Network


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

Hotspot


Dive into the research topics where Ran-Sook Woo is active.

Publication


Featured researches published by Ran-Sook Woo.


Molecular Pharmacology | 2005

Mefenamic acid shows neuroprotective effects and improves cognitive impairment in in vitro and in vivo Alzheimer's disease models.

Yuyoung Joo; Hye-Sun Kim; Ran-Sook Woo; Cheol Hyoung Park; Ki-Young Shin; Jean-Pyo Lee; Keun-A Chang; Seonghan Kim; Yoo-Hun Suh

Nonsteroidal anti-inflammatory drugs (NSAIDs) exert anti-inflammatory, analgesic, and antipyretic activities and suppress prostaglandin synthesis by inhibiting cyclooxygenase, an enzyme that catalyzes the formation of prostaglandin precursors from arachidonic acid. Epidemiological observations indicate that the long-term treatment of patients suffering from rheumatoid arthritis with NSAIDs results in reduced risk and delayed onset of Alzheimers disease. In this study, we investigated the therapeutic potential for Alzheimers disease of mefenamic acid, a commonly used NSAID that is a cyclooxygenase-1 and 2 inhibitor with only moderate anti-inflammatory properties. We found that mefenamic acid attenuates the neurotoxicities induced by amyloid β peptide (Aβ)1–42 treatment and the expression of a Swedish double mutation (KM595/596NL) of amyloid precursor protein (Swe-APP) or the C-terminal fragments of APP (APP-CTs) in neuronal cells. We also show that mefenamic acid decreases the production of the free radical nitric oxide and reduces cytochrome c release from mitochondria induced by Aβ1–42, Swe-APP, or APP-CTs in neuronal cells. In addition, mefenamic acid up-regulates expression of the antiapoptotic protein Bcl-XL. Moreover, our study demonstrates for the first time that mefenamic acid improves learning and memory impairment in an Aβ1–42-infused Alzheimers disease rat model. Taking these in vitro and in vivo results together, our study suggests that mefenamic acid could be used as a therapeutic agent in Alzheimers disease.


Neuroscience | 2012

Neuregulin-1 protects against neurotoxicities induced by Swedish amyloid precursor protein via the ErbB4 receptor

Ran-Sook Woo; Ji-Hye Lee; Hye Sun Kim; C.-H. Baek; Dae-Yong Song; Yoo-Hun Suh; Tai-Kyoung Baik

Neuregulin-1 (NRG1) plays an important role in the development and plasticity of the brain and exhibits potent neuroprotective properties. However, little information on its role in Alzheimers disease (AD) is known. The neuroprotective effect and mechanisms of NRG1 in SH-SY5Y cells overexpressing the Swedish mutant form of amyloid precursor protein (Swe-APP) and primary cortical neuronal cells treated with amyloid beta peptide(1-42) (Aβ(1-42)) were investigated in this study. NRG1 attenuated Swe-APP- or Aβ(1-42)-induced lactate dehydrogenase (LDH) release in a concentration-dependent manner. The mitigating effects of NRG1 on neuronal cell death were blocked by ErbB4 inhibition, a key NRG1 receptor, which suggests a role of ErbB4 in the neuroprotective function of NRG1. Moreover, NRG1 reduced the number of Swe-APP- and Aβ(1-42)-induced TUNEL-positive SH-SY5Y cells and primary cortical neurons, respectively. NRG1 reduced the accumulation of reactive oxygen species and attenuated Swe-APP-induced mitochondrial membrane potential loss. NRG1 also induced the upregulation of the expression of the anti-apoptotic protein, Bcl-2, and decreased caspase-3 activation. Collectively, our results demonstrate that NRG1 exerts neuroprotective effects via the ErbB4 receptor, which suggests the neuroprotective potential of NRG1 in AD.


Neuroscience Research | 2011

Role of activating transcription factor 3 in ischemic penumbra region following transient middle cerebral artery occlusion and reperfusion injury.

Dae-Yong Song; Kyoung-Min Oh; Ha-Nul Yu; Chae-Ri Park; Ran-Sook Woo; Sung-Sam Jung; Tai-Kyoung Baik

The activating transcription factor 3 (ATF3) is expressed by various types of cellular insults. It has been suggested to serve diverse functions in both cellular survival and death signal cascades, but the exact role of ATF3 in brain ischemia is little known so far. Thus, the authors examined the expression pattern of ATF3 following middle cerebral artery occlusion (MCAO) and reperfusion injury. At 1-2 days after MCAO and reperfusion injury, numerous number of ATF3-immunoreacitive (-ir) nuclei was observed in the ipsilateral peri-infarct cortex, but declined rapidly at 3 days. Almost all ATF3-ir nuclei were co-localized with NeuN-ir neurons. Neither GFAP- nor OX42-ir neuroglia were co-localized with ATF3. Double labeling of Fluoro-Jade B with ATF3 showed that ATF3-ir nuclei mismatched with Fluoro-Jade B-ir neurons. To further examine the role of ATF3 in ischemic peri-infarct regions, double immunofluorescent labeling of ATF3/caspase 3, ATF3/Bcl-xl, and ATF3/HSP27 was conducted. Semiquantitive estimation showed that about 15% of ATF3-ir neurons also expressed caspase 3. However, about only 0.4% and 2.6% of ATF3-ir neurons were double-stained with Bcl-xl and Hsp27, respectively. Consequently, it would be suggested that ATF3 seem to play an important role in caspase-dependent neuronal apoptotic signal transduction pathways caused by focal cerebral ischemia and reperfusion injury.


Anatomy & Cell Biology | 2010

Expression of ErbB4 in the apoptotic neurons of Alzheimer's disease brain

Ran-Sook Woo; Ji-Hye Lee; Ha-Nul Yu; Dae-Yong Song; Tai-Kyoung Baik

Neuregulin-1 (NRG1) signaling participates in the synaptic plasticity, maintenance or regulation of adult brain. Although ErbB4, a key NRG1 receptor, is expressed in multiple regions in the adult animal brain, little is known about its localization in Alzheimers disease (AD) brains. We previously reported that ErbB4 immunoreactivity showed regional difference in the hippocampus of age-matched control. In the present paper, immunohistochemical characterization of the distribution of ErbB4 receptor in the hippocampus relative to pathology staging were performed in age-matched control (Braak stage 0, n=6) and AD (Braak stage I/V, n=10). Here, we found that ErbB4 immunoreactivity was significantly increased in apoptotic hippocampal pyramidal neurons in the brains of AD patients, compared to those of age-matched control subjects. In AD brains, ErbB4 immunoreactivity was demonstrated to colocalize with the apoptotic signal Bax in apoptotic hippocampal pyramidal neurons. These results suggest that up-regulation of ErbB4 immunoreactivity in apoptotic neuron may involve in the progression of pathology of AD.


Neuroscience | 2016

Adolescent mice show anxiety- and aggressive-like behavior and the reduction of long-term potentiation in mossy fiber-CA3 synapses after neonatal maternal separation.

S.Y. Shin; Seung Ho Han; Ran-Sook Woo; S.H. Jang; Sun Seek Min

Exposure to maternal separation (MS) during early life is an identified risk factor for emotional disorders such as anxiety and depression later in life. This study investigated the effects of neonatal MS on the behavior and long-term potentiation (LTP) as well as basic synaptic transmission at hippocampal CA3-CA1 and mossy fiber (MF)-CA3 synapses in adolescent mice for 19days. When mice were adolescents, we measured depression, learning, memory, anxious and aggressive behavior using the forced swimming test (FST), Y-maze, Morris water maze (MWM), elevated plus maze (EPM), three consecutive days of the open field test, the social interaction test, the tube-dominance test and the resident-intruder test. The results showed that there was no difference in FST, Y-maze, and MWM performance. However, MS mice showed more anxiety-like behavior in the EPM test and aggressive-like behavior in the tube-dominance and resident-intruder tests. In addition, the magnitude of LTP and release probability in the MF-CA3 synapses was reduced in the MS group but not in the CA3-CA1 synapse. Our results indicate that early life stress due to MS may induce anxiety- and aggressive-like behavior during adolescence, and these effects are associated with synaptic plasticity at the hippocampal MF-CA3 synapses.


PLOS ONE | 2014

Neuritin Attenuates Cognitive Function Impairments in Tg2576 Mouse Model of Alzheimer's Disease

Yoori Choi; Ki-Hwan Lee; Junghwa Ryu; Hyoun Geun Kim; A Young Jeong; Ran-Sook Woo; Jun-Ho Lee; Jin Won Hyun; Seokyung Hahn; Joung-Hun Kim; Hye-Sun Kim

Neuritin, also known as CPG15, is a neurotrophic factor that was initially discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin plays multiple roles in the process of neural development and synaptic plasticity, although its binding receptor(s) and downstream signaling effectors remain unclear. In this study, we found that the cortical and hippocampal expression of neuritin is reduced in the brains of Alzheimers disease (AD) patients and demonstrated that viral-mediated expression of neuritin in the dentate gyrus of 13-month-old Tg2576 mice, an AD animal model, attenuated a deficit in learning and memory as assessed by a Morris water maze test. We also found that neuritin restored the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neuron cultures prepared from Tg2576 mice. It was also shown that viral-mediated expression of neuritin in the dentate gyrus of 7-week-old Sprague-Dawley rats increased neurogenesis in the hippocampus. Taken together, our results demonstrate that neuritin restores the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neurons from Tg2576 neurons, and also attenuates cognitive function deficits in Tg2576 mouse model of AD, suggesting that neuritin possesses a therapeutic potential for AD.


Journal of Biological Chemistry | 2015

Neuregulin 1 Controls Glutamate Uptake by Up-regulating Excitatory Amino Acid Carrier 1 (EAAC1)

Ha-Nul Yu; Woo-Kyu Park; Ki-Hoan Nam; Dae-Yong Song; Hye-Sun Kim; Tai-Kyoung Baik; Ran-Sook Woo

Background: NRG1 and its receptor ErbB4 are schizophrenia susceptibility genes. Results: NRG1 induced the up-regulation of EAAC1 with an increase in glutamate uptake. Conclusion: NRG1/ErbB4 signaling influences glutamate uptake by increasing the EAAC1 protein level. Significance: These results contribute to understanding of a possible mechanism of NRG1/ErbB4 signaling that may be linked to the neural circuitry disruption in schizophrenia. Neuregulin 1 (NRG1) is a trophic factor that is thought to have important roles in the regulating brain circuitry. Recent studies suggest that NRG1 regulates synaptic transmission, although the precise mechanisms remain unknown. Here we report that NRG1 influences glutamate uptake by increasing the protein level of excitatory amino acid carrier (EAAC1). Our data indicate that NRG1 induced the up-regulation of EAAC1 in primary cortical neurons with an increase in glutamate uptake. These in vitro results were corroborated in the prefrontal cortex (PFC) of mice given NRG1. The stimulatory effect of NRG1 was blocked by inhibition of the NRG1 receptor ErbB4. The suppressed expression of ErbB4 by siRNA led to a decrease in the expression of EAAC1. In addition, the ablation of ErbB4 in parvalbumin (PV)-positive neurons in PV-ErbB4−/− mice suppressed EAAC1 expression. Taken together, our results show that NRG1 signaling through ErbB4 modulates EAAC1. These findings link proposed effectors in schizophrenia: NRG1/ErbB4 signaling perturbation, EAAC1 deficit, and neurotransmission dysfunction.


Journal of Chemical Neuroanatomy | 2015

Changes of gene expression of Gal3, Hsp27, Lcn2, and Timp1 in rat substantia nigra following medial forebrain bundle transection using a candidate gene microarray

Yoon-Jung Choy; Sung-Young Hong; Sang-Jin Pack; Ran-Sook Woo; Tai-Kyoung Baik; Dae-Yong Song

Neuroinflammation is an early event and important contributor to the pathobiology of neurodegenerative diseases. Neuroglia, especially microglia, are a major central nervous system population that can modulate neuroinflammation. To determine potential key molecules in this process, we employed microarray analysis in the substantia nigra (SN) following medial forebrain bundle (MFB) transection and analyzed the temporal expression profiles of candidate genes implicated in neuroglial activation and functional maturation. The DNA microarray analyzed, 8913 probes. Sixty nine genes were up-regulated and 11 genes were down-regulated at least twofold compared to normal control. Of the 80 genes, 23 were related to cell metabolism, 3 related to apoptosis, 27 related to immunity. Among them, 4 genes (Galectin 3, Heat shock protein 27, Lipocalin 2, Tissue inhibitory metalloproteinase 1) seemed to be related to the neuroglial function. The candidate genes were subjected to quantitative real-time PCR, Western blotting, and immunohistochemical approaches. Expression changes similar to the microarray were evident. In a double immunofluorescence assay, Galectin 3 almost completely co-localized with OX6-positive activated microglia, and Heat shock protein 27 mainly co-localized with glial fibrillary acidic protein (GFAP) positive astrocytes. Lipocalin 2, except for a few matches of GFAP positive astrocytes, did not co-localized with any of neuroglial markers. This is the first study to evaluate gene expression changes in the SN following MFB transection, which has been used as a parkinsonian animal model. Several candidate genes with potential roles in neuroglial activation and functional maturation were identified. The molecular significance of the candidate genes in neuroglial activation and neuroinflammation remains unclear.


International Journal of Neuroscience | 2012

Plasminogen Activator Inhibitor-1 Promotes Synaptogenesis and Protects Against Aβ 1-42 -Induced Neurotoxicity in Primary Cultured Hippocampal Neurons

Harim Cho; Yuyoung Joo; Seonghan Kim; Ran-Sook Woo; Sang Hyung Lee; Hye-Sun Kim

ABSTRACT Plasminogen activator inhibitor-1 (PAI-1) is a soluble factor that is released from astrocytes, the most abundant type of glial cell in the brain. PAI-1 was initially identified as inhibiting two types of plasminogen activators, that is, tissue-type plasminogen and urokinase activators that are known to lead to the proteolytic degradation of the extracellular matrix. Recently, PAI-1 was reported to mediate the neuroprotective activity of transforming growth factor-β1 against N-methyl-D-aspartate receptor-mediated excitotoxicity and to be involved in angiogenesis following ischemic stroke, independently of the effects via the inhibition of tissue-type plasminogen and urokinase-type plasminogen activators. In this study, we examined whether PAI-1 influences synaptogenesis and neurotoxicity induced by amyloid beta peptide1-42 (Aß1-42) in rat primary hippocampal neurons. Using immunostaining, treatment with PAI-1 for 24 h was found to significantly upregulate synaptophysin, postsynaptic density-95, and the polysialylated form of neural cell adhesion molecule, compared to treatment with vehicle alone. In addition, PAI-1 has neuroprotective effects against Aβ1-42-induced cytotoxicity in rat primary cultured hippocampal neurons. Taken together, our results suggest that PAI-1 has therapeutic potential in Alzheimers disease by promoting synaptogenesis and by demonstrating neuroprotective effects against Aβ1-42-oligomer-induced neurotoxicity in rat primary cultured hippocampal neurons.


Journal of Pharmacy and Pharmacology | 2016

Blocking the phosphatidylinositol 3-kinase pathway inhibits neuregulin-1-mediated rescue of neurotoxicity induced by Aβ1–42

Tai-Kyoung Baik; Young-Jung Kim; Se-Mi Kang; Dae-Yong Song; Sun Seek Min; Ran-Sook Woo

Neuregulin‐1 (NRG1) has an important role in both the development and the plasticity of the brain as well as neuroprotective properties. In this study, we investigated the downstream pathways of NRG1 signalling and their role in the prevention of Aβ1–42‐induced neurotoxicity.

Collaboration


Dive into the Ran-Sook Woo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hye-Sun Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoo-Hun Suh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuyoung Joo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junghwa Ryu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge