Network


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

Hotspot


Dive into the research topics where Young Woo Han is active.

Publication


Featured researches published by Young Woo Han.


Journal of Immunology | 2009

Functional Modulation of Dendritic Cells and Macrophages by Japanese Encephalitis Virus through MyD88 Adaptor Molecule-Dependent and -Independent Pathways

Abi G. Aleyas; Junu A. George; Young Woo Han; Masudur Rahman; Seon Ju Kim; Sang-Bae Han; Byung Sam Kim; Koanhoi Kim; Seong Kug Eo

Dendritic cells (DCs) are potent initiators of T cell-mediated immunity that undergo maturation during viral infections. However, few reports describing the interactions of DCs with Japanese encephalitis virus (JEV), which remains the most frequent cause of acute and epidemic viral encephalitis, are available. In this study, we investigated the interaction of JEV with DCs and macrophages. JEV replicated its viral RNA in both cells with different efficiency, and JEV infection of macrophages followed the classical activation pathway of up-regulation of tested costimulatory molecules and proinflammatory cytokine production (IL-6, TNF-α, and IL-12). On the contrary, JEV-infected DCs failed to up-regulate costimulatory molecules such as CD40 and MHC class II. Of more interest, along with production of proinflammatory cytokines, DCs infected by JEV released antiinflammatory cytokine IL-10, which was not detected in macrophages. Moreover, signaling through MyD88 molecule, a pan-adaptor molecule of TLRs, and p38 MAPK in JEV-infected DCs was found to play a role in the production of cytokines and subversion of primary CD4+ and CD8+ T cell responses. We also found that IL-10 released from JEV-infected DCs led to a reduction in the priming of CD8+ T cells, but not CD4+ T cells. Taken together, our data suggest that JEV induces functional impairment of DCs through MyD88-dependent and -independent pathways, which subsequently leads to poor CD4+ and CD8+ T cell responses, resulting in boosting viral survival and dissemination in the body.


PLOS Pathogens | 2014

Distinct dictation of Japanese encephalitis virus-induced neuroinflammation and lethality via triggering TLR3 and TLR4 signal pathways.

Young Woo Han; Jin Young Choi; Erdenebelig Uyangaa; Seong Bum Kim; Jin Hyoung Kim; Bum Seok Kim; Koanhoi Kim; Seong Kug Eo

Japanese encephalitis (JE) is major emerging neurologic disease caused by JE virus. To date, the impact of TLR molecules on JE progression has not been addressed. Here, we determined whether each TLR modulates JE, using several TLR-deficient mouse strains (TLR2, TLR3, TLR4, TLR7, TLR9). Surprisingly, among the tested TLR-deficient mice there were contrasting results in TLR3−/− and TLR4−/− mice, i.e. TLR3−/− mice were highly susceptible to JE, whereas TLR4−/− mice showed enhanced resistance to JE. TLR3 ablation induced severe CNS inflammation characterized by early infiltration of inflammatory CD11b+Ly-6Chigh monocytes along with profoundly increased viral burden, proinflammatory cytokine/chemokine expression as well as BBB permeability. In contrast, TLR4−/− mice showed mild CNS inflammation manifested by reduced viral burden, leukocyte infiltration and proinflammatory cytokine expression. Interestingly, TLR4 ablation provided potent in vivo systemic type I IFN innate response, as well as ex vivo type I IFN production associated with strong induction of antiviral PRRs (RIG-I, MDA5), transcription factors (IRF-3, IRF-7), and IFN-dependent (PKR, Oas1, Mx) and independent ISGs (ISG49, ISG54, ISG56) by alternative activation of IRF3 and NF-κB in myeloid-derived DCs and macrophages, as compared to TLR3−/− myeloid-derived cells which were more permissive to viral replication through impaired type I IFN innate response. TLR4 ablation also appeared to mount an enhanced type I IFN innate and humoral, CD4+ and CD8+ T cell responses, which were mediated by altered immune cell populations (increased number of plasmacytoid DCs and NK cells, reduced CD11b+Ly-6Chigh monocytes) and CD4+Foxp3+ Treg number in lymphoid tissue. Thus, potent type I IFN innate and adaptive immune responses in the absence of TLR4 were closely coupled with reduced JE lethality. Collectively, these results suggest that a balanced triggering of TLR signal array by viral components during JE progression could be responsible for determining disease outcome through regulating negative and positive factors.


Microbiology and Immunology | 2006

Cytokine GM-CSF Genetic Adjuvant Facilitates Prophylactic DNA Vaccine against Pseudorabies Virus through Enhanced Immune Responses

Hyun A Yoon; Abi G. Aleyas; Junu A. George; Seong Ok Park; Young Woo Han; John Hwa Lee; Jeong-Gon Cho; Seong Kug Eo

Granulocyte/macrophage colony‐stimulatory factor (GM‐CSF) is an attractive adjuvant for a DNA vaccine on account of its ability to recruit antigen‐presenting cells (APCs) to the site of antigen synthesis as well as its ability to stimulate the maturation of dendritic cells (DCs). This study evaluated the utility of GM‐CSF cDNA as a DNA vaccine adjuvant for glycoprotein B (gB) of pseudorabies virus (PrV) in a murine model. The co‐injection of GM‐CSF DNA enhanced the levels of serum PrV‐specific IgG with a 1.5‐ to 2‐fold increase. Moreover, GM‐CSF co‐injection inhibited the production of IgG2a isotype. However, it enhanced production of an IgG1 isotype resulting in humoral responses biased to the Th2‐type against PrV antigen. In contrast, the co‐administration of GM‐CSF DNA enhanced the T cell‐mediated immunity biased to the Th1‐type, as judged by the significantly higher level of cytokine IL‐2 and IFN‐γ production but not IL‐4. When challenged with a lethal dose of PrV, the GM‐CSF co‐injection enhanced the resistance against a PrV infection. This suggests that co‐inoculation with a vector expressing GM‐CSF enhanced the protective immunity against a PrV infection. This immunity was caused by the induction of increased humoral and cellular immunity in response to PrV antigen.


Journal of Immunology | 2010

Multifront assault on antigen presentation by Japanese encephalitis virus subverts CD8+ T cell responses.

Abi G. Aleyas; Young Woo Han; Junu A. George; Bumseok Kim; Koanhoi Kim; Chong-Kil Lee; Seong Kug Eo

Japanese encephalitis virus (JEV) is a frequent cause of acute and epidemic viral encephalitis. However, there is little information describing the mechanisms by which JEV subverts immune responses that may predispose the host to secondary infections. In this study, we found that JEV induced the subversion of CD8+ T cell responses in a transient manner that was closely correlated with viral multiplication. Subsequently, analysis using a TCR-transgenic system revealed that CD8+ T cells purified from JEV-infected mice showed impaired responses, and that naive CD8+ T cells adoptively transferred into JEV-infected recipients showed less expanded responses. Furthermore, JEV altered the splenic dendritic cell (DC) subpopulation via preferential depletion of CD8α+CD11c+ DCs without changing the plasmacytoid DCs and induced a significant reduction in the surface expression of MHC class II and CD40, but not MHC class I, CD80, and CD86 molecules. Finally, JEV was shown to inhibit the presentation of MHC class I-restricted Ag in DCs, and this immune suppression was ameliorated via the activation of DCs by TLR agonists. Collectively, our data indicate that JEV precludes the functions of Ag-presenting machinery, such as depletion of CD8α+CD11c+ DCs and downregulation of MHC class I-restricted Ag presentation, thereby leading to immune subversion of CD8+ T cells.


European Journal of Immunology | 2012

Impaired cross-presentation of CD8α+ CD11c+ dendritic cells by Japanese encephalitis virus in a TLR2/MyD88 signal pathway-dependent manner.

Abi G. Aleyas; Young Woo Han; Ajit Mahadev Patil; Seong Bum Kim; Koanhoi Kim; Seong Kug Eo

Cross‐presentation is the pathway by which exogenous antigens are routed for presentation by MHC class I molecules leading to activation of antiviral CD8+ T‐cell responses. However, there is little information describing the modulation of cross‐presentation and the impact of pathogen‐derived signals associated with Japanese encephalitis virus (JEV), which is one of the most common causes of encephalitis in humans. In this study, we demonstrate that JEV infection could suppress in vivo cross‐presentation of soluble and cell‐associated antigens, thereby generating weak CD8+ T‐cell responses to exogenous antigens, as evaluated by CFSE dilution of adoptively transferred CD8+ T cells and in vivo CTL killing activity. Furthermore, CD8α+CD11c+ dendritic cells (DCs), which are known to be far more efficient at cross‐presenting soluble antigens, played a specific role in contributing to JEV‐mediated inhibition of the cross‐presentation of exogenous antigens through interference with effective antigen uptake. Finally, this study provides evidence that TLR2‐MyD88 and p38 MAPK signal pathway might be involved in JEV‐mediated inhibition of cross‐presentation of soluble and cell‐associated antigens. These observations suggest that the modulation of cross‐presentation of exogenous antigens through TLR signaling has important implications for antiviral immune responses against JEV infection and the development of effective vaccination strategies.


Veterinary Microbiology | 2012

Oral co-administration of live attenuated Salmonella enterica serovar Typhimurium expressing chicken interferon-α and interleukin-18 enhances the alleviation of clinical signs caused by respiratory infection with avian influenza virus H9N2.

Masudur Rahman; Erdenebileg Uyangaa; Young Woo Han; Seong Bum Kim; Jin Hyoung Kim; Jin Young Choi; Seong Kug Eo

The combined use of cytokines has shown synergistic and/or additive effects in controlling several viral infections of livestock animals. However, little is known concerning the practical use of chicken cytokine combinations to control avian diseases. Here, we investigated the antiviral efficacy of oral co-administration of chicken interferon-α (chIFN-α) and chicken interleukin-18 (chIL-18) using attenuated Salmonella enterica serovar Typhimurium in chickens infected with avian influenza virus (AIV) H9N2. Our results demonstrate that oral co-administration of S. enterica serovar Typhimurium expressing chIFN-α and chIL-18 produced a greater alleviation of clinical signs caused by respiratory infection with AIV H9N2 in chickens, when compared to administration of S. enterica serovar Typhimurium expressing either chIFN-α or chIL-18 alone. Mortality, clinical symptom severity, and feed and water intake were used to access treatment effectiveness. This enhancement of antiviral immunity was further confirmed by evidence of reduced rectal shedding and decreased replication of AIV H9N2 in several different tissues of challenged chickens including trachea, lung, cecal tonsil, and brain. Furthermore, oral co-administration of chIFN-α and chIL-18 more efficiently modulated the immune responses of chickens against AIV H9N2 by enhancing both humoral and Th1-biased cell-mediated immunity, compared to single administration of either construct. Therefore, our results suggest that the combined administration of two chicken cytokines, chIFN-α and chIL-18, using attenuated S. enterica serovar Typhimurium as an oral carrier, provides an effective means for controlling respiratory disease caused by AIV H9N2 infection.


Veterinary Microbiology | 2012

Oral administration of Salmonella enterica serovar Typhimurium expressing swine interleukin-18 induces Th1-biased protective immunity against inactivated vaccine of pseudorabies virus.

Seong Bum Kim; Seon Ju Kim; Byung Min Lee; Young Woo Han; Masudur Rahman; Erdenebileg Uyangaa; Jin Hyoung Kim; Jin Young Choi; Dong Jin Yoo; Koanhoi Kim; Seong Kug Eo

Enhancing and/or modulating innate and adaptive immunity by cytokines appears to be greatly useful to provide effective protective immunity against infectious diseases. However, an effective delivery system for mass administration in livestock industry is needed because of limitations such as cost, labor, time, and protein stability. Here the immunomodulatory functions of swine interleukine-18 (swIL-18), known as IFN-γ-inducing factor (IGIF), were evaluated in a vaccination model of pseudorabies virus (PrV) using attenuated Salmonella enterica serovar Typhimurium as the oral delivery system. The oral administration of S. enterica serovar Typhimurium expressing swIL-18 prior to vaccination with inactivated PrV vaccine induced enhanced levels of serum PrV-specific IgG and its IgG2 isotype, compared to administration of S. enterica serovar Typhimurium harboring the empty vector. Furthermore, S. enterica serovar Typhimurium expressing swIL-18 mounted Th1-biased cellular immune responses against PrV antigen, as evaluated by the production of IFN-γ and IL-4 from peripheral blood mononuclear cells of piglets. Subsequently, Th1-biased immunity induced by S. enterica serovar Typhimurium expressing swIL-18 showed rapid response and rendered piglets displayed more alleviated clinical signs following the virulent PrV challenge. Also, this alleviation of clinical signs was further confirmed by the reduction of nasal excretion of PrV after challenge. The present study demonstrates the extended use of immunomodulatory functions of swIL-18 orally delivered by attenuated S. enterica serovar Typhimurium.


Immunology and Cell Biology | 2009

Genetic co-transfer of CCR7 ligands enhances immunity and prolongs survival against virulent challenge of pseudorabies virus

Young Woo Han; Abi G. Aleyas; Junu A. George; Seon Ju Kim; Hye Kyung Kim; Dong Jin Yoo; Seong Ho Kang; Seong Kug Eo

The CC chemokine receptor 7 (CCR7) and cognate CCR7 ligands, CCL19 and CCL21, help establish microenvironments in lymphoid tissue that can facilitate encounters between naive T cells and mature dendritic cells (DCs). This study was conducted to determine if CCR7 ligands can augment the immunogenicity of a DNA vaccine that expresses glycoprotein B (gB) of the pseudorabies virus (PrV). The genetic co‐transfer of CCR7 ligands along with a PrV DNA vaccine increased the levels of serum PrV‐specific immunoglobulin (Ig) G by 2‐ to 2.5‐fold. In addition, the level of PrV‐specific IgG2a isotype was significantly enhanced by co‐injection of CCR7 ligand DNA, which indicates that CCR7 ligand biases the humoral immunity toward the Th1‐type pattern. The co‐injection of CCR7 ligand DNA consistently enhanced the level of Th1‐type cytokines (IL‐2 and IFN‐γ) produced by stimulated immune cells when compared with a group that was vaccinated with the PrV DNA vaccine. Also, the genetic co‐transfer of CCR7 ligand DNAs with PrV DNA vaccine provided prolonged survival against a virulent challenge by PrV. Moreover, the co‐administration of CCR7 ligand DNA increased the number of mature DCs into the secondary lymphoid tissues, which appeared to enhance the proliferation of PrV‐immune CD4+ T cells. Taken together, these findings indicate that CCR7 ligands are an attractive adjuvant for a PrV DNA vaccine that can offer protective immunity against the PrV.


Immunology and Cell Biology | 2006

Differential segregation of protective immunity by encoded antigen in DNA vaccine against pseudorabies virus

Hyun A Yoon; Abi G. Aleyas; Junu A. George; Seong Ok Park; Young Woo Han; Seong Ho Kang; Jeong Gon Cho; Seong Kug Eo

A murine model immunized with plasmid DNA vaccine expressing three glycoproteins pCIgB, pCIgC and pCIgD were used to examine the relative potency of major glycoproteins as well as the contribution of immunological parameters in providing protective immunity against the pseudorabies virus (PrV). Among the three glycoprotein‐encoded plasmid DNA vaccines, pCIgB produced the strongest response of PrV‐specific IgG in the sera. pCIgB and pCIgD also induced a contrast pattern of immunity that was biased to the Th1 and Th2 types, respectively. pCIgC showed the potent inducer of CD8+ T‐cell‐mediated CTL activity against PrV. In addition, a cocktail vaccination of all three glycoprotein‐encoded plasmid DNA vaccines induced the production of both cytokine types, Th1 and Th2 with levels that were the same as that of each immunogen. With regard to protective efficacy, pCIgB induced the most effective protection against a virulent virus challenge and a cocktail vaccination appeared to offer complete protection against a 5 LD 50 challenge, but not a 10 LD 50 one. pCIgD induced protection that was same as pCIgB, but pCIgC offered no effective protection. These results show the relative potency of the three glycoprotein‐encoded PrV DNA vaccines in inducing protective immunity against PrV infection. The results in this study support previous results showing the importance of Th1‐type CD4+ T cells and their antibodies in conferring protection.


BMC Veterinary Research | 2012

Enhancement of Th1-biased protective immunity against avian influenza H9N2 virus via oral co-administration of attenuated Salmonella enterica serovar Typhimurium expressing chicken interferon-α and interleukin-18 along with an inactivated vaccine

Masudur Rahman; Erdenebileg Uyangaa; Young Woo Han; Seong Bum Kim; Jin Hyoung Kim; Jin Young Choi; Seong Kug Eo

BackgroundControl of currently circulating re-assorted low-pathogenicity avian influenza (LPAI) H9N2 is a major concern for both animal and human health. Thus, an improved LPAI H9N2 vaccination strategy is needed to induce complete immunity in chickens against LPAI H9N2 virus strains. Cytokines play a crucial role in mounting both the type and extent of an immune response generated following infection with a pathogen or after vaccination. To improve the efficacy of inactivated LPAI H9N2 vaccine, attenuated Salmonella enterica serovar Typhimurium was used for oral co-administration of chicken interferon-α (chIFN-α) and chicken interleukin-18 (chIL-18) as natural immunomodulators.ResultsOral co-administration of S. enterica serovar Typhimurium expressing chIFN-α and chIL-18, prior to vaccination with inactivated AI H9N2 vaccine, modulated the immune response of chickens against the vaccine antigen through enhanced humoral and Th1-biased cell-mediated immunity, compared to chickens that received single administration of S. enterica serovar Typhimurium expressing either chIFN-α or chIL-18. To further test the protective efficacy of this improved vaccination regimen, immunized chickens were intra-tracheally challenged with a high dose of LPAI H9N2 virus. Combined administration of S. enterica serovar Typhimurium expressing chIFN-α and chIL-18 showed markedly enhanced protection compared to single administration of the construct, as determined by mortality, clinical severity, and feed and water intake. This enhancement of protective immunity was further confirmed by reduced rectal shedding and replication of AIV H9N2 in different tissues of challenged chickens.ConclusionsOur results indicate the value of combined administration of chIFN-α and chIL-18 using a Salmonella vaccine strain to generate an effective immunization strategy in chickens against LPAI H9N2.

Collaboration


Dive into the Young Woo Han's collaboration.

Top Co-Authors

Avatar

Seong Kug Eo

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Koanhoi Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Seong Bum Kim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jin Hyoung Kim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Abi G. Aleyas

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jin Young Choi

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Junu A. George

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Hwa Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Seon Ju Kim

Chonbuk National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge