Network


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

Hotspot


Dive into the research topics where XiaoOu Mao is active.

Publication


Featured researches published by XiaoOu Mao.


Neuroscience | 2000

Induction of vascular endothelial growth factor and hypoxia-inducible factor-1α by global ischemia in rat brain

Kunlin Jin; XiaoOu Mao; Tetsuya Nagayama; P.C Goldsmith; David A. Greenberg

Vascular endothelial growth factor is an angiogenic and neurotrophic peptide whose expression is transcriptionally induced in hypoxic tissues through the action of hypoxia-inducible factor-1alpha. To determine if this signaling pathway is activated in the ischemic brain, and might therefore participate in adaptive processes such as angiogenesis and neuroprotection, we examined the expression of vascular endothelial growth factor and hypoxia-inducible factor-1alpha in cerebral cortex and hippocampus following transient global cerebral ischemia in the rat. Northern analysis showed ischemia-inducible expression of multiple vascular endothelial growth factor messenger ribonucleic acid splice variants between 4 and 24h. Western analysis and immunocytochemistry demonstrated the concerted induction of vascular endothelial growth factor and hypoxia-inducible factor-1alpha in the same, apparently neuronal, cells in vulnerable regions of cortex and hippocampus after 15min of ischemia, which persisted for as long as 4 to 72h of reperfusion. These findings demonstrate that hypoxia-sensitive vascular endothelial growth factor signaling can be induced in neurons in global cerebral ischemia in vivo, and are consistent with the hypothesis that ischemic insults trigger hypoxia-sensing and adaptive downstream molecular responses in central neurons.


Journal of Cerebral Blood Flow and Metabolism | 2010

Transplantation of human neural precursor cells in Matrigel scaffolding improves outcome from focal cerebral ischemia after delayed postischemic treatment in rats

Kunlin Jin; XiaoOu Mao; Lin Xie; Veronica Galvan; Bin Lai; Yaoming Wang; Olivia Gorostiza; Xiaomei Wang; David A. Greenberg

Transplantation of neural cells is a potential approach for stroke treatment, but disruption of tissue architecture may limit transplant efficacy. One strategy for enhancing the ability of transplants to restore brain structure and function is to administer cells together with biomaterial scaffolding. We electrocoagulated the distal middle cerebral artery in adult rats and, 3 weeks later, injected one of the following into the infarct cavity: artificial cerebrospinal fluid, Matrigel scaffolding, human embryonic stem cell-derived neuronal precursor cells, scaffolding plus cells, or cells cultured in and administered together with scaffolding. Five weeks after transplantation, the latter two groups showed ∼50% and ∼60% reductions, respectively, in infarct cavity volume. Rats given cells cultured in and administered together with scaffolding also showed (1) survival and neuronal differentiation of transplanted cells shown by immunostaining for neuronal marker proteins and cleaved caspase-3, and by patch-clamp recording, 8 weeks after transplantation and (2) improved outcome on tests of sensorimotor and cognitive functions, 4 to 9 weeks after transplantation. These results indicate that transplantation of human neural cells together with biomaterial scaffolding has the potential to improve the outcome from stroke, even when treatment is delayed for several weeks after the ischemic event.


Journal of Cerebral Blood Flow and Metabolism | 2009

Involvement of Notch1 Signaling in Neurogenesis in the Subventricular Zone of Normal and Ischemic Rat Brain in Vivo

Xiaomei Wang; XiaoOu Mao; Lin Xie; David A. Greenberg; Kunlin Jin

The Notch1 signaling pathway is regarded as one of the main regulators of neural stem cell behavior during development, but its role in the adult brain is less well understood. We found that Notch1 was mainly expressed in doublecortin (DCX)-positive cells corresponding to newborn neurons, whereas the Notch1 ligand, Jagged1, was predominantly expressed in glial fibrillary acidic protein (GFAP)-positive astrocytic cells in the subventricular zone (SVZ) of the normal adult brain. These findings were confirmed by conditional depletion of DCX-positive cells in transgenic mice carrying herpes simplex virus thymidine kinase (HSV-TK) under the control of the DCX promoter. In addition, the activated form of Notch1 (Notch intracellular domain, NICD) and its downstream transcriptional targets, Hes1 and sonic hedgehog (Shh), were also expressed in SVZ cells. Increased activation of Notch1 signaling increased SVZ cell proliferation, whereas inhibiting Notch1 signaling resulted in a reduction of proliferating cells in the SVZ. Levels of NICD, Hes1, and Shh were increased in the SVZ at 4 and 24 h after focal cerebral ischemia. Finally, ischemia-induced cell proliferation in the SVZ was blocked by inhibition of the Notch1 signaling pathway, suggesting that Notch1 signaling may have a key role in normal adult and ischemia-induced neurogenesis.


Journal of Immunology | 2014

mTOR Signaling Inhibition Modulates Macrophage/Microglia-Mediated Neuroinflammation and Secondary Injury via Regulatory T Cells after Focal Ischemia

Luokun Xie; Fen Sun; Jixian Wang; XiaoOu Mao; Lin Xie; Shao-Hua Yang; Dong-Ming Su; James W. Simpkins; David A. Greenberg; Kunlin Jin

Signaling by the mammalian target of rapamycin (mTOR) plays an important role in the modulation of both innate and adaptive immune responses. However, the role and underlying mechanism of mTOR signaling in poststroke neuroinflammation are largely unexplored. In this study, we injected rapamycin, a mTOR inhibitor, by the intracerebroventricular route 6 h after focal ischemic stroke in rats. We found that rapamycin significantly reduced lesion volume and improved behavioral deficits. Notably, infiltration of γδ T cells and granulocytes, which are detrimental to the ischemic brain, was profoundly reduced after rapamycin treatment, as was the production of proinflammatory cytokines and chemokines by macrophages and microglia. Rapamycin treatment prevented brain macrophage polarization toward the M1 type. In addition, we also found that rapamycin significantly enhanced anti-inflammation activity of regulatory T cells (Tregs), which decreased production of proinflammatory cytokines and chemokines by macrophages and microglia. Depletion of Tregs partially elevated macrophage/microglia-induced neuroinflammation after stroke. Our data suggest that rapamycin can attenuate secondary injury and motor deficits after focal ischemia by enhancing the anti-inflammation activity of Tregs to restrain poststroke neuroinflammation.


Brain Research | 2011

Effect of human neural precursor cell transplantation on endogenous neurogenesis after focal cerebral ischemia in the rat.

Kunlin Jin; Lin Xie; XiaoOu Mao; Maeve B. Greenberg; Alexander Moore; Botao Peng; Rose B. Greenberg; David A. Greenberg

Little is known about the relationship between neuronal cell transplantation and endogenous neurogenesis after experimental stroke. We found previously that transplantation of neuronal precursors derived from BG01 human embryonic stem cells reduced infarct volume and improved behavioral outcome after distal middle cerebral artery occlusion (MCAO) in rats. In this study, transplantation was performed 14 days after distal MCAO and doublecortin (Dcx)-expressing cells in the subventricular zone (SVZ) and subgranular zone of dentate gyrus (SGZ) were counted 60 days post-transplant. Transplantation increased neurogenesis (Dcx expression) in ipsilateral SVZ, but not in contralateral SVZ or either SGZ, in both young adult (3-month-old) and aged (24-month-old) rats. These findings suggest that cell-based therapy for stroke may be associated with changes in endogenous adaptive processes, including neurogenesis.


Neuroscience | 2000

Induction of vascular endothelial growth factor receptors and phosphatidylinositol 3'-kinase/Akt signaling by global cerebral ischemia in the rat

Kunlin Jin; XiaoOu Mao; Tetsuya Nagayama; P.C Goldsmith; David A. Greenberg

Vascular endothelial growth factor is an angiogenic peptide that binds to tyrosine kinase receptors on target cells to activate signal transduction pathways involving phosphatidylinositol 3-kinase and the serine-threonine protein kinase, Akt. To determine whether this signaling pathway is activated in cerebral ischemia, we examined the expression of vascular endothelial growth factor receptors 1 and 2, and phosphatidylinositol 3-kinase-activated phospho-Akt, in the cerebral cortex and hippocampus following transient global cerebral ischemia in the rat. Western blot analysis and immunocytochemistry demonstrated induction of vascular endothelial growth factor receptor 1 and 2 expression, and of anti-phosphatidylinositol 3-kinase-immunoprecipitated phospho-Akt, in vulnerable regions of the cortex and hippocampus after 15 min of global ischemia and 4-72 h of reperfusion. These findings demonstrate that vascular endothelial growth factor receptors and receptor-coupled signal transduction pathways are induced in ischemic brain in vivo, and could therefore participate in endogenous neuroprotective responses to ischemia.


PLOS ONE | 2012

Conditional Depletion of Neurogenesis Inhibits Long-Term Recovery after Experimental Stroke in Mice

Xiaomei Wang; XiaoOu Mao; Lin Xie; Fen Sun; David A. Greenberg; Kunlin Jin

We reported previously that ablation of doublecortin (DCX)-immunopositive newborn neurons in mice worsens anatomical and functional outcome measured 1 day after experimental stroke, but whether this effect persists is unknown. We generated transgenic mice that express herpes simplex virus thymidine kinase under control of the DCX promoter (DCX-TK transgenic mice). DCX-expressing and recently divided cells in the rostral subventricular zone (SVZ) and hippocampus of DCX-TK transgenic mice, but not wild-type mice, were specifically depleted after ganciclovir (GCV) treatment for 14 days. Focal cerebral ischemia was induced by permanent distal middle cerebral artery occlusion (MCAO) on day 14 of vehicle or GCV treatment, and mice were killed 12 weeks after MCAO. Infarct volume was significantly increased and neurologic deficits were more severe in GCV- compared to vehicle-treated DCX-TK transgenic mice at first 8 weeks, after depletion of DCX- and bromodeoxyuridine-immunoreactive cells in the SVZ and dentate gyrus following focal ischemia. Our results indicate that endogenous neurogenesis in a critical period following experimental stroke influences the course of long-term recovery.


Journal of Neurotrauma | 2013

Neurogenesis in Adult Human Brain after Traumatic Brain Injury

Weiming Zheng; Qichuan ZhuGe; Ming Zhong; Gourong Chen; Bei Shao; Hong Wang; XiaoOu Mao; Lin Xie; Kunlin Jin

While much work has been conducted regarding the neurogenesis response to traumatic brain injury (TBI) in rodents, it remains largely unknown whether neurogenesis in adult human brain also responds to TBI in a similar manner. Here, we performed immunocytochemistry on 11 brain specimens from patients with traumatic brain injury, who underwent surgical intervention. We found that expression of neural stem/progenitor cell (NSC) protein markers, including DCX, TUC4, PSA-NCAM, SOX2 and NeuroD, was increased in the perilesional cortex of human brain after TBI compared to that of normal brain. Confocal images showed that these NSC proteins were expressed in one single cell. We also found that proliferative markers were expressed in NSC protein-positive cells after TBI, and the number of proliferative NSCs was significantly increased after TBI. Our data suggest that TBI may also induce neurogenesis in human brain.


Journal of Cerebral Blood Flow and Metabolism | 2001

Fas (CD95) May Mediate Delayed Cell Death in Hippocampal CA1 Sector after Global Cerebral Ischemia

Kunlin Jin; Steven H. Graham; XiaoOu Mao; Tetsuya Nagayama; Roger P. Simon; David A. Greenberg

Cell death–regulatory genes like caspases and bcl-2 family genes are involved in delayed cell death in the CA1 sector of hippocampus after global cerebral ischemia, but little is known about the mechanisms that trigger their expression. The authors found that expression of Fas and Fas-ligand messenger ribonucleic acid and protein was induced in vulnerable CA1 neurons at 24 and 72 hours after global ischemia. Fas-associating protein with a novel death domain (FADD) also was upregulated and immunoprecipitated and co-localized with Fas. Caspase-10 was activated and interacted with FADD protein to an increasing extent as the duration of ischemia increased. Moreover, caspase-10 co-localized with both FADD and caspase-3. These findings suggest that Fas-mediated death signaling may play an important role in signaling hippocampal neuronal death in CA1 after global cerebral ischemia.


Journal of Cerebral Blood Flow and Metabolism | 2008

Neurogenesis after primary intracerebral hemorrhage in adult human brain.

Jianfeng Shen; Lin Xie; XiaoOu Mao; Yongqing Zhou; Renya Zhan; David A. Greenberg; Kunlin Jin

Neurogenesis occurs in discrete regions of normal brains of adult mammals including humans, and is induced in response to brain injury and neurodegenerative disease. Whether intracerebral hemorrhage can also induce neurogenesis in human brain is unknown. Specimens were obtained from patients with primary intracerebral hemorrhage undergoing surgical evacuation of an intracerebral hematoma, and evaluated by two-photon laser scanning confocal microscopy. We found that neural stem/progenitor cell-specific protein markers were expressed in cells located in the perihematomal regions of the basal ganglia and parietal lobe of the adult human brain after primary intracerebral hemorrhage (n = 5). Cells in this region also expressed cell proliferation markers, which colocalized to the same cells that expressed neural stem/progenitor cell-specific proteins. Our data suggest that intracerebral hemorrhage induces neurogenesis in the adult human brain.

Collaboration


Dive into the XiaoOu Mao's collaboration.

Top Co-Authors

Avatar

Kunlin Jin

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

David A. Greenberg

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lin Xie

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Fen Sun

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaomei Wang

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Ming Zhong

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Botao Peng

Buck Institute for Research on Aging

View shared research outputs
Researchain Logo
Decentralizing Knowledge