Network


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

Hotspot


Dive into the research topics where Won-Ha Lee is active.

Publication


Featured researches published by Won-Ha Lee.


Immunological Reviews | 2006

CD4+ T-cell memory: generation and multi-faceted roles for CD4+ T cells in protective immunity to influenza

Susan L. Swain; Javed N. Agrewala; D. Brown; Dawn M. Jelley-Gibbs; Susanne Golech; Gail E. Huston; Stephen C. Jones; Cris Kamperschroer; Won-Ha Lee; K. Kai McKinstry; Eulogia Román; Tara M. Strutt; Nan Ping Weng

Summary:  We have outlined the carefully orchestrated process of CD4+ T‐cell differentiation from naïve to effector and from effector to memory cells with a focus on how these processes can be studied in vivo in responses to pathogen infection. We emphasize that the regulatory factors that determine the quality and quantity of the effector and memory cells generated include (i) the antigen dose during the initial T‐cell interaction with antigen‐presenting cells; (ii) the dose and duration of repeated interactions; and (iii) the milieu of inflammatory and growth cytokines that responding CD4+ T cells encounter. We suggest that heterogeneity in these regulatory factors leads to the generation of a spectrum of effectors with different functional attributes. Furthermore, we suggest that it is the presence of effectors at different stages along a pathway of progressive linear differentiation that leads to a related spectrum of memory cells. Our studies particularly highlight the multifaceted roles of CD4+ effector and memory T cells in protective responses to influenza infection and support the concept that efficient priming of CD4+ T cells that react to shared influenza proteins could contribute greatly to vaccine strategies for influenza.


The Journal of Neuroscience | 2009

Lipocalin-2 is an autocrine mediator of reactive astrocytosis.

Shinrye Lee; Jae-Yong Park; Won-Ha Lee; Ho Kim; Hae Chul Park; Kiyoshi Mori; Kyoungho Suk

Astrocytes, the most abundant glial cell type in the brain, provide metabolic and trophic support to neurons and modulate synaptic activity. In response to a brain injury, astrocytes proliferate and become hypertrophic with an increased expression of intermediate filament proteins. This process is collectively referred to as reactive astrocytosis. Lipocalin 2 (lcn2) is a member of the lipocalin family that binds to small hydrophobic molecules. We propose that lcn2 is an autocrine mediator of reactive astrocytosis based on the multiple roles of lcn2 in the regulation of cell death, morphology, and migration of astrocytes. lcn2 expression and secretion increased after inflammatory stimulation in cultured astrocytes. Forced expression of lcn2 or treatment with LCN2 protein increased the sensitivity of astrocytes to cytotoxic stimuli. Iron and BIM (Bcl-2-interacting mediator of cell death) proteins were involved in the cytotoxic sensitization process. LCN2 protein induced upregulation of glial fibrillary acidic protein (GFAP), cell migration, and morphological changes similar to characteristic phenotypic changes termed reactive astrocytosis. The lcn2-induced phenotypic changes of astrocytes occurred through a Rho–ROCK (Rho kinase)–GFAP pathway, which was positively regulated by nitric oxide and cGMP. In zebrafishes, forced expression of rat lcn2 gene increased the number and thickness of cellular processes in GFAP-expressing radial glia cells, suggesting that lcn2 expression in glia cells plays an important role in vivo. Our results suggest that lcn2 acts in an autocrine manner to induce cell death sensitization and morphological changes in astrocytes under inflammatory conditions and that these phenotypic changes may be the basis of reactive astrocytosis in vivo.


Journal of Immunology | 2005

TLR4, but Not TLR2, Signals Autoregulatory Apoptosis of Cultured Microglia: A Critical Role of IFN-β as a Decision Maker

Dae Young Jung; Heasuk Lee; Bo-Young Jung; Jiyeon Ock; Myung-Shik Lee; Won-Ha Lee; Kyoungho Suk

TLRs mediate diverse signaling after recognition of evolutionary conserved pathogen-associated molecular patterns such as LPS and lipopeptides. Both TLR2 and TLR4 are known to trigger a protective immune response as well as cellular apoptosis. In this study, we present evidence that TLR4, but not TLR2, mediates an autoregulatory apoptosis of activated microglia. Brain microglia underwent apoptosis upon stimulation with TLR4 ligand (LPS), but not TLR2 ligands (Pam3Cys-Ser-Lys4, peptidoglycan, and lipoteichoic acid). Based on studies using TLR2-deficient or TLR4 mutant mice and TLR dominant-negative mutants, we also demonstrated that TLR4, but not TLR2, is necessary for microglial apoptosis. The critical difference between TLR2 and TLR4 signalings in microglia was IFN regulatory factor-3 (IRF-3) activation, followed by IFN-β expression: while TLR4 agonist induced the activation of IRF-3/IFN-β pathway, TLR2 did not. Nevertheless, both TLR2 and TLR4 agonists strongly induced NF-κB activation and NO production in microglia. Neutralizing Ab against IFN-β attenuated TLR4-mediated microglial apoptosis. IFN-β alone, however, did not induce a significant cell death. Meanwhile, TLR2 activation induced microglial apoptosis with help of IFN-β, indicating that IFN-β production following IRF-3 activation determines the apoptogenic action of TLR signaling. TLR4-mediated microglial apoptosis was mediated by MyD88 and Toll/IL-1R domain-containing adaptor-inducing IFN-β, and was associated with caspase-11 and -3 activation rather than Fas-associated death domain protein/caspase-8 pathway. Taken together, TLR4 appears to signal a microglial apoptosis via autocrine/paracrine IFN-β production, which may act as an apoptotic sensitizer.


Neuropharmacology | 2008

Inhibition of glial inflammatory activation and neurotoxicity by tricyclic antidepressants

Jaegyu Hwang; Long Tai Zheng; Jiyeon Ock; Maan-Gee Lee; Sang-Hyun Kim; Ho-Won Lee; Won-Ha Lee; Hae Chul Park; Kyoungho Suk

Glial activation and neuroinflammatory processes play an important role in the pathogenesis of neurodegenerative diseases such as Alzheimers disease, Parkinsons disease, and HIV dementia. Activated glial cells can secrete various proinflammatory cytokines and neurotoxic mediators, which may contribute to neuronal cell death. Inhibition of glial activation may alleviate neurodegeneration under these conditions. In the present study, the antiinflammatory and neuroprotective effects of tricyclic antidepressants were investigated using cultured brain cells as a model. The results showed that clomipramine and imipramine significantly decreased the production of nitric oxide or tumor necrosis factor-alpha (TNF-alpha) in microglia and astrocyte cultures. Clomipramine and imipramine also attenuated the expression of inducible nitric oxide synthase and proinflammatory cytokines such as interleukin-1beta and TNF-alpha at mRNA levels. In addition, clomipramine and imipramine inhibited IkappaB degradation, nuclear translocation of the p65 subunit of NF-kappaB, and phosphorylation of p38 mitogen-activated protein kinase in the lipopolysaccharide-stimulated microglia cells. Moreover, clomipramine and imipramine were neuroprotective as the drugs reduced microglia-mediated neuroblastoma cell death in a microglia/neuron co-culture. Therefore, these results imply that clomipramine and imipramine have antiinflammatory and neuroprotective effects in the central nervous system by modulating glial activation.


Journal of Immunology | 2007

A Dual Role of Lipocalin 2 in the Apoptosis and Deramification of Activated Microglia

Shinrye Lee; Jayoung Lee; Sangseop Kim; Jae-Yong Park; Won-Ha Lee; Kiyoshi Mori; Sang-Hyun Kim; Kyoungho Suk

Activated microglia are thought to undergo apoptosis as a self-regulatory mechanism. To better understand molecular mechanisms of the microglial apoptosis, apoptosis-resistant variants of microglial cells were selected and characterized. The expression of lipocalin 2 (lcn2) was significantly down-regulated in the microglial cells that were resistant to NO-induced apoptosis. lcn2 expression was increased by inflammatory stimuli in microglia. The stable expression of lcn2 as well as the addition of rLCN2 protein augmented the sensitivity of microglia to the NO-induced apoptosis, while knockdown of lcn2 expression using short hairpin RNA attenuated the cell death. Microglial cells with increased lcn2 expression were more sensitive to other cytotoxic agents as well. Thus, inflammatory activation of microglia may lead to up-regulation of lcn2 expression, which sensitizes microglia to the self-regulatory apoptosis. Additionally, the stable expression of lcn2 in BV-2 microglia cells induced a morphological change of the cells into the round shape with a loss of processes. Treatment of primary microglia cultures with the rLCN2 protein also induced the deramification of microglia. The deramification of microglia was closely related with the apoptosis-prone phenotype, because other deramification-inducing agents such as cAMP-elevating agent forskolin, ATP, and calcium ionophore also rendered microglia more sensitive to cell death. Taken together, our results suggest that activated microglia may secrete LCN2 protein, which act in an autocrine manner to sensitize microglia to the self-regulatory apoptosis and to endow microglia with an amoeboid form, a canonical morphology of activated microglia in vivo.


Molecular Pharmacology | 2006

Decursin Inhibits Induction of Inflammatory Mediators by Blocking Nuclear Factor-κB Activation in Macrophages

Jung-Hee Kim; Ji-Hye Jeong; Sung-Tak Jeon; Ho Kim; Jiyeon Ock; Kyoungho Suk; Sang-In Kim; Kyung-Sik Song; Won-Ha Lee

In the course of screening inhibitors of matrix metalloproteinase (MMP)-9 induction in macrophages, we isolated decursin, a coumarin compound, from the roots of Angelicae gigas. As a marker for the screening and isolation, we tested expression of MMP-9 in RAW264.7 cells and THP-1 cells after treatment with bacterial lipopolysaccharide (LPS), the TLR-4 ligand. Decursin suppressed MMP-9 expression in cells stimulated by LPS in a dose-dependent manner at concentrations below 60 μM with no sign of cytotoxicity. The suppressive effect of decursin was observed not only in cells stimulated with ligands for TLR4, TLR2, TLR3, and TLR9 but also in cells stimulated with interleukin (IL)-1β, and tumor necrosis factor (TNF)-α, indicating that the molecular target of decursin is common signaling molecules induced by these stimulants. In addition to the suppression of MMP-9 expression, decursin blocked nitric oxide production and cytokine (IL-8, MCP-1, IL-1β, and TNF-α) secretion induced by LPS. To find out the molecular mechanism responsible for the suppressive effect of decursin, we analyzed signaling molecules involved in the TLR-mediated activation of MMP-9 and cytokines. Decursin blocked phosphorylation of IκB and nuclear translocation of NF-κB in THP-1 cells activated with LPS. Furthermore, expression of a luciferase reporter gene under the promoter containing NF-κB binding sites was blocked by decursin. These data indicate that decursin is a novel inhibitor of NF-κB activation in signaling induced by TLR ligands and cytokines.


The FASEB Journal | 2013

Secreted protein lipocalin-2 promotes microglial M1 polarization

Eunha Jang; Shinrye Lee; Jong-Heon Kim; Jae-Hong Kim; Jung-Wan Seo; Won-Ha Lee; Kiyoshi Mori; Kazuwa Nakao; Kyoungho Suk

Activated macrophages are classified into two different forms: classically activated (M1) or alternatively activated (M2) macrophages. The presence of M1/M2 phenotypic polarization has also been suggested for microglia. Here, we report that the secreted protein lipocalin 2 (LCN2) amplifies M1 polarization of activated microglia. LCN2 protein (EC50 1 μg/ml), but not glutathione S‐transferase used as a control, increased the M1‐related gene expression in cultured mouse microglial cells after 8–24 h. LCN2 was secreted from M1‐polarized, but not M2‐polarized, microglia. LCN2 inhibited phosphorylation of STAT6 in IL‐4‐stimulated microglia, suggesting LCN2 suppression of the canonical M2 signaling. In the lipopolysaccharide (LPS)‐induced mouse neuroinflammation model, the expression of LCN2 was notably increased in microglia. Primary microglial cultures derived from LCN2‐deficient mice showed a suppressed M1 response and enhanced M2 response. Mice lacking LCN2 showed a markedly reduced M1‐related gene expression in microglia after LPS injection, which was consistent with the results of histological analysis. Neuroinflammation‐associated impairment in motor behavior and cognitive function was also attenuated in the LCN2‐deficient mice, as determined by the rotarod performance test, fatigue test, open‐field test, and object recognition task. These findings suggest that LCN2 is an M1‐amplifier in brain microglial cells.—Jang, E., Lee, S., Kim, J.‐H., Kim, J.‐H., Seo, J.‐W., Lee, W.‐H., Mori, K., Nakao, K., Suk, K. Secreted protein lipocalin‐2 promotes microglial M1 polarization. FASEB J. 27, 1176–1190 (2013). www.fasebj.org


Journal of Experimental Medicine | 2007

Rapid default transition of CD4 T cell effectors to functional memory cells

K. Kai McKinstry; Susanne Golech; Won-Ha Lee; Gail E. Huston; Nan-ping Weng; Susan L. Swain

The majority of highly activated CD4 T cell effectors die after antigen clearance, but a small number revert to a resting state, becoming memory cells with unique functional attributes. It is currently unclear when after antigen clearance effectors return to rest and acquire important memory properties. We follow well-defined cohorts of CD4 T cells through the effector-to-memory transition by analyzing phenotype, important functional properties, and gene expression profiles. We find that the transition from effector to memory is rapid in that effectors rested for only 3 d closely resemble canonical memory cells rested for 60 d or longer in the absence of antigen. This is true for both Th1 and Th2 lineages, and occurs whether CD4 T cell effectors rest in vivo or in vitro, suggesting a default pathway. We find that the effector–memory transition at the level of gene expression occurs in two stages: a rapid loss of expression of a myriad of effector-associated genes, and a more gradual gain of expression of a cohort of genes uniquely associated with memory cells rested for extended periods.


Biochemical Pharmacology | 2009

Suppression of the TRIF-dependent signaling pathway of Toll-like receptors by luteolin.

Jun Kyung Lee; So Young Kim; Yoon Sun Kim; Won-Ha Lee; Daniel H. Hwang; Joo Young Lee

Toll-like receptors (TLRs) play important roles in induction of innate immune responses for both host defense against invading pathogens and wound healing after tissue injury. Since dysregulation of TLR-mediated immune responses is closely linked to many chronic diseases, modulation of TLR activation by small molecules may have therapeutic potential against such diseases. Expression of the majority of lipopolysaccharide-induced TLR4 target genes is mediated through a MyD88-independent (TRIF-dependent) signaling pathway. In order to evaluate the therapeutic potential of the flavonoid luteolin we examined its effect on TLR-stimulated signal transduction via the TRIF-dependent pathway. Luteolin suppressed activation of Interferon regulatory factor 3 (IRF3) and NFkappaB induced by TLR3 and TLR4 agonists resulting in the decreased expression of target genes such as TNF-alpha, IL-6, IL-12, IP-10, IFNbeta, CXCL9, and IL-27 in macrophages. Luteolin attenuated ligand-independent activation of IRF3 or NFkappaB induced by TLR4, TRIF, or TBK1, while it did not inhibit TLR oligomerization. Luteolin inhibited TBK1-kinase activity and IRF3 dimerization and phosphorylation, leading to the reduction of TBK1-dependent gene expression. Structural analogs of luteolin such as quercetin, chrysin, and eriodictyol also inhibited TBK1-kinase activity and TBK1-target gene expression. These results demonstrate that TBK1 is a novel target of anti-inflammatory flavonoids resulting in the down-regulation of the TRIF-dependent signaling pathway. These results suggest that the beneficial activities of these flavonoids against inflammatory diseases may be attributed to the modulation of TLR-mediated inflammatory responses.


Journal of Biological Chemistry | 2011

Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration.

Shinrye Lee; Jong-Heon Kim; Jae-Hong Kim; Jung-Wan Seo; Hyung-Soo Han; Won-Ha Lee; Kiyoshi Mori; Kazuwa Nakao; Jonathan Barasch; Kyoungho Suk

Background: LCN2 has been implicated in cell morphology and migration. Results: LCN2 promotes cell migration through up-regulation of chemokines (CXCL10) in brain both in vitro and in vivo. Conclusion: LCN2 is a chemokine inducer in the CNS and may accelerate cell migration under inflammatory conditions in an autocrine or paracrine manner. Significance: LCN2 could be targeted to therapeutically modulate glial responses in various neuroinflammatory disease conditions. The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.

Collaboration


Dive into the Won-Ha Lee's collaboration.

Top Co-Authors

Avatar

Kyoungho Suk

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Won-Jung Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Min Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Jong-Heon Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Shinrye Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eun-Ju Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jae-Hong Kim

Kyungpook National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge