Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 9141, 2024 04 21.
Article in English | MEDLINE | ID: mdl-38644371

ABSTRACT

Tuberculosis remains a large health threat, despite the availability of the tuberculosis vaccine, BCG. As BCG efficacy gradually decreases from adolescence, BCG-Prime and antigen-booster may be an efficient strategy to confer vaccine efficacy. Mycobacterial DNA-binding protein 1 (MDP1, namely Rv2986c, hupB or HU) is a major Mycobacterium tuberculosis protein that induces vaccine-efficacy by co-administration with CpG DNA. To produce MDP1 for booster-vaccine use, we have created recombinant MDP1 produced in both Escherichia coli (eMDP1) and Mycolicibacterium smegmatis (mMDP1), an avirulent rapid-growing mycobacteria. We tested their immunogenicity by checking interferon (IFN)-gamma production by stimulated peripheral blood cells derived from BCG-vaccinated individuals. Similar to native M. tuberculosis MDP1, we observed that most lysin resides in the C-terminal half of mMDP1 are highly methylated. In contrast, eMDP1 had less post-translational modifications and IFN-gamma stimulation. mMDP1 stimulated the highest amount of IFN-gamma production among the examined native M. tuberculosis proteins including immunodominant MPT32 and Antigen 85 complex. MDP1-mediated IFN-gamma production was more strongly enhanced when combined with a new type of CpG DNA G9.1 than any other tested CpG DNAs. Taken together, these results suggest that the combination of mMDP1 and G9.1 possess high potential use for human booster vaccine against tuberculosis.


Subject(s)
BCG Vaccine , Bacterial Proteins , DNA-Binding Proteins , Interferon-gamma , Mycobacterium tuberculosis , Protein Processing, Post-Translational , Humans , Interferon-gamma/metabolism , Bacterial Proteins/immunology , BCG Vaccine/immunology , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mycobacterium tuberculosis/immunology , Recombinant Proteins/immunology , Oligodeoxyribonucleotides/pharmacology , Tuberculosis/prevention & control , Tuberculosis/immunology , CpG Islands , Mycobacterium smegmatis/immunology , Mycobacterium smegmatis/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , Female
2.
Vaccine ; 41(8): 1447-1456, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36702691

ABSTRACT

Mucosal vaccines offer several advantages over transdermal vaccines, including the ability to acquire systemic and mucosal immunities. Smoking is a huge public health threat and major risk factor for various diseases that exacerbate or prolong respiratory symptoms and conditions. However, its impact on the efficacy of mucosal vaccines remains partially explored. Thus, this study investigates the effects of smoking on mucosal vaccine reactivity by assessing the induction of Th1 immunity, a vital response in infection defense. Cigarette smoke condensate was prepared as a substitute for mainstream smoke. We intranasally administered diphtheria toxoid as an antigen and natural CpG oligonucleotide G9.1, which enhances the Th1-type antibody (Ab) response in a plasmacytoid dendritic cells (pDCs) dependent manner, as an adjuvant to mice to assess the effect of cigarette smoke condensate on Ab responses. The mechanism of its effect was evaluated using human peripheral blood mononuclear cells and their pDC-rich fraction cultured with or without G9.1. In mice, cigarette smoke condensate tended to decrease diphtheria toxoid-specific Ab response, with a higher reduction in Th1-type IgG2 Ab response than in Th2-type IgG1 Ab response. In human peripheral blood mononuclear cells, cigarette smoke condensate significantly reduced the induction of IFN-α production by G9.1. Moreover, G9.1-induced increases in the CD83 expression in pDCs and the CD80 expression in DCs were suppressed via treatment with cigarette smoke condensate. Among the mechanisms suggested were decreased expression of toll-like receptor 9 mRNA, decreased expression of mRNA for IFN regulatory factor 7, and increased CpG methylation of its promoter region. The analysis of Tbet and GATA3 expressions revealed that cigarette smoke condensate exhibits Th1-directed immunostimulatory activity at a steady state but becomes more Th2-directed under G9.1 stimulation. In conclusion, smoking could reduce mucosal vaccine responses by decreasing pDC activation and, consequently, Th1-dominant immunity.


Subject(s)
Cigarette Smoking , Interferon-alpha , Animals , Humans , Mice , Dendritic Cells , Diphtheria Toxoid , Leukocytes, Mononuclear , RNA, Messenger/genetics , Smoking
3.
Tuberculosis (Edinb) ; 128: 102067, 2021 05.
Article in English | MEDLINE | ID: mdl-33752142

ABSTRACT

Tuberculosis is a major threat to global health and its increased incidence in adolescents as well as onset in the elderly presents a serious problem. One strategy to control tuberculosis involves taking advantage of Bacillus Calmette-Guérin's (BCG) superior effects on childhood tuberculosis. Accordingly, here we aimed to develop a booster vaccine for adults who received the BCG vaccine during early childhood. Therefore, we first devised a system to assess the efficacy of a candidate booster vaccine. Specifically, variant strain BCG-II, a minor component of BCG-Tokyo strain, which elicits weak immunity, was administered to guinea pigs. Vaccine-induced immunity and protection against Mycobacterium tuberculosis (Mtb) infection were evaluated using skin delayed-type hypersensitivity (DTH) and Mtb colony forming unit counts in organs, respectively. Candidate booster vaccine containing the mycobacterial DNA-binding protein 1 (MDP1) as antigen and CpG oligodeoxynucleotide G9.1 as adjuvant increased T-bet expression and IFN-γ production in human peripheral blood mononuclear cells. Intradermal administration of MDP1 or MDP1 and G9.1 to unimmunized guinea pigs produced DTH on MDP1-inoculated skin. Boosting BCG-II-primed guinea pigs with this protocol effectively enhanced DTH against MDP1 and protection against Mtb infection, particularly when combined with G9.1. The candidate vaccine may contribute to efforts to prevent tuberculosis.


Subject(s)
Bacterial Proteins/immunology , DNA-Binding Proteins/immunology , Oligodeoxyribonucleotides/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/blood , BCG Vaccine/immunology , Cytokines/immunology , Female , Guinea Pigs , Humans , Immunization, Secondary , Leukocytes, Mononuclear , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium bovis
4.
Vaccine ; 37(36): 5382-5389, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31345642

ABSTRACT

This study examined the protective efficacy of and immune response to a nasal influenza vaccine combined with a novel mucosal oligodeoxynucleotide (ODN) adjuvant, CpG ODN G9.1 (G9.1), in a model of infection limited to the upper respiratory tract (URT) and a model of infection in the lower respiratory tract (LRT). Mice were nasally primed with an A/California/7/2009 (Cal7) split vaccine (X179A) plus G9.1 and were then nasally given a booster with X179A alone. When mice were challenged with either a large (infection of the LRT) or small (infection limited to the URT) volume of live Cal7 influenza virus, mice nasally given G9.1 combined with X179A had a markedly higher rate of protection against infection limited to the URT. Moreover, this group of mice promptly recovered from an infection of the LRT. When mice were subcutaneously (s.c.) given X179A as a current form of vaccination, they had no protection from an infection limited to the URT but they did recover from an infection of the LRT. The patterns of protection were closely correlated with influenza virus-specific mucosal secretory IgA (SIgA) or serum IgG antibody (Ab) responses. Thus, SIgA Abs responses play an important role in protection from an infection limited to the URT while influenza virus-specific serum IgG Ab responses help to protect from an infection of the LRT. A finding of note is that lungs from mice nasally given G9.1 had low levels of type I IFN-associated protein- and transcription factor-specific mRNA expression. These results suggest that nasal G9.1 can be used as an effective and safe mucosal adjuvant for influenza vaccines since this nasal vaccine system elicits both mucosal SIgA and serum IgG Ab responses that provide complete protection without inducing potent inflammatory responses.


Subject(s)
Influenza, Human/immunology , Influenza, Human/prevention & control , Oligodeoxyribonucleotides/immunology , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Kinetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae/immunology , Orthomyxoviridae/pathogenicity , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/therapeutic use
5.
Hum Vaccin Immunother ; 11(3): 755-60, 2015.
Article in English | MEDLINE | ID: mdl-25751765

ABSTRACT

Bacterial DNA comprising palindromic sequences and containing unmethylated CpG is recognized by toll-like receptor 9 of plasmacytoid dendritic cells (pDCs) and induces the production of interferon-α and chemokines, leading to the activation of a Th1 immune response. Therefore, synthetic equivalents of bacterial DNA (CpG oligodeoxynucleotides) have been developed for clinical applications. They are usually phosphorothioated for in vivo use; this approach also leads to adverse effects as reported in mouse models.Mucosal vaccines that induce both mucosal and systemic immunity received substantial attention in recent years. For their development, phosphodiester-linked oligodeoxynucleotides, including the sequence of a palindromic CpG DNA may be advantageous as adjuvants because their target pDCs are present right there, in the mucosa of the vaccination site. In addition, the probability of adverse effects is believed to be low. Here, we review the discovery of such CpG oligodeoxynucleotides and their possible use as mucosal adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Mucosal/drug effects , Oligodeoxyribonucleotides/pharmacology , Adjuvants, Immunologic/administration & dosage , Administration, Mucosal , Animals , Dendritic Cells/drug effects , Humans , Oligodeoxyribonucleotides/administration & dosage
6.
PLoS One ; 9(2): e88846, 2014.
Article in English | MEDLINE | ID: mdl-24586411

ABSTRACT

BACKGROUND: CpG oligodeoxynucleotides (ODNs), resembling bacterial DNA, are currently tested in clinical trials as vaccine adjuvants. They have the nuclease-resistant phosphorothioate bond; the immune responses elicited differ according to the CpG ODN sequence and vaccination method. To develop a CpG ODN that can induce plasmacytoid dendritic cell (pDC)-mediated T(H)1 immunity through the mucosa, we constructed phosphodiester G9.1 comprising one palindromic CpG motif with unique polyguanosine-runs that allows degradation similar to naturally occurring bacterial DNA. METHODS: T(H)1 and T(H)2 immunity activation was evaluated by cytokine production pattern and T-bet/GATA-3 ratio in human peripheral blood mononuclear cells and mouse bone marrow cells. Adjuvanticity was evaluated in mice administered G9.1 with diphtheria toxoid (DT) through nasal vaccination. RESULTS: G9.1 exhibited stronger IFN-α-inducing activity than A-class CpG ODN2216 and increased T-bet/GATA-3 ratio by enhancing T-bet expression. Nasally administered G9.1 plus DT induced DT-specific mucosal IgA and serum IgG, but not IgE, responses with antitoxin activity in C57BL/6 and BALB/c mice, possibly due to IFN/BAFF production. Induction of T(H)1, but not T(H)2-type Abs depended completely on pDCs, the first in vivo demonstration by CpG ODNs. CONCLUSIONS: G9.1 is a promising mucosal adjuvant for induction of pDC-mediated T(H)1 immunity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Dendritic Cells/immunology , Mucous Membrane/immunology , Oligodeoxyribonucleotides/immunology , Th1 Cells/immunology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , DNA, Bacterial/immunology , Dendritic Cells/drug effects , Diphtheria Toxoid/immunology , Female , Humans , Interferon-alpha/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/drug effects , Th1 Cells/drug effects
7.
Immun Ageing ; 10(1): 25, 2013 Jun 22.
Article in English | MEDLINE | ID: mdl-23799936

ABSTRACT

BACKGROUND: Mycobacterium bovis bacillus Calmette Guérin (BCG) vaccine, which has been inoculated to more than one billion people world-wide, has significant effect in preventing tuberculous meningitis and miliary tuberculosis (TB) in neonate and early childhood. However, BCG fails to adequately protect against pulmonary TB and reactivation of latent infections in adults. To overcome this problem, adequate booster is urgently desired in adult who received prior BCG vaccination, and appropriate animal models that substitute human cases would be highly valuable for further experimentation. FINDINGS: The booster effect of the synthesized CpG oligomer (Oligo-B) on aged mice which had been primarily vaccinated with BCG at the age of 4-week old. The specific Th1 type reaction, production of interferon-γ, in response to TB antigens, purified protein derivatives (PPD) and protection against challenge with Mycobacterium tuberculosis (MTB) H37Rv decreased with increasing age and were not observed in 89-week old mice. In order to rejuvenate the Th1 type response against PPD and protection activity against MTB infection, Oligo-B, which is known to augment Th1 responses, was administered as a booster to 81-90-week old mice (late 50's in human equivalent) vaccinated with BCG at 4-week old. The boosting with Oligo-B increased the number of CD4+ CD44high CD62Lhigh, central memory type T cell. Furthermore, the Oligo-B boosting rejuvenated the ability of mice to protect against infection with MTB H37Rv. CONCLUSIONS: Th1-adjuvant CpG oligo DNA, such as Oligo-B, may be a promising booster when coupled with BCG priming.

8.
Cancer Sci ; 101(3): 767-73, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20067466

ABSTRACT

Glutathione S-transferase mu (GSTM1) is mainly known as a detoxification enzyme but it has also been shown to be a negative regulator of apoptosis-related signaling cascades. Recently GSTM1 has been reported to be a significant risk factor for hematological relapse in childhood acute lymphoblastic leukemia, although the underlying mechanism remains largely unknown. Glucocorticoids play a crucial role in the treatment of childhood acute lymphoblastic leukemia, therefore we hypothesized that GSTM1 plays important roles in glucocorticoid-induced apoptotic pathways. To clarify the relationship between GSTM1 and drug resistance, GSTM1 was transfected into a T-acute lymphoblastic leukemia cell line, CCRF-CEM (CEM), and we established the GSTM1-expressing cell lines CEM/M1-4 and CEM/M1-9. Transduction of GSTM1 into CEM selectively decreased cellular sensitivity to dexamethasone in a manner that was independent of glutathione conjugation, but was due to apoptosis inhibition. Dexamethasone-induced p38-MAPK and Bim activation were concomitantly suppressed. Interestingly, nuclear factor kappa b (NF-kappaB) p50 activity was upregulated in GSTM1-expressing CEM. Inhibition of NF-kappaB by the pharmacological agent BAY11-7082 greatly enhanced the sensitivity of the GSTM1-expressing CEM to dexamethasone and was accompanied by an increase in Bim expression. Thus, we propose that GSTM1, a novel regulator of dexamethasone-induced apoptosis, causes dexamethasone resistance by suppression of Bim through dual mechanisms of both downregulation of p38-MAPK and upregulation of NF-kappaB p50.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis/drug effects , Dexamethasone/pharmacology , Glutathione Transferase/physiology , Membrane Proteins/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Bcl-2-Like Protein 11 , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , NF-kappa B p50 Subunit/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Immunol ; 177(7): 4841-52, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16982926

ABSTRACT

CpG DNA induces plasmacytoid dendritic cells (pDC) to produce type I IFN and chemokines. However, it has not been fully elucidated how the TLR9 signaling pathway is linked to these gene expressions. We examined the mechanisms involving the TLR9 and type I IFN signaling pathways, in relation to CpG DNA-induced IFN-alpha, IFN regulatory factor (IRF)-7, and chemokines CXCL10 and CCL3 in human pDC. In pDC, NF-kappaB subunits p65 and p50 were constitutively activated. pDC also constitutively expressed IRF-7 and CCL3, and the gene expressions seemed to be regulated by NF-kappaB. CpG DNA enhanced the NF-kappaB p65/p50 activity, which collaborated with p38 MAPK to up-regulate the expressions of IRF-7, CXCL10, and CCL3 in a manner independent of type I IFN signaling. We then examined the pathway through which IFN-alpha is expressed. Type I IFN induced the expression of IRF-7, but not of IFN-alpha, in a NF-kappaB-independent way. CpG DNA enabled the type I IFN-treated pDC to express IFN-alpha in the presence of NF-kappaB/p38 MAPK inhibitor, and chloroquine abrogated this effect. With CpG DNA, IRF-7, both constitutively and newly expressed, moved to the nuclei independently of NF-kappaB/p38 MAPK. These findings suggest that, in CpG DNA-stimulated human pDC, the induction of IRF-7, CXCL10, and CCL3 is mediated by the NF-kappaB/p38 MAPK pathway, and that IRF-7 is activated upstream of the activation of NF-kappaB/p38 MAPK in chloroquine-sensitive regulatory machinery, thereby leading to the expression of IFN-alpha.


Subject(s)
Chemokines/biosynthesis , CpG Islands/immunology , Dendritic Cells/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Blotting, Western , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Humans , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon-alpha/immunology , Interferon-alpha/metabolism , NF-kappa B/immunology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , p38 Mitogen-Activated Protein Kinases/immunology
10.
J Leukoc Biol ; 74(5): 942-51, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12960242

ABSTRACT

Leukocytosis in tobacco smokers has been well recognized; however, the exact cause has not been elucidated. To test the hypothesis that tobacco nicotine stimulates neutrophils in the respiratory tract to produce IL-8, which causes neutrophilia in vivo, we examined whether nicotine induces neutrophil-IL-8 production in vitro; the causative role of NF-kappaB in its production, in association with the possible production of reactive oxygen intermediates that activate NF-kappaB; and the nicotinic acetylcholine receptors (nAChRs) involved in IL-8 production. Nicotine stimulated neutrophils to produce IL-8 in both time- and concentration-dependent manners with a 50% effective concentration of 1.89 mM. A degradation of IkappaB-alpha/beta proteins and an activity of NF-kappaB p65 and p50 were enhanced following nicotine treatment. The synthesis of superoxide and the oxidation of dihydrorhodamine 123 (DHR) were also enhanced. The NOS inhibitor, nomega-Nitro-l-arginine methyl ester, prevented nicotine-induced IL-8 production, with an entire abrogation of DHR oxidation, IkappaB degradation, and NF-kappaB activity. Neutrophils spontaneously produced NO whose production was not increased, but rather decreased by nicotine stimulation, suggesting that superoxide, produced by nicotine, generates peroxynitrite by reacting with preformed NO, which enhances the NF-kappaB activity, thereby producing IL-8. The nAChRs seemed to be involved in IL-8 production. In smokers, blood IL-8 levels were significantly higher than those in nonsmokers. In conclusion, nicotine stimulates neutrophil-IL-8 production via nAChR by generating peroxynitrite and subsequent NF-kappaB activation, and the IL-8 appears to contribute to leukocytosis in tobacco smokers.


Subject(s)
Interleukin-8/blood , NF-kappa B/blood , Neutrophils/immunology , Nicotine/pharmacology , Peroxynitrous Acid/blood , Humans , I-kappa B Proteins/blood , Interleukin-8/biosynthesis , Kinetics , NF-KappaB Inhibitor alpha , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Peroxidase/blood , Protein Transport , Reactive Oxygen Species/metabolism , Smoking/blood , Tumor Necrosis Factor-alpha/pharmacology
12.
J Leukoc Biol ; 72(5): 1011-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429724

ABSTRACT

Human plasmacytoid or CD4(+)CD11c(-) type 2 dendritic cell precursors (PDC) were identified as natural type I interferon (IFN)-producing cells in response to viral and bacterial infection. They represent effector cells of innate immunity and link it to the distinct adaptive immunity by differentiating into mature DC. It has been reported that oligodeoxyribonucleotides containing unmethylated CpG motifs (CpG DNA) stimulate PDC to produce IFN-alpha, but the molecular mechanisms involved remain unknown. We found that CpG-DNA-induced IFN-alpha production in PDC was completely impaired by the inhibitor of the p38 mitogen-activated protein kinase (MAPK) pathway. Expression of IFN regulatory factor (IRF)-7 was enhanced by CpG-DNA treatment, which was preceded by the phosphorylation of signal transducer and activator of transcription (STAT)1 on Tyr-701, as well as its enhanced phosphorylation on Ser-727. All of these events were also suppressed by the p38 MAPK inhibitor. STAT1, STAT2, and IRF-9, components of IFN-stimulated gene factor 3 (ISGF3), were recognized in the nuclear fraction of CpG-DNA-treated cells. Neither anti-IFN-alpha/beta antibodies (Ab) nor anti-IFNAR Ab suppressed STAT1 phosphorylation, enhancement of IRF-7 expression, or IFN-alpha production in the early phase of the culture. These results suggest that CpG DNA induces p38 MAPK-dependent phosphorylation of STAT1 in a manner independent of IFN-alpha/beta, which may cause ISGF3 formation to increase the transcription of the IRF-7 gene, thereby leading to IFN-alpha production in human PDC.


Subject(s)
DNA-Binding Proteins/metabolism , Dendritic Cells/immunology , Interferon-alpha/biosynthesis , Mitogen-Activated Protein Kinases/physiology , Oligodeoxyribonucleotides/pharmacology , Trans-Activators/metabolism , Cell Nucleus/chemistry , Cells, Cultured , DNA-Binding Proteins/analysis , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3 , Interferon Regulatory Factor-7 , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Interferon-alpha/genetics , Kinetics , Models, Immunological , Phosphorylation , RNA, Messenger/biosynthesis , STAT1 Transcription Factor , STAT2 Transcription Factor , Serine/metabolism , Stem Cells/drug effects , Stem Cells/immunology , Trans-Activators/analysis , Trans-Activators/chemistry , Transcription Factors/analysis , Transcription Factors/biosynthesis , Tyrosine/metabolism , p38 Mitogen-Activated Protein Kinases
13.
Jpn J Infect Dis ; 55(2): 37-44, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12082305

ABSTRACT

DNA containing an unmethylated CpG motif has a potent immunostimulatory effect on the vertebrate immune system. Because such CpG motifs are relatively common in bacterial DNA, but rare in mammalian animal and plant DNA, they may be an evolutionary adaptation augmenting innate immunity, most likely in response to pathogens that replicate within the host cells, such as viruses and intracellular bacteria. Microbial infection induces innate immunity by triggering pattern-recognition systems. The infected cells produce proinflammatory cytokines that directly combat microbial invaders and express costimulating surface molecules, which develop adaptive immunity by inducing distinct T cell differentiation. Bacterial DNA with unmethylated CpG-DNA stimulates vertebrate immature immune cells to induce maturation and to produce TNF-alpha as well as Th1-type cytokines, IL-12 and IFN-gamma. Therefore, CpG-DNA functions as an adjuvant for regulating the initiation of Th1 differentiation. The roles of immunostimulatory CpG motifs in DNA vaccine developments and in therapeutic applications have been discussed.


Subject(s)
BCG Vaccine/immunology , CpG Islands/immunology , DNA, Bacterial/immunology , Tuberculosis/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/pharmacology , Animals , Antineoplastic Agents/immunology , Base Sequence , CpG Islands/genetics , DNA, Bacterial/genetics , DNA, Bacterial/therapeutic use , Humans , Immunization , Tuberculosis/prevention & control , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...