Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Faraday Discuss ; 250(0): 335-347, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-37965681

ABSTRACT

The scarcity of n-type polymers with high electrical conductivity (σ) and power factor (PF) is the major challenge for organic thermoelectrics (OTEs). By integrating cyano functionalities and an intramolecular conformation lock, we herein synthesize a new electron-deficient building block, CNg4T2, bearing long 1,4,7,10-tetraoxahendecyl side chains, and then further develop two n-type polythiophene derivatives, CNg4T2-2FT and CNg4T2-CNT2, with 3,4-difluorothiophene and 3,3'-dicyano-2,2'-bithiophene as co-units, respectively. Compared with CNg4T2-2FT, CNg4T2-CNT2 features a deeper-positioned lowest unoccupied molecular orbital (LUMO) while maintaining a high degree of backbone coplanarity. As a consequence, the CNg4T2-CNT2 film with molecular dopant N-DMBI delivered an impressive σ of 13.2 S cm-1 and a high PF of up to 10.84 µW m-1 K-2, significantly outperforming CNg4T2-2FT and benchmark n-type polymer N2200 films. To the best of our knowledge, this PF of CNg4T2-CNT2 devices is the highest value for n-type polythiophenes in OTEs. Further characterizations indicate that the high performance of CNg4T2-CNT2-based devices is attributed to the high doping efficiency and ordered packing of polymer chains. Our study demonstrates that CNg4T2 is a highly appealing electron-deficient building block for n-type OTE polymers and also suggests that fine-tuning of the polymer backbone is a powerful approach to accessing high-performance n-type polymers for OTE devices.

2.
Nat Nanotechnol ; 18(2): 205-216, 2023 02.
Article in English | MEDLINE | ID: mdl-36522556

ABSTRACT

When under stress, plants release molecules to activate their defense system. Detecting these stress-related molecules offers the possibility to address stress conditions and prevent the development of diseases. However, detecting endogenous signalling molecules in living plants remains challenging due to low concentrations of these analytes and interference with other compounds; additionally, many methods currently used are invasive and labour-intensive. Here we show a non-destructive surface-enhanced Raman scattering (SERS)-based nanoprobe for the real-time detection of multiple stress-related endogenous molecules in living plants. The nanoprobe, which is placed in the intercellular space, is optically active in the near-infrared region (785 nm) to avoid interferences from plant autofluorescence. It consists of a Si nanosphere surrounded by a corrugated Ag shell modified by a water-soluble cationic polymer poly(diallyldimethylammonium chloride), which can interact with multiple plant signalling molecules. We measure a SERS enhancement factor of 2.9 × 107 and a signal-to-noise ratio of up to 64 with an acquisition time of ~100 ms. To show quantitative multiplex detection, we adopted a binding model to interpret the SERS intensities of two different analytes bound to the SERS hot spot of the nanoprobe. Under either abiotic or biotic stress, our optical nanosensors can successfully monitor salicylic acid, extracellular adenosine triphosphate, cruciferous phytoalexin and glutathione in Nasturtium officinale, Triticum aestivum L. and Hordeum vulgare L.-all stress-related molecules indicating the possible onset of a plant disease. We believe that plasmonic nanosensor platforms can enable the early diagnosis of stress, contributing to a timely disease management of plants.


Subject(s)
Metal Nanoparticles , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Gold/chemistry , Polymers , Glutathione , Metal Nanoparticles/chemistry
3.
Front Immunol ; 12: 778829, 2021.
Article in English | MEDLINE | ID: mdl-34868052

ABSTRACT

Since the coronavirus disease outbreak in 2019, several antibody therapeutics have been developed to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Antibody therapeutics are effective in neutralizing the virus and reducing hospitalization in patients with mild and moderate infections. These therapeutics target the spike protein of SARS-CoV-2; however, emerging mutations in this protein reduce their efficiency. In this study, we developed a universal SARS-CoV-2 neutralizing antibody. We generated a humanized monoclonal antibody, MG1141A, against the receptor-binding domain of the spike protein through traditional mouse immunization. We confirmed that MG1141A could effectively neutralize live viruses, with an EC50 of 92 pM, and that it exhibited effective Fc-mediated functions. Additionally, it retained its neutralizing activity against the alpha (UK), beta (South Africa), and gamma (Brazil) variants of SARS-CoV-2. Taken together, our study contributes to the development of a novel antibody therapeutic approach, which can effectively combat emerging SARS-CoV-2 mutations.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/therapy , SARS-CoV-2/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Antibody Affinity , Complementarity Determining Regions/chemistry , Epitopes , Humans , Immunization , Mice , Molecular Docking Simulation , Protein Interaction Domains and Motifs , Receptors, IgG/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
4.
Vaccine ; 35(33): 4126-4132, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28669617

ABSTRACT

BACKGROUND: In Korea, measles occurs mainly in infants <12months of age, who are unvaccinated. In addition, vaccine populations, including adolescents and young adults, can become infected though importation. Thus, the question arises whether the current level of herd immunity in Korea is now insufficient for protecting against measles infection. METHODS: Age-specific measles seroprevalence was evaluated by performing enzyme immunoassays and plaque reduction-neutralization tests on 3050 subjects aged 0-50years (birth cohort 1964-2014) and 480 subjects aged 2-30years (birth cohort 1984-2012). RESULTS: The overall seropositivity and measles antibody concentrations were 71.5% and 1366mIU/mL, respectively. Progressive decline in antibody levels and seropositivity were observed over time after vaccination in infants, adolescents, and young adults. The accumulation of potentially susceptible individuals in the population was confirmed by comparing data from 2010 and 2014 seroprevalence surveys. The statistical correlation between measles incidence and measles seronegativity was determined. CONCLUSIONS: Waning levels of measles antibodies with increasing time post-vaccination suggests that measles susceptibility is potentially increasing in Korea. This trend may be related to limitations of vaccine-induced immunity in the absence of natural boosting by the wild virus, compared to naturally acquired immunity triggered by measles infection. This study provides an important view into the current measles herd immunity in Korea.


Subject(s)
Antibodies, Viral/blood , Measles Vaccine/immunology , Measles/immunology , Measles/prevention & control , Adolescent , Adult , Age Factors , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Infant, Newborn , Male , Measles Vaccine/administration & dosage , Middle Aged , Neutralization Tests , Republic of Korea , Seroepidemiologic Studies , Viral Plaque Assay , Young Adult
5.
Emerg Infect Dis ; 22(1): 100-4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26691200

ABSTRACT

An outbreak of nosocomial infections with Middle East respiratory syndrome coronavirus occurred in South Korea in May 2015. Spike glycoprotein genes of virus strains from South Korea were closely related to those of strains from Riyadh, Saudi Arabia. However, virus strains from South Korea showed strain-specific variations.


Subject(s)
Genetic Variation/genetics , Middle East Respiratory Syndrome Coronavirus/genetics , Spike Glycoprotein, Coronavirus/genetics , Coronavirus Infections/epidemiology , Coronavirus Infections/genetics , Coronavirus Infections/virology , Cross Infection/epidemiology , Cross Infection/virology , Disease Outbreaks , Humans , Male , Republic of Korea/epidemiology , Saudi Arabia/epidemiology
6.
Sci Rep ; 5: 17548, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626303

ABSTRACT

Although the roles of dendritic cells (DCs) in adaptive defense have been defined well, the contribution of DCs to T cell-independent innate defense and subsequent neuroimmunopathology in immune-privileged CNS upon infection with neurotropic viruses has not been completely defined. Notably, DC roles in regulating innate CD11b(+)Ly-6C(hi) monocyte functions during neuroinflammation have not yet been addressed. Using selective ablation of CD11c(hi)PDCA-1(int/lo) DCs without alteration in CD11c(int)PDCA-1(hi) plasmacytoid DC number, we found that CD11c(hi) DCs are essential to control neuroinflammation caused by infection with neurotropic Japanese encephalitis virus, through early and increased infiltration of CD11b(+)Ly-6C(hi) monocytes and higher expression of CC chemokines. More interestingly, selective CD11c(hi) DC ablation provided altered differentiation and function of infiltrated CD11b(+)Ly-6C(hi) monocytes in the CNS through Flt3-L and GM-CSF, which was closely associated with severely enhanced neuroinflammation. Furthermore, CD11b(+)Ly-6C(hi) monocytes generated in CD11c(hi) DC-ablated environment had a deleterious rather than protective role during neuroinflammation, and were more quickly recruited into inflamed CNS, depending on CCR2, thereby exacerbating neuroinflammation via enhanced supply of virus from the periphery. Therefore, our data demonstrate that CD11c(hi) DCs provide a critical and unexpected role to preserve the immune-privileged CNS in lethal neuroinflammation via regulating the differentiation, function, and trafficking of CD11b(+)Ly-6C(hi) monocytes.


Subject(s)
Antigens, Ly/immunology , CD11c Antigen/immunology , Cell Differentiation/immunology , Central Nervous System/immunology , Encephalitis, Arbovirus/immunology , Flavivirus Infections/immunology , Monocytes/immunology , Animals , Antigens, Ly/genetics , CD11c Antigen/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cell Movement/immunology , Central Nervous System/pathology , Central Nervous System/virology , Dendritic Cells/immunology , Dendritic Cells/pathology , Encephalitis Viruses, Japanese , Encephalitis, Arbovirus/genetics , Flavivirus Infections/genetics , Mice , Mice, Transgenic , Monocytes/pathology
7.
J Neuroinflammation ; 12: 216, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26597582

ABSTRACT

BACKGROUND: Japanese encephalitis (JE), a neuroinflammation caused by zoonotic JE virus, is the major cause of viral encephalitis worldwide and poses an increasing threat to global health and welfare. To date, however, there has been no report describing the regulation of JE progression using immunomodulatory tools for developing therapeutic strategies. We tested whether blocking the 4-1BB signaling pathway would regulate JE progression using murine JE model. METHODS: Infected wild-type and 4-1BB-knockout (KO) mice were examined daily for mortality and clinical signs, and neuroinflammation in the CNS was evaluated by infiltration of inflammatory leukocytes and cytokine expression. In addition, viral burden, JEV-specific T cell, and type I/II IFN (IFN-I/II) innate responses were analyzed. RESULTS: Blocking the 4-1BB signaling pathway significantly increased resistance to JE and reduced viral burden in extraneural tissues and the CNS, rather than causing a detrimental effect. In addition, treatment with 4-1BB agonistic antibody exacerbated JE. Furthermore, JE amelioration and reduction of viral burden by blocking the 4-1BB signaling pathway were associated with an increased frequency of IFN-II-producing NK and CD4(+) Th1 cells as well as increased infiltration of mature Ly-6C(hi) monocytes in the inflamed CNS. More interestingly, DCs and macrophages derived from 4-1BB KO mice showed potent and rapid IFN-I innate immune responses upon JEV infection, which was coupled to strong induction of PRRs (RIG-I, MDA5), transcription factors (IRF7), and antiviral ISG genes (ISG49, ISG54, ISG56). Further, the ablation of 4-1BB signaling enhanced IFN-I innate responses in neuron cells, which likely regulated viral spread in the CNS. Finally, we confirmed that blocking the 4-1BB signaling pathway in myeloid cells derived from hematopoietic stem cells (HSCs) played a dominant role in ameliorating JE. In support of this finding, HSC-derived leukocytes played a dominant role in generating the IFN-I innate responses in the host. CONCLUSIONS: Blocking the 4-1BB signaling pathway ameliorates JE via divergent enhancement of IFN-II-producing NK and CD4(+) Th1 cells and mature Ly-6C(hi) monocyte infiltration, as well as an IFN-I innate response of myeloid-derived cells. Therefore, regulation of the 4-1BB signaling pathway with antibodies or inhibitors could be a valuable therapeutic strategy for the treatment of JE.


Subject(s)
Antigens, Ly/biosynthesis , Encephalitis, Japanese/metabolism , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Monocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/deficiency , Animals , Cell Differentiation/physiology , Encephalitis, Japanese/immunology , Encephalitis, Japanese/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Signal Transduction/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
8.
Emerg Infect Dis ; 21(11): 2084-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26488745

ABSTRACT

In May 2015, Middle East respiratory syndrome coronavirus infection was laboratory confirmed in South Korea. Patients were a man who had visited the Middle East, his wife, and a man who shared a hospital room with the index patient. Rapid laboratory confirmation will facilitate subsequent prevention and control for imported cases.


Subject(s)
Coronavirus Infections/diagnosis , Coronavirus/pathogenicity , Cross Infection/virology , Travel , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Cross Infection/transmission , Humans , Male , Middle East , Republic of Korea/epidemiology
9.
Genome Announc ; 3(4)2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26272558

ABSTRACT

The full genome sequence of a Middle East respiratory syndrome coronavirus (MERS-CoV) was identified from cultured and isolated in Vero cells. The viral genome sequence has high similarity to 53 human MERS-CoVs, ranging from 99.5% to 99.8% at the nucleotide level.

10.
J Vet Med Sci ; 77(4): 395-403, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25502364

ABSTRACT

Newcastle disease (ND) is a highly contagious disease of chickens causing significant economic losses worldwide. Due to limitations in the efficacy against currently circulating ND viruses, existing vaccination strategies require improvements, and incorporating immunomodulatory cytokines with existing vaccines might be a novel approach. Here, we investigated the systemic and mucosal immunomodulatory properties of oral co-administration of chicken interleukin-18 (chIL-18) and chicken interferon-α (chIFN-α) using attenuated Salmonella enterica serovar Typhimurium on an inactivated ND vaccine. Our results demonstrate that oral administration of S. enterica serovar Typhimurium expressing chIL-18 or chIFN-α provided enhanced systemic and mucosal immune responses, as determined by serum hemagglutination inhibition antibody and NDV Ag-specific IgG as well as NDV Ag-specific IgA in lung and duodenal lavages of chickens immunized with inactivated ND vaccine via the intramuscular or intranasal route. Notably, combined oral administration of S. enterica serovar Typhimurium expressing chIL-18 and chIFN-α significantly enhanced systemic and mucosal immunity in ND-vaccinated chickens, compared to single administration of S. enterica serovar Typhimurium expressing chIL-18 or chIFN-α. In addition, oral co-administration of S. enterica serovar Typhimurium expressing chIL-18 and chIFN-α provided enhanced NDV Ag-specific proliferation of peripheral blood mononuclear cells and Th1-biased cell-mediated immunity, compared to single administration of either construct. Therefore, our results provide valuable insight into the modulation of systemic and mucosal immunity by incorporation of immunomodulatory chIL-18 and chIFN-α using Salmonella vaccines into existing ND vaccines.


Subject(s)
Chickens , Interferon-alpha/metabolism , Interleukin-18/metabolism , Interleukin-18/pharmacology , Newcastle Disease/prevention & control , Salmonella typhimurium/metabolism , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Interferon-alpha/administration & dosage , Interferon-alpha/pharmacology , Interleukin-18/administration & dosage , Specific Pathogen-Free Organisms , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Vaccines/administration & dosage
11.
PLoS Pathog ; 10(9): e1004319, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25188232

ABSTRACT

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-6C(high) 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.


Subject(s)
Brain/immunology , Encephalitis Virus, Japanese/pathogenicity , Encephalitis, Japanese/complications , Inflammation/etiology , Signal Transduction , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/physiology , Animals , Blotting, Western , Brain/metabolism , Brain/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Encephalitis, Japanese/virology , Enzyme-Linked Immunosorbent Assay , Immunity, Innate , Inflammation/metabolism , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/virology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
BMC Vet Res ; 8: 105, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22776696

ABSTRACT

BACKGROUND: Control 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. RESULTS: Oral 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. CONCLUSIONS: Our 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.


Subject(s)
Influenza in Birds/prevention & control , Interferon-alpha/metabolism , Interleukin-18/metabolism , Salmonella typhimurium/physiology , Th1 Cells/physiology , Viral Vaccines/immunology , Administration, Oral , Animals , Cell Proliferation , Chickens , Influenza A Virus, H9N2 Subtype , Interferon-alpha/administration & dosage , Interferon-alpha/pharmacology , Interleukin-18/administration & dosage , Interleukin-18/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, Inactivated/immunology , Virus Replication , Virus Shedding
13.
Eur J Immunol ; 42(10): 2655-66, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22706912

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/immunology , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 2/metabolism , Animals , Antigens, Viral/metabolism , CD11c Antigen/metabolism , CD8 Antigens/metabolism , Cell Proliferation , Cytotoxicity, Immunologic , Dendritic Cells/virology , Lymphocyte Activation , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
14.
Immune Netw ; 12(2): 48-57, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22740790

ABSTRACT

Acute viral encephalitis caused by neurotrophic viruses, such as mosquito-borne flaviviruses, is an emerging and re-emerging disease that represents an immense global health problem. Considerable progression has been made in understanding the pathogenesis of acute viral encephalitis, but the immune-pathological processes occurring during the progression of encephalitis and the roles played by various molecules and cellular components of the innate and adaptive systems still remain undefined. Recent findings reveal the significant contribution of Toll-like receptors (TLRs) and regulatory CD4(+) T cells in the outcomes of infectious diseases caused by neurotrophic viruses. In this review, we discuss the ample evidence focused on the roles of TLRs and CD4(+) helper T cell subsets on the progression of acute viral encephalitis. Finally, we draw attention to the importance of these molecules and cellular components in defining the pathogenesis of acute viral encephalitis, thereby providing new therapeutic avenues for this disease.

15.
Microbiol Immunol ; 56(8): 529-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22551253

ABSTRACT

The co-administration of two or more cytokines may generate additive or synergistic effects for controlling infectious diseases. However, the practical use of cytokine combinations for the modulation of immune responses against inactivated vaccine has not been demonstrated in livestock yet, primarily due to protein stability, production, and costs associated with mass administration. In light of the current situation, we evaluated the immunomodulatory functions of the combined administration of swine interleukin-18 (swIL-18) and interferon-α (swIFN-α) against an inactivated PrV vaccine using attenuated Salmonella enterica serovar Typhimurium as a cytokine delivery system. Co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α produced enhanced Th1-biased humoral and cellular immune responses against the inactivated PrV vaccine, when compared to single administration of S. enterica serovar Typhimurium expressing either swIL-18 or swIFN-α. Also, enhanced immune responses in co-administered piglets occurred rapidly after virulent PrV challenge, and piglets that received co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α displayed a greater alleviation of clinical severity following the virulent PrV challenge, as determined by clinical scores and cumulative daily weight gain. Furthermore, this enhancement was confirmed by reduced nasal shedding of PrV following viral challenge. Therefore, these results suggest that oral co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α provide enhanced Th1-biased immunity against inactivated PrV vaccine to alleviate clinical signs caused by PrV challenge.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Genetic Vectors/administration & dosage , Herpesvirus 1, Suid/immunology , Interferon-alpha/administration & dosage , Interleukin-18/administration & dosage , Pseudorabies Vaccines/immunology , Th1 Cells/immunology , Animals , Body Weight , Disease Models, Animal , Immunity, Cellular , Immunity, Humoral , Interferon-alpha/genetics , Interleukin-18/genetics , Pseudorabies/immunology , Pseudorabies/pathology , Pseudorabies/prevention & control , Pseudorabies Vaccines/administration & dosage , Salmonella typhimurium/genetics , Severity of Illness Index , Swine , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
16.
Vet Microbiol ; 157(3-4): 448-55, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22245401

ABSTRACT

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.


Subject(s)
Antiviral Agents/therapeutic use , Chickens/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/drug therapy , Interferon-alpha/therapeutic use , Interleukin-18/therapeutic use , Salmonella typhimurium , Administration, Oral , Animals , Antibodies, Viral/blood , Antiviral Agents/immunology , Chickens/virology , Hemagglutination Inhibition Tests , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/immunology , Interferon-alpha/immunology , Interleukin-18/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Virus Replication , Virus Shedding
17.
Vet Microbiol ; 155(2-4): 172-82, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-21940117

ABSTRACT

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.


Subject(s)
Interleukin-18/genetics , Pseudorabies Vaccines/immunology , Pseudorabies/immunology , Salmonella typhimurium/genetics , Swine Diseases/immunology , Th1 Cells/immunology , Administration, Oral , Animals , Herpesvirus 1, Suid/immunology , Immunity, Cellular , Interleukin-4/metabolism , Leukocytes, Mononuclear/immunology , Pseudorabies/prevention & control , Sus scrofa , Swine , Swine Diseases/prevention & control , Vaccines, Inactivated/immunology
18.
Comp Immunol Microbiol Infect Dis ; 34(4): 369-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21719106

ABSTRACT

The enhanced effect of cytokine combinations has been assessed empirically, based on their immunobiological mechanisms. However, far less is known of the enhanced protection of practical cytokine combinations against viral infection in the livestock industry, due to cost and production issues associated with mass administration. This study demonstrates the enhanced protection of oral co-administration of swine interferon-α (swIFN-α) and interleukin-18 (swIL-18) against infection with transmissible gastroenteritis virus (TGEV) in piglets using attenuated Salmonella enterica serovar Typhimurium as carrier of cytokine proteins. A single oral co-administration of S. enterica serovar Typhimurium expressing swIFN-α and swIL-18 induced enhanced alleviation of the severity of diarrhea caused by TGEV infection, compared to piglets administered S. enterica serovar Typhimurium expressing swIFN-α or swIL-18 alone. This enhancement was further observed by the reduction of TGEV shedding and replication, and the expression of IFN-stimulated gene products in the intestinal tract. The results suggest that the combined administration of the swIFN-α and swIL-18 cytokines using attenuated S. enterica serovar Typhimurium as an oral carrier provides enhanced protection against intestinal tract infection with TGEV.


Subject(s)
Gastroenteritis, Transmissible, of Swine/prevention & control , Immunity, Active , Interferon-alpha/immunology , Interleukin-18/immunology , Intestines/immunology , Salmonella typhimurium/genetics , Transmissible gastroenteritis virus/drug effects , Vaccination , Vaccines, Attenuated/administration & dosage , Administration, Oral , Animals , Female , Gastroenteritis, Transmissible, of Swine/immunology , Gastroenteritis, Transmissible, of Swine/metabolism , Gastroenteritis, Transmissible, of Swine/virology , Interferon-alpha/genetics , Interleukin-18/genetics , Intestines/virology , Livestock , Mice , Plasmids , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/chemistry , Salmonella typhimurium/immunology , Swine , Transfection , Transmissible gastroenteritis virus/growth & development , Transmissible gastroenteritis virus/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Load/drug effects , Viral Load/immunology , Virus Replication/drug effects , Virus Replication/immunology
19.
Vet Microbiol ; 154(1-2): 140-51, 2011 Dec 29.
Article in English | MEDLINE | ID: mdl-21764226

ABSTRACT

Low pathogenic avian influenza (LPAI) H9N2 has attracted considerable attention due to severe commercial losses in the poultry industry. Furthermore, avian influenza virus (AIV) H9N2-infected chickens can be a reservoir for viral transmission to mammals including pigs and humans, complicating control of viral mutants. Chicken interferon-alpha (chIFN-α) may be useful as an exogenous antiviral agent to control AIV H9N2 infection. However, a superior vehicle for administration of chIFN-α is needed because of challenges of protein stability, production cost, and labor associated with mass administration. Presently, oral administration of single dose of attenuated Salmonella enterica serovar Typhimurium expressing chIFN-α alleviated clinical signs and histopathological changes caused by respiratory infection with AIV H9N2 and reduced the excretion of virus in cloacal swab samples. Similarly, chickens administered S. enterica serovar Typhimurium expressing chIFN-α showed inhibited replication of AIV H9N2 in several different tissues including trachea, lung, cecal tonsil, and brain. Furthermore, immune responses specific for challenged AIV H9N2 were enhanced in chickens administered S. enterica serovar Typhimurium expressing chIFN-α, as determined by hemagglutination inhibition assay of sera, proliferation and IFN-γ and interleukin-4 expression by AIV H9N2 antigen-stimulated peripheral blood mononuclear cells and splenocytes. Therefore, oral administration of S. enterica serovar Typhimurium expressing chIFN-α can successfully control clinical signs caused by respiratory infection with AIV H9N2, which provides valuable insight into the use of attenuated Salmonella vaccine as an oral delivery system of chIFN-α to prevent the replication of AIV H9N2 in respiratory tract.


Subject(s)
Chickens/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Interferon-alpha/pharmacology , Respiratory Tract Infections/veterinary , Salmonella typhimurium/immunology , Adaptive Immunity , Administration, Oral , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Base Sequence , Hemagglutination Inhibition Tests , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza in Birds/immunology , Interferon-alpha/genetics , Interferon-gamma/immunology , Interleukin-4/immunology , Leukocytes, Mononuclear/immunology , Lung/pathology , Lung/virology , Molecular Sequence Data , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Salmonella typhimurium/genetics , Spleen/cytology , Spleen/immunology , Trachea/pathology , Trachea/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virus Replication
20.
Comp Immunol Microbiol Infect Dis ; 34(4): 335-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543119

ABSTRACT

Oral administration of attenuated Salmonella vaccine may provide valuable advantages such as low cost, easy preparation, and safety. Attenuated Salmonella vaccines also serve as carriers of foreign antigens and immunomodulatory cytokines. Presently, an attenuated Salmonella enterica serovar Typhimurium strain was used as a carrier for open reading frame 7 (ORF7) protein of porcine reproductive and respiratory syndrome virus (PRRSV), a swine pathogen of significant global economic importance. Initially, an attenuated S. enterica serovar Typhimurium expressing ORF7 gene derived from PRRSV Korean isolate was constructed. Following oral administration of a single dose of the attenuated Salmonella vaccine expressing PRRSV ORF7, humoral and cell-mediated immune responses specific for ORF7 were induced at both systemic and mucosal sites including spleen, mesenteric lymph node, Peyer's patch, and laminar propria, as evaluated by determining serum ORF7-specific IgG and mucosal IgA responses, as well as Th1- and Th2-type cytokine production from antigen-stimulated T cells. The induced humoral responses were sustained for at least 12weeks post-immunization. In particular, the immunized mice displayed immune responses to both the foreign ORF7 antigen and Salmonella itself. The results indicate the value of attenuated S. enterica serovar Typhimurium as an oral carrier of PRRSV antigenic proteins to induce effective systemic and mucosal immunity.


Subject(s)
Immunity, Mucosal , Lymph Nodes/immunology , Lymphocytes/immunology , Peyer's Patches/immunology , Porcine respiratory and reproductive syndrome virus/genetics , Salmonella Infections, Animal/prevention & control , Spleen/immunology , Vaccination , Vaccines, Attenuated/administration & dosage , Administration, Oral , Animals , Antigens, Bacterial , Antigens, Viral/immunology , Base Sequence , Cells, Cultured , Female , Immunoglobulin A/analysis , Immunoglobulin A/biosynthesis , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Lymph Nodes/cytology , Lymph Nodes/microbiology , Lymphocyte Activation , Lymphocytes/microbiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Open Reading Frames/genetics , Open Reading Frames/immunology , Peyer's Patches/cytology , Peyer's Patches/microbiology , Porcine respiratory and reproductive syndrome virus/chemistry , Porcine respiratory and reproductive syndrome virus/immunology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Spleen/cytology , Spleen/microbiology , Swine , Th1-Th2 Balance
SELECTION OF CITATIONS
SEARCH DETAIL
...