 can be regulated by hypoxia in vivo and mediates hypoxia-induced neurogenesis. To determine causative links between Notch1, neurogenesis and hypoxia, we examined multiple steps of hippocampal neurogenesis followed intermittent hypoxia (IH) in wild type (WT) and Notch1 heterozygous deficient (N+/-) mice. We found that IH increased NSC proliferation, newborn neuron survival and migration, and spine morphogenesis in dentate gyrus of hippocampus, as well as neurogenesis in olfactory bulb in WT mice. However, IH-enhanced neurogenesis was inhibited in N+/- mice. It was shown that Notch1 signaling was activated following IH in WT mice, but not in N+/- mice. Our data indicated that IH, as a novel external stimulus, enhances neurogenesis at multiple stages and that Notch1 is activated by hypoxia in vivo and required for hypoxia-induced neurogenesis. These results suggest IH as a novel therapeutic strategy for degenerative neurological disorders and provide evidence for causative links between Notch1, neurogenesis and hypoxia.


Stroke | 2011

Loss of NB-3 Aggravates Cerebral Ischemia by Impairing Neuron Survival and Neurite Growth

Xin Huang; Jia Sun; Tong Zhao; Kuiwu Wu; Kazutada Watanabe; Zhi-Cheng Xiao; Lingling Zhu; Ming Fan

Background and Purpose— NB-3 is a member of the F3/contactin family of neural recognition molecules, which are crucial for cell morphogenesis and motility. NB-3 is expressed in neurons and plays an important role in axonal extension and neuronal survival. However, the role of NB-3 in cerebral ischemic injury remains unknown. Methods— Adult male wild-type and NB-3 knockout mice were subjected to ischemic injury by unilateral middle cerebral carotid artery occlusion for 3 hours, 6 hours, and 12 hours. Ischemic infarction volumes were then determined by 2, 3, 5-triphenyltetrazolium chloride staining. Neurological dysfunction analysis was also performed. Primary culture of neuronal cells from wild-type and knockout animals was also used for analysis of neuronal survival and neurite outgrowth. Results— NB-3 expression in the ischemic hemisphere was decreased after transient middle cerebral artery occlusion (MCAO). NB-3-knockout mice developed a 2.6-fold larger infarct volume and exhibited increased neurological deficit scores after transient middle cerebral artery occlusion compared with control mice. Substrate with NB-3 promoted neuronal survival and neurite outgrowth in vitro, whereas neurite outgrowth and neuronal survival were significantly reduced in NB-3-deficient neurons. In addition, NB-3 deficiency renders neurons more susceptible to oxygen–glucose deprivation-induced damage and NB-3 as substrate could partially through homophilic mechanisms. Conclusions— These data demonstrate that NB-3 deficiency may aggravate brain damage after middle cerebral artery occlusion by impairing neuronal survival and neurite growth.


Cellular and Molecular Neurobiology | 2011

Small ncRNA Expression and Regulation Under Hypoxia in Neural Progenitor Cells

Zhao-hui Liu; Guang Yang; Tong Zhao; Guojun Cao; Lei Xiong; Wei Xia; Xin Huang; Li-Ying Wu; Kuiwu Wu; Ming Fan; Ningsheng Shao; Lingling Zhu

Small non-coding RNA (ncRNA) plays critical roles in a large number of cellular processes, including neural development, cell survival and cell determination. Our previous work showed that low oxygen promoted the survival and proliferation of neural progenitor cells (NPCs) in vitro. In this study, we examine the expression and regulation of small ncRNAs in the hypoxia-driven proliferation of NPCs. The expression profiles of ncRNAs in NPCs under hypoxia were detected using microarray analysis. Results of significance analysis of microarrays (SAM) revealed that 15 small RNAs were up-regulated at least threefold and 11 were down-regulated under hypoxic conditions. The differentially expressed small ncRNAs were confirmed by quantitative RT-PCR, and miR-210 was observed to be highly expressed in NPCs under hypoxic conditions. Further study showed that hypoxia-inducible factor (HIF)-1α had a direct impact on the putative promoter regions of miR-210. From these results, we conclude that some small ncRNAs participate in the regulation of the proliferation of NPCs under hypoxia and that miR-210 is directly regulated by HIF-1α.


PLOS ONE | 2015

Reduced Cerebral Oxygen Content in the DG and SVZ In Situ Promotes Neurogenesis in the Adult Rat Brain In Vivo

Kuan Zhang; Yanzhao Zhou; Tong Zhao; Li-Ying Wu; Xin Huang; Kuiwu Wu; Lun Xu; Dahu Li; Shuhong Liu; Yong-Qi Zhao; Ming Fan; Lingling Zhu

Neurogenesis in the adult brain occurs mainly within two neurogenic structures, the dentate gyrus (DG) of the hippocampus and the sub-ventricular zone (SVZ) of the forebrain. It has been reported that mild hypoxia promoted the proliferation of Neural Stem Cells (NSCs)in vitro. Our previous study further demonstrated that an external hypoxic environment stimulated neurogenesis in the adult rat brain in vivo. However, it remains unknown how external hypoxic environments affect the oxygen content in the brain and result in neurogenesis. Here we use an optical fiber luminescent oxygen sensor to detect the oxygen content in the adult rat brain in situ under normoxia and hypoxia. We found that the distribution of oxygen in cerebral regions is spatiotemporally heterogeneous. The Po2 values in the ventricles (45∼50 Torr) and DG (approximately 10 Torr) were much higher than those of other parts of the brain, such as the cortex and thalamus (approximately 2 Torr). Interestingly, our in vivo studies showed that an external hypoxic environment could change the intrinsic oxygen content in brain tissues, notably reducing oxygen levels in both the DG and SVZ, the major sites of adult neurogenesis. Furthermore, the hypoxic environment also increased the expression of HIF-1α and VEGF, two factors that have been reported to regulate neurogenesis, within the DG and SVZ. Thus, we have demonstrated that reducing the oxygen content of the external environment decreased Po2 levels in the DG and SVZ. This reduced oxygen level in the DG and SVZ might be the main mechanism triggering neurogenesis in the adult brain. More importantly, we speculate that varying oxygen levels may be the physiological basis of the regionally restricted neurogenesis in the adult brain.


Cellular Reprogramming | 2011

Gene Expression Profiles and Metabolic Changes in Embryonic Neural Progenitor Cells Under Low Oxygen

Lingling Zhu; Tong Zhao; Xin Huang; Zhao-hui Liu; Li-Ying Wu; Kuiwu Wu; Ming Fan

Hypoxia promotes the proliferation of neural progenitor cells (NPCs), and low oxygen is a useful tool for expansion of NPCs in vitro. To further understand the regulation of the mechanisms involved, we first identified the gene expression profile of NPCs and characterized their metabolic changes in vitro under 3% oxygen. NPCs derived from E13.5 rat mesencephalon were cultured under either normoxia or hypoxia for 24 h and 72 h. Total RNA was subjected to cDNA microarray analysis of 5705 genes. The results showed that approximately 1.24% of gene expression changed under low oxygen at the two time points. Among the 142 differentially expressed genes, the greatest number was involved in glycolysis and metabolism. The metabolic changes of NPCs under low oxygen conditions were also assayed. The glucose content in the conditioned medium incubated in low oxygen decreased significantly; however, the levels of pyruvate and lactic acid increased compared to conditioned medium cultured in normoxia. The NPCs under low oxygen consumed more glucose and produced energy by glycolysis. The information gained from gene expression and metabolic analyses of NPCs under low oxygen conditions will provide new approaches for the evaluation of NPCs as potential in vivo cellular therapeutics.

Collaboration


Dive into the Kuiwu Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tong Zhao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Li-Ying Wu

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Ming Fan

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Yong-Qi Zhao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuhong Liu

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Kuan Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan Wu

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge