Network


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

Hotspot


Dive into the research topics where Hae Sook Noh is active.

Publication


Featured researches published by Hae Sook Noh.


The FASEB Journal | 2003

Flavonoid wogonin from medicinal herb is neuroprotective by inhibiting inflammatory activation of microglia

Heasuk Lee; Young Ok Kim; Hocheol Kim; Sun Yeou Kim; Hae Sook Noh; Sang Soo Kang; Gyeong Jae Cho; Wan Sung Choi; Kyoungho Suk

Wogonin (5,7‐dihydroxy‐8‐methoxyflavone), a flavonoid originated from the root of a medicinal herb Scutellaria baicalensis Georgi, has been previously shown to have anti‐inflammatory activities in various cell types including macrophages. In this work, we have found that wogonin is a potent neuroprotector from natural source. Wogonin inhibited inflammatory activation of cultured brain microglia by diminishing lipopolysaccharide‐induced tumor necrosis factor‐α (TNF‐α), interleukin‐1β, and nitric oxide (NO) production. Wogonin inhibited NO production by suppressing inducible NO synthase (iNOS) induction and NF‐κB activation in microglia. Inhibition of inflammatory activation of microglia by wogonin led to the reduction in microglial cytotoxicity toward cocultured PC12 cells, supporting a neuroprotective role for wogonin in vitro. The neuroprotective effect of wogonin was further demonstrated in vivo using two experimental brain injury models; transient global ischemia by four‐vessel occlusion and excitotoxic injury by systemic kainate injection. In both animal models, wogonin conferred neuroprotection by attenuating the death of hippocampal neurons, and the neuroprotective effect was associated with inhibition of the inflammatory activation of microglia. Hippocampal induction of inflammatory mediators such as iNOS and TNF‐α was reduced by wogonin in the global ischemia model, and microglial activation was markedly down‐regulated by wogonin in the kainate injection model as judged by microglia‐specific isolectin B4 staining. Taken together, our results indicate that wogonin exerts its neuroprotective effect by inhibiting microglial activation, which is a critical component of pathogenic inflammatory responses in neurodegenerative diseases. The current study emphasizes the importance of medicinal herbs and their constituents as an invaluable source for the development of novel neuroprotective drugs.


Epilepsy Research | 2003

The protective effect of a ketogenic diet on kainic acid-induced hippocampal cell death in the male ICR mice.

Hae Sook Noh; Yoon Sook Kim; Hee Po Lee; Ki Myung Chung; Dong Wook Kim; Sang Soo Kang; Gyeong Jae Cho; Wan Sung Choi

This study was designed to evaluate the antiapoptotic effects of a ketogenic diet (KD) through histological (cresyl violet staining, TUNEL staining and immunohistochemistry) and behavioral studies using kainic acid (KA, 25mg/kg i.p.)-induced seizures in male ICR mice. KA-induced seizure in rodents is widely used as an experimental model for human temporal lobe epilepsy because of their behavioral and pathological similarities. A KA-induced seizure causes neuronal damage in hippocampal pyramidal neurons and involves a caspase-3-mediated apoptotic pathway. In this study, the seizure onset time of the KD-fed group was delayed compared to that of the group fed a normal diet (ND) after a systemic KA injection. Histological studies revealed that KA caused pyknosis in most of the hippocampal areas in the ND-fed group, however, well-preserved pyramidal neurons were detected in the hippocampus of mice that had been on KD for 1 month, which began on postnatal day 21. The number of TUNEL-positive cells and caspase-3-positive cells in the hippocampus of the KD-fed group was lower than that of the ND-fed group. These findings indicate that KD has an antiepileptic effect via a neuroprotective action that involves the inhibition of caspase-3-mediated apoptosis of hippocampal neurons.


Journal of Neuroscience Research | 2006

Acetoacetate protects neuronal cells from oxidative glutamate toxicity

Hae Sook Noh; Young-Sool Hah; Rashidova Nilufar; Jaehee Han; Jae-Hwan Bong; Sang Soo Kang; Gyeong Jae Cho; Wan Sung Choi

Glutamate cytotoxicity contributes to neuronal degeneration in many central nervous system (CNS) diseases, such as epilepsy and ischemia. We previously reported that a high‐fat and low‐carbohydrate diet, the ketogenic diet (KD), protects against kainic acid‐induced hippocampal cell death in mice. We hypothesized based on these findings that ketosis resulting from KD might inhibit glutamate cytotoxicity, resulting in inhibition of hippocampal neuronal cell death. Therefore, we investigated the role of ketone bodies [acetoacetate (AA) and β‐hydroxybutyrate (β‐OHB)] both in a mouse hippocampal cell line (HT22) and in rat primary hippocampal neurons. As a result, we found that pretreatment with 5 mM lithium AA and 4 mM Na β‐OHB protected the HT22 hippocampal cell line and primary hippocampal neuronal culture against 5 mM glutamate toxicity and that up to 2 hr of pretreatment with 5 mM AA had a protective effect against 5 mM glutamate toxicity in the HT22 cell line. Pretreatment with 5 mM AA decreased ROS production of HT22 cell line at 2 and 8 hr exposure of glutamate, and it decreased the appearance of annexin V‐positive HT22 cells, which are indicative of an early stage of apoptosis, and propidium iodide‐positive HT22 cells, which are indicative of necrosis.


Neuroscience Letters | 2010

Autophagy protects the rotenone-induced cell death in α-synuclein overexpressing SH-SY5Y cells

Shorafidinkhuja Dadakhujaev; Hae Sook Noh; Eun Joo Jung; Joon Yung Cha; Seon Mi Baek; Ji Hye Ha; Deok Ryong Kim

Loss of dopaminergic cells induced by alpha-synuclein accumulation in substantia nigra causes the development of Parkinsons disease (PD). To date, although autophagy has been implicated in the pathology of PD, the molecular mechanism is still unclear. To study the role of autophagy in PD pathogenesis, we established stable SH-SY5Y cell lines overexpressing wild-type or mutant alpha-synuclein proteins (A30P or A53T). Overexpression of mutant alpha-synuclein induced some protein aggregates and cell death in the absence of drug. LC3-II protein, a critical marker for autophagy, was produced in an autophagy-dependent manner. The rotenone-induced cell death was interrupted by autophagy stimulation. Autophagy activation also restored the mitochondrial membrane potential (MMP) impaired by rotenone in mutant alpha-synuclein expressing cells. Additionally, autophagy activation significantly relieved rotenone-induced ROS accumulation and HIF-1alpha expression in neuronal cells expressing mutant alpha-synuclein proteins. These findings indicate that autophagy plays an important scavenger role against harmful influence of toxic protein aggregates produced in rotenone-treated cells.


Brain Research | 2003

Suppression by ethanol of male reproductive activity.

Jin Hyun Kim; Hyun Joon Kim; Hae Sook Noh; Gu Seob Roh; Sang Soo Kang; Gyeong Jae Cho; Sang Kyu Park; Byung Ju Lee; Wan Sung Choi

Ethanol has been known to suppress reproductive activity in laboratory animals and humans through the inhibition of luteinizing hormone (LH) release by reduction of gonadotropin-releasing hormone (GnRH) secretion from the hypothalamus. There are, however, little data is available regarding the effect of ethanol on GnRH gene expression. Thus, the present study was designed to evaluate the effect of ethanol on GnRH gene expression and reproductive activity at all levels of the hypothalamus-pituitary-gonad (HPG) axis simultaneously. To this end, ethanol (3 g/kg i.p., 15% v/v in saline) was administered to adult male rats for 10 days. Serum levels of LH and testosterone were significantly decreased by ethanol. Using Northern blot analysis and in situ hybridization, the present study showed the reduction in GnRH mRNA levels in the hypothalamus by prolonged ethanol administration. The content of LH in the anterior pituitary was also significantly reduced by ethanol. In addition, steroidogenic acute regulatory protein (StAR) mRNA levels were significantly decreased by ethanol, suggesting a cause for the reduced production of testosterone under this condition. These results indicate that ethanol affects the HPG axis at all the levels. Especially, suppressed GnRH mRNA levels in the hypothalamus of ethanol-treated rats strongly demonstrated that hypothalamus is the major action site of ethanol on the HPG axis. Decreased serum LH level may affect the steroidogenesis in the testis, at least in part, through the inhibition of StAR gene expression that induces part of dysfunctions of reproductive activity.


Cancer Letters | 2012

Cathepsin D inhibits oxidative stress-induced cell death via activation of autophagy in cancer cells

Young-Sool Hah; Hae Sook Noh; Ji Hye Ha; Jin Sook Ahn; Jong Ryeal Hahm; Hee Young Cho; Deok Ryong Kim

Cathepsin D (CatD), a lysosomal aspartic protease, plays an essential role in tumor progression and apoptosis. However, the function of CatD in cell death is not yet fully understood. In this study, we identified CatD as one of up-regulated proteins in human malignant glioblastoma M059J cells that lack the catalytic subunit of DNA-PK compared with its isogenic M059K cells with normal DNA-PK activity. M059J cells were relatively more resistant to genotoxic stress than M059K cells. Overexpression of wild-type CatD but not catalytically inactive mutant CatD (D295N) inhibited H(2)O(2)-induced cell death in HeLa cells. Furthermore, knockdown of CatD expression abolished anti-apoptotic effect by CatD in the presence of H(2)O(2). Interestingly, high expression of CatD in HeLa cells significantly activated autophagy: increase of acidic autophagic vacuoles, LC3-II formation, and GFP-LC3 puncta. These results suggest that CatD can function as an anti-apoptotic mediator by inducing autophagy under cellular stress. In conclusion, inhibition of autophagy could be a novel strategy for the adjuvant chemotherapy of CatD-expressing cancers.


Molecules and Cells | 2010

Propofol protects the autophagic cell death induced by the ischemia/reperfusion injury in rats

Hae Sook Noh; Il Woo Shin; Ji Hye Ha; Young-Sool Hah; Seon Mi Baek; Deok Ryong Kim

Autophagy has been implicated in cardiac cell death during ischemia/reperfusion (I/R). In this study we investigated how propofol, an antioxidant widely used for anesthesia, affects the autophagic cell death induced by the myocardial I/R injury. The infarction size in the myocardium was dramatically reduced in rats treated with propofol during I/R compared with untreated rats. A large number of autophagic vacuoles were observed in the cardiomyocytes of I/R-injured rats but rarely in I/R-injured rats treated with propofol. While LC3-II formation, an autophagy marker, was up-regulated in the I/R-injured myocardium, it was significantly down-regulated in the myocardial tissues of I/R-injured and propofol-treated rats. Moreover, propofol inhibited the I/R-induced expression of Beclin-1, and it accelerated phosphorylation of mTOR during I/R and Beclin-1/Bcl-2 interaction in cells, which indicates that it facilitates the inhibitory pathway of autophagy. These data suggest that propofol protects the autophagic cell death induced by the myocardial I/R injury.


Journal of Neuroscience Research | 2006

Ketogenic diet protects the hippocampus from kainic acid toxicity by inhibiting the dissociation of bad from 14-3-3.

Hae Sook Noh; Yoon Sook Kim; Young Hee Kim; Jae Yoon Han; Chang Hwan Park; Ahn Ki Kang; Hee Suk Shin; Sang Soo Kang; Gyeong Jae Cho; Wan Sung Choi

The ketogenic diet (KD) is often effective for intractable epilepsy, but its antiepileptic mechanisms remain largely unknown. Within the cell death/survival pathway, Akt and its downstream protein Bad play an important role in kainic acid (KA)‐induced cell death. Therefore, we investigated the effects of a KD on KA‐induced changes in the Akt/Bad/14‐3‐3 signaling pathway by evaluating Akt, Bad, 14‐3‐3, and cleaved caspase‐3 expression levels as well as their relative interactions. Our results showed that a KD did not affect the expression levels of Akt, Bad, Bcl‐xL, Bax, and 14‐3‐3 but increased phospho‐Akt [serine 473; p‐Akt (Ser473)] and phospho‐Bad [serine 136; p‐Bad (Ser136)] expression levels as well as decreased cleaved caspase‐3 levels following a KA‐induced seizure in the hippocampus. Furthermore, we found that a KD increased the protein–protein interaction between 14‐3‐3 and p‐Bad (Ser136), which might be phosphorylated by p‐Akt (Ser473), and decreased interaction of Bad and Bcl‐xL. These results suggest that a KD might protect, at least partially, the hippocampus from KA‐induced cell death via inhibiting the dissociation of Bad from 14‐3‐3.


Brain Research | 2006

Increased nitric oxide caused by the ketogenic diet reduces the onset time of kainic acid-induced seizures in ICR mice.

Hae Sook Noh; Dong Wook Kim; Gyeong Jae Cho; Wan Sung Choi; Sang Soo Kang

Although the antiepileptic effects of the ketogenic diet (KD) are well documented, the mechanisms underlying this action remain obscure. Nitric oxide (NO) has long been thought to play a role in regulating seizures. However, the effects of the KD on endogenous NO production have not been characterized. Therefore, the present study was designed to examine the effect of the KD on endogenous NO production, as well as the precise role of NO in kainic acid (KA)-induced seizures, in male ICR mice. We first found that preadministration of the KD for 4 weeks increased endogenous NO generation in the hippocampus. We also demonstrated that the increase in NO induced by the KD resulted from increased neuronal NO synthase (nNOS) activity and exerted an antiepileptic effect on KA-induced seizures, based on the results of experiments using NOS-knockout mice and two NOS inhibitors, N-omega-nitro-L-arginine methyl ester (L-NAME) and 7-nitroindazole (7-NI). These data suggest that the antiepileptic effects of the KD might be mediated, at least in part, by increased NO levels in the hippocampus.


Brain Research | 2005

Ketogenic diet prevents clusterin accumulation induced by kainic acid in the hippocampus of male ICR mice

Hae Sook Noh; Dong Wook Kim; Sang Soo Kang; Gyeong Jae Cho; Wan Sung Choi

We investigated the effect of ketogenic diet (KD) on clusterin accumulation in the kainic acid (KA)-induced seizure model. Two days after KA administration, strong clusterin-like immunoreactivity (IR) was detected in the hippocampus in the normal diet (ND)-fed mice. But in the KD-fed mice, few clusterin-like IR was detected. These results indicate that KD has neuroprotective effects throughout diminishing nuclear clusterin accumulation that is involved in caspase-3 independent cell death mechanism.

Collaboration


Dive into the Hae Sook Noh's collaboration.

Top Co-Authors

Avatar

Sang Soo Kang

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Wan Sung Choi

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Gyeong Jae Cho

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Deok Ryong Kim

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Young-Sool Hah

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Ji Hye Ha

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Yoon Sook Kim

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jong Ryeal Hahm

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Seon Mi Baek

Gyeongsang National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge