Network


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

Hotspot


Dive into the research topics where Yoon-Seong Kim is active.

Publication


Featured researches published by Yoon-Seong Kim.


Experimental and Molecular Medicine | 2006

Microglia, major player in the brain inflammation : their roles in the pathogenesis of Parkinson’s disease

Yoon-Seong Kim; Tong H. Joh

Inflammation, a self-defensive reaction against various pathogenic stimuli, may become harmful self-damaging process. Increasing evidence has linked chronic inflammation to a number of neurodegenerative disorders including Alzheimers disease (AD), Parkinsons disease (PD), and multiple sclerosis. In the central nervous system, microglia, the resident innate immune cells play major role in the inflammatory process. Although they form the first line of defense for the neural parenchyma, uncontrolled activation of microglia may directly toxic to neurons by releasing various substances such as inflammatory cytokines (IL-1β, TNF-α, IL-6), NO, PGE and superoxide. Moreover, our recent study demonstrated that activated microglia phagocytose not only damaged cell debris but also neighboring intact cells. It further supports their active participation in self-perpetuating neuronal damaging cycles. In the following review, we discuss microglial responses to damaging neurons, known activators released from injured neurons and how microglia cause neuronal degeneration. In the last part, microglial activation and their role in PD are discussed in depth.


The Journal of Neuroscience | 2005

Matrix Metalloproteinase-3: A Novel Signaling Proteinase from Apoptotic Neuronal Cells That Activates Microglia

Yoon-Seong Kim; Sung Soo Kim; Jeong Je Cho; Dong Hee Choi; Onyou Hwang; Dong Hoon Shin; Hong Sung Chun; M. Flint Beal; Tong H. Joh

Microglial activation and inflammation are associated with progressive neuronal apoptosis in neurodegenerative human brain disorders. We sought to investigate molecular signaling mechanisms that govern activation of microglia in apoptotic neuronal degeneration. We report here that the active form of matrix metalloproteinase-3 (MMP-3) was released into the serum-deprived media (SDM) of PC12 cells and other media of apoptotic neuronal cells within 2-6 h of treatment of the cells, and SDM and catalytic domain of recombinant MMP-3 (cMMP-3) activated microglia in primary microglia cultures as well as BV2 cells, a mouse microglia cell line. Both SDM and cMMP-3 induced generation of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), IL-1β, and interleukin-1 receptor antagonist but not IL-12 and inducible nitric oxide synthase, which are readily induced by lipopolysaccharide, in microglia, suggesting that there is a characteristic pattern of microglial cytokine induction by apoptotic neurons. Neither glial cell line-derived neurotrophic factor nor anti-inflammatory cytokines, such as IL-10 and transforming growth factor-β1, were induced. SDM and cMMP-3 extensively released TNF-α from microglia and activated the nuclear factor-κB pathway, and these microglial responses were totally abolished by preincubation with an MMP-3 inhibitor, NNGH [N-isobutyl-N-(4-methoxyphenylsulfonyl)-glycylhydroxamic acid]. MMP-3-mediated microglial activation mostly depended on ERK (extracellular signal-regulated kinase) phosphorylation but not much on either JNK (c-Jun N-terminal protein kinase) or p38 activation. Conditioned medium of SDM- or cMMP-3-activated BV2 cells caused apoptosis of PC12 cells. These results strongly suggest that the distinctive signal of neuronal apoptosis is the release of active form of MMP-3 that activates microglia and subsequently exacerbates neuronal degeneration. Therefore, the release of MMP-3 from apoptotic neurons may play a major role in degenerative human brain disorders, such as Parkinsons disease.


Molecular Brain Research | 1999

Cultures of astrocytes and microglia express interleukin 18

Bruno Conti; Larry Park; Noel Y. Calingasan; Yoon-Seong Kim; Hocheol Kim; Youngmee Bae; Gary E. Gibson; Tong H. Joh

Interleukin 18 (IL-18 or interferon-gamma inducing factor) is a recently discovered pro-inflammatory cytokine and powerful stimulator of the cell-mediated immune response. IL-18 is produced by several sources including monocytes/macrophages, keratinocytes and the zona reticularis and zona fasciculata of the adrenal cortex. IL-18 occurs in brain but its cellular source in the CNS has never been investigated. The presence of IL-18 and its response to stimulation in the brain was tested with primary cultures of microglia, astrocytes and hippocampal neurons. IL-18 mRNA was present in astrocytes and microglia, but not in neurons. The endotoxin lipopolysaccharide (LPS) did not affect IL-18 in astrocytes, but LPS robustly increased IL-18 mRNA in microglia. IL-18 protein was constitutively expressed in astrocytes and induced in microglia by LPS. The levels of interleukin-1beta converting enzyme (ICE), an activating enzyme, and caspase 3 (CPP32), an inactivating enzyme, were assessed to investigate the presence of the appropriate processing enzymes in the cultured cells. ICE was present at constitutive levels in microglia and astrocytes suggesting that these cell types may produce and secrete matured IL-18. Active forms of CPP32 were not detectable in either cell type indicating the absence of a degradative pathway of IL-18. The present results demonstrate that microglia and astrocytes are sources of brain IL-18 and add a new member to the family of cytokines produced in the brain.


Journal of Biological Chemistry | 2004

Transglutaminase 2 induces nuclear factor-κB activation via a novel pathway in BV-2 microglia

Jongmin Lee; Yoon-Seong Kim; Dong-Hee Choi; Moon Suk Bang; Tai Ryoon Han; Tong H. Joh; Soo-Youl Kim

Transglutaminase 2 (TGase 2) expression is increased in inflammatory diseases. We demonstrated previously that inhibitors of TGase 2 reduce nitric oxide (NO) generation in a lipopolysaccharide (LPS)-treated microglial cell line. However, the precise mechanism by which TGase 2 promotes inflammation remains unclear. We found that TGase 2 activates the transcriptional activator nuclear factor (NF)-κB and thereby enhances LPS-induced expression of inducible nitric-oxide synthase. TGase 2 activates NF-κB via a novel pathway. Rather than stimulating phosphorylation and degradation of the inhibitory subunit α of NF-κB (I-κBα), TGase2 induces its polymerization. This polymerization results in dissociation of NF-κB and its translocation to the nucleus, where it is capable of up-regulating a host of inflammatory genes, including inducible nitric-oxide synthase and tumor necrosis factor α (TNF-α). Indeed, TGase inhibitors prevent depletion of monomeric I-κBα in the cytosol of cells overexpressing TGase 2. In an LPS-induced rat brain injury model, TGase inhibitors significantly reduced TNF-α synthesis. The findings are consistent with a model in which LPS-induced NF-κB activation is the result of phosphorylation of I-κBα by I-κB kinase as well as I-κBα polymerization by TGase 2. Safe and stable TGase2 inhibitors may be effective agents in diseases associated with inflammation.


The FASEB Journal | 2007

A pivotal role of matrix metalloproteinase-3 activity in dopaminergic neuronal degeneration via microglial activation

Yoon-Seong Kim; Dong Hee Choi; Michelle L. Block; Stefan Lorenzl; Lichuan Yang; Youn Jung Kim; Shuei Sugama; Byung Pil Cho; Onyou Hwang; Susan E. Browne; Soo Yul Kim; Jau-Shyong Hong; M. Flint Beal; Tong H. Joh

Recent studies have demonstrated that activated microglia play an important role in dopamine (DA) neuronal degeneration in Parkinson disease (PD) by generating NADPH‐oxidase (NADPHO)‐derived superoxide. However, the molecular mechanisms that underlie microglial activation in DA cell death are still disputed. We report here that matrix metalloproteinase‐3 (MMP‐3) was newly induced and activated in stressed DA cells, and the active form of MMP‐3 (actMMP‐3) was released into the medium. The released actMMP‐3, as well as catalytically active recombinant MMP‐3 (cMMP‐3) led to microglial activation and superoxide generation in microglia and enhanced DA cell death. cMMP‐3 caused DA cell death in mesencephalic neuron‐glia mixed culture of wild‐type (WT) mice, but this was attenuated in the culture of NADPHO subunit null mice (gp91phox‐/‐), suggesting that NADPHO mediated the cMMP‐3‐induced microglial production of superoxide and DA cell death. Furthermore, in the N‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP)‐injected animal model of PD, nigrostriatal DA neuronal degeneration, microglial activation, and superoxide generation were largely attenuated in MMP‐3‐/‐mice. These results indicate that actMMP‐3 released from stressed DA neurons is responsible for microglial activation and generation of NADPHO‐derived superoxide and eventually enhances nigrostriatal DA neuronal degeneration. Our results could lead to a novel therapeutic approach to PD. Kim, Y. S., Choi, D. H., Block, M. L., Lorenzl, S., Yang, L., Kim, Y. J., Sugama, S., Cho, B. P., Ywang, O., Browne, S. E., Kim, S. Y., Hong, J.‐S., Beal, M. F., Jon, T. H. A pivotal role of matrix metalloproteinase‐3 activity in dopaminergic neuronal degeneration via microglial activation. FASEB J. 21, 179–187 (2007)


Journal of Biological Chemistry | 2004

Transglutaminase 2 induces NF-κB activation via a novel pathway in BV-2 microglia

Jong-Min Lee; Yoon-Seong Kim; Dong-Hee Choi; Moon Suk Bang; Tay R. Han; Tong H. Joe; Soo-Youl Kim

Transglutaminase 2 (TGase 2) expression is increased in inflammatory diseases. We demonstrated previously that inhibitors of TGase 2 reduce nitric oxide (NO) generation in a lipopolysaccharide (LPS)-treated microglial cell line. However, the precise mechanism by which TGase 2 promotes inflammation remains unclear. We found that TGase 2 activates the transcriptional activator nuclear factor (NF)-κB and thereby enhances LPS-induced expression of inducible nitric-oxide synthase. TGase 2 activates NF-κB via a novel pathway. Rather than stimulating phosphorylation and degradation of the inhibitory subunit α of NF-κB (I-κBα), TGase2 induces its polymerization. This polymerization results in dissociation of NF-κB and its translocation to the nucleus, where it is capable of up-regulating a host of inflammatory genes, including inducible nitric-oxide synthase and tumor necrosis factor α (TNF-α). Indeed, TGase inhibitors prevent depletion of monomeric I-κBα in the cytosol of cells overexpressing TGase 2. In an LPS-induced rat brain injury model, TGase inhibitors significantly reduced TNF-α synthesis. The findings are consistent with a model in which LPS-induced NF-κB activation is the result of phosphorylation of I-κBα by I-κB kinase as well as I-κBα polymerization by TGase 2. Safe and stable TGase2 inhibitors may be effective agents in diseases associated with inflammation.


Journal of Neurochemistry | 2008

A novel intracellular role of matrix metalloproteinase-3 during apoptosis of dopaminergic cells

Dong Hee Choi; Eun-Mee Kim; Hyo Jin Son; Tong H. Joh; Yoon-Seong Kim; Dong-Hou Kim; M. Flint Beal; Onyou Hwang

We have previously demonstrated that the active form of matrix metalloproteinase‐3 (actMMP‐3) is released from dopamine(DA)rgic neurons undergoing apoptosis. Herein, whether actMMP‐3 might be generated intracellularly, and if so, whether it is involved in apoptosis of DArgic neurons itself was investigated in primary cultured DArgic neurons of wild‐type, MMP‐3 knockout animals, and CATH.a cells. During apoptosis, gene expression of MMP‐3 is induced, specifically among the various classes of MMPs, generating the proform (55 kDa) which is subsequently cleaved to the catalytically active actMMP‐3 (48 kDa) involving a serine protease. Intracellular actMMP‐3 activity is directly linked to apoptotic signaling in DArgic cells: (i) Pharmacologic inhibition of enzymatic activity, repression of gene expression by siRNA, and gene deficiency all lead to protection; (ii) pharmacologic inhibition causes attenuation of DNA fragmentation and caspase 3 activation, the indices of apoptosis; and (iii) inhibition of the pro‐apoptotic enzyme c‐Jun N‐terminal protein kinase leads to repression of MMP‐3 induction. Under the cell stress condition, MMP‐3 is released as actMMP‐3 rather than the proform (proMMP‐3), and catalytically active MMP‐3 added to the medium does not cause cell death. Thus, actMMP‐3 seems to have a novel intracellular role in apoptotic DArgic cells and this finding provides an insight into the pathogenesis of Parkinson’s disease.


Neuropharmacology | 2011

Fluoxetine prevents MPTP-induced loss of dopaminergic neurons by inhibiting microglial activation

Young Cheul Chung; Sang Ryong Kim; Ju-Young Park; Eun Sook Chung; Keun W. Park; So Y. Won; Eugene Bok; Minyoung Jin; Eun S. Park; Sung-Hwa Yoon; Hyuk Wan Ko; Yoon-Seong Kim; Byung Kwan Jin

Parkinsons disease (PD) is characterized by degeneration of nigrostriatal dopaminergic (DA) neurons. Mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) exhibit microglial activation-induced oxidative stress and inflammation, and nigrostriatal DA neuronal damage, and thus serve as an experimental model of PD. Here, we report that fluoxetine, one of the most commonly prescribed antidepressants, prevents MPTP-induced degeneration of nigrostriatal DA neurons and increases striatal dopamine levels with the partial motor recovery. This was accompanied by inhibiting transient expression of proinflammatory cytokines and inducible nitric oxide synthase; and attenuating microglial NADPH oxidase activation, reactive oxygen species/reactive nitrogen species production, and consequent oxidative damage. Interestingly, fluoxetine was found to protect DA neuronal damage from 1-methyl-4-phenyl-pyridinium (MPP(+)) neurotoxicity in co-cultures of mesencephalic neurons and microglia but not in neuron-enriched mesencephalic cultures devoid of microglia. The present in vivo and in vitro findings show that fluoxetine may possess anti-inflammatory properties and inhibit glial activation-mediated oxidative stress. Therefore, we carefully propose that neuroprotection of fluoxetine might be associated with its anti-inflammatory properties and could be employed as novel therapeutic agents for PD and other disorders associated with neuroinflammation and microglia-derived oxidative damage.


Antioxidants & Redox Signaling | 2012

NADPH Oxidase 1-Mediated Oxidative Stress Leads to Dopamine Neuron Death in Parkinson's Disease

Dong-Hee Choi; Ana Clara Cristóvão; Subhrangshu Guhathakurta; Jongmin Lee; Tong H. Joh; M. Flint Beal; Yoon-Seong Kim

AIM Oxidative stress has long been considered as a major contributing factor in the pathogenesis of Parkinsons disease. However, molecular sources for reactive oxygen species in Parkinsons disease have not been clearly elucidated. Herein, we sought to investigate whether a superoxide-producing NADPH oxidases (NOXs) are implicated in oxidative stress-mediated dopaminergic neuronal degeneration. RESULTS Expression of various Nox isoforms and cytoplasmic components were investigated in N27, rat dopaminergic cells. While most of Nox isoforms were constitutively expressed, Nox1 expression was significantly increased after treatment with 6-hydroxydopamine. Rac1, a key regulator in the Nox1 system, was also activated. Striatal injection of 6-hydroxydopamine increased Nox1 expression in dopaminergic neurons in the rat substantia nigra. Interestingly, it was localized into the nucleus, and immunostaining for DNA oxidative stress marker, 8-oxo-dG, was increased. Nox1 expression was also found in the nucleus of dopaminergic neurons in the substantia nigra of Parkinsons disease patients. Adeno-associated virus-mediated Nox1 knockdown or Rac1 inhibition reduced 6-hydroxydopamine-induced oxidative DNA damage and dopaminergic neuronal degeneration significantly. INNOVATION Nox1/Rac1 could serve as a potential therapeutic target for Parkinsons disease. CONCLUSION We provide evidence that dopaminergic neurons are equipped with the Nox1/Rac1 superoxide-generating system. Stress-induced Nox1/Rac1 activation causes oxidative DNA damage and neurodegeneration. Reduced dopaminergic neuronal death achieved by targeting Nox1/Rac1, emphasizes the impact of oxidative stress caused by this system on the pathogenesis and therapy in Parkinsons disease.


Experimental and Molecular Medicine | 2007

Celastrol inhibits production of nitric oxide and proinflammatory cytokines through MAPK signal transduction and NF-κB in LPS-stimulated BV-2 microglial cells

Hyo Won Jung; Yoo Sun Chung; Yoon-Seong Kim; Yong-Ki Park

Excessive production of nitric oxide (NO) and proinflammatory cytokines from activated microglia play an important role in human neurodegenerative disorders. Here, we investigated whether celastrol, which has been used as a potent anti-inflammatory and anti-oxidative agent in Chinese medicine, attenuates excessive production of NO and proinflammatory cytokines such as TNF-α and IL-1β in LPS-stimulated BV-2 cells, a mouse microglial cell line. We report here that the LPS-elicited excessive production of NO, TNF-α, and IL-1β in BV-2 cells was largely inhibited in the presence of celastrol, and the attenuation of inducible iNOS and these cytokines resulted from the reduced expression of mRNAs of iNOS and these cytokines, respectively. The molecular mechanisms that underlie celastrol-mediated attenuation were the inhibition of LPS-induced phosphorylation of MAPK/ERK1/2 and the DNA binding activity of NF-κB in BV-2 cells. The results indicate that celastrol effectively attenuated NO and proinflammatory cytokine production via the inhibition of ERK1/2 phosphorylation and NF-κB activation in LPS-activated microglia. Thus, celastrol may be an effective therapeutic candidate for use in the treatment of neurodegenerative human brain disorders.

Collaboration


Dive into the Yoon-Seong Kim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Goun Je

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce T. Volpe

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Sambuddha Basu

University of Central Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge