Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Microbiol Immunol ; 61(6): 195-205, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28463465

ABSTRACT

To develop safe vaccines for inducing mucosal immunity to major pulmonary bacterial infections, appropriate vaccine antigens (Ags), delivery systems and nontoxic molecular adjuvants must be considered. Such vaccine constructs can induce Ag-specific immune responses that protect against mucosal infections. In particular, it has been shown that simply mixing the adjuvant with the bacterial Ag is a relatively easy means of constructing adjuvant-based mucosal vaccine preparations; the resulting vaccines can elicit protective immunity. DNA-based nasal adjuvants targeting mucosal DCs have been studied in order to induce Ag-specific mucosal and systemic immune responses that provide essential protection against microbial pathogens that invade mucosal surfaces. In this review, initially a plasmid encoding the cDNA of Flt3 ligand (pFL), a molecule that is a growth factor for DCs, as an effective adjuvant for mucosal immunity to pneumococcal infections, is introduced. Next, the potential of adding unmethylated CpG oligodeoxynucleotide and pFL together with a pneumococcal Ag to induce protection from pneumococcal infections is discussed. Pneumococcal surface protein A has been used as vaccine for restoring mucosal immunity in older persons. Further, our nasal pFL adjuvant system with phosphorylcholine-keyhole limpet hemocyanin (PC-KLH) has also been used in pneumococcal vaccine development to induce complete protection from nasal carriage by Streptococcus pneumoniae. Finally, the possibility that anti-PC antibodies induced by nasal delivery of pFL plus PC-KLH may play a protective role in prevention of atherogenesis and thus block subsequent development of cardiovascular disease is discussed.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Dendritic Cells/immunology , Immunity, Mucosal/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Vaccines, DNA/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , DNA, Complementary/immunology , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Phosphorylcholine/administration & dosage , Phosphorylcholine/immunology , Pneumococcal Vaccines/administration & dosage , Vaccines, DNA/administration & dosage
2.
Plant Cell Rep ; 35(3): 667-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26661780

ABSTRACT

KEY MESSAGE: The first Good Manufacturing Practices production of a purification-free rice-based oral cholera vaccine (MucoRice-CTB) from transgenic plants in a closed cultivation system yielded a product meeting regulatory requirements. Despite our knowledge of their advantages, plant-based vaccines remain unavailable for human use in both developing and industrialized countries. A leading, practical obstacle to their widespread use is producing plant-based vaccines that meet governmental regulatory requirements. Here, we report the first production according to current Good Manufacturing Practices of a rice-based vaccine, the cholera vaccine MucoRice-CTB, at an academic institution. To this end, we established specifications and methods for the master seed bank (MSB) of MucoRice-CTB, which was previously generated as a selection-marker-free line, evaluated its propagation, and given that the stored seeds must be renewed periodically. The production of MucoRice-CTB incorporated a closed hydroponic system for cultivating the transgenic plants, to minimize variations in expression and quality during vaccine manufacture. This type of molecular farming factory can be operated year-round, generating three harvests annually, and is cost- and production-effective. Rice was polished to a ratio of 95 % and then powdered to produce the MucoRice-CTB drug substance, and the identity, potency, and safety of the MucoRice-CTB product met pre-established release requirements. The formulation of MucoRice-CTB made by fine-powdering of drug substance and packaged in an aluminum pouch is being evaluated in a physician-initiated phase I study.


Subject(s)
Cholera Vaccines/genetics , Oryza/genetics , Plants, Genetically Modified/genetics , Technology, Pharmaceutical/methods , Administration, Oral , Animals , Blotting, Western , Cholera/immunology , Cholera/microbiology , Cholera/prevention & control , Cholera Toxin/toxicity , Cholera Vaccines/administration & dosage , Cholera Vaccines/immunology , Cost-Benefit Analysis , Diarrhea/chemically induced , Diarrhea/immunology , Diarrhea/prevention & control , Drug Packaging , Drug Stability , Humans , Immunization/methods , Mice , Oryza/growth & development , Plants, Genetically Modified/growth & development , Powders , Reproducibility of Results , Technology, Pharmaceutical/economics , Vibrio cholerae/immunology
3.
PLoS One ; 10(9): e0139368, 2015.
Article in English | MEDLINE | ID: mdl-26422280

ABSTRACT

Cholera toxin (CT) induces severe diarrhea in humans but acts as an adjuvant to enhance immune responses to vaccines when administered orally. Nasally administered CT also acts as an adjuvant, but CT and CT derivatives, including the B subunit of CT (CTB), are taken up from the olfactory epithelium and transported to the olfactory bulbs and therefore may be toxic to the central nervous system. To assess the toxicity, we investigated whether nasally administered CT or CT derivatives impair the olfactory system. In mice, nasal administration of CT, but not CTB or a non-toxic CT derivative, reduced the expression of olfactory marker protein (OMP) in the olfactory epithelium and olfactory bulbs and impaired odor responses, as determined with behavioral tests and optical imaging. Thus, nasally administered CT, like orally administered CT, is toxic and damages the olfactory system in mice. However, CTB and a non-toxic CT derivative, do not damage the olfactory system. The optical imaging we used here will be useful for assessing the safety of nasal vaccines and adjuvants during their development for human use and CT can be used as a positive control in this test.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Administration, Intranasal/adverse effects , Cholera Toxin/administration & dosage , Olfactory Mucosa/drug effects , Animals , Bacterial Vaccines/metabolism , Bacterial Vaccines/pharmacology , Cholera Toxin/immunology , Male , Mice, Inbred BALB C , Olfactory Marker Protein/metabolism , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism
4.
Immune Netw ; 15(1): 9-15, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25713504

ABSTRACT

Streptococcus pneumoniae (the pneumococcus) causes a major upper respiratory tract infection often leading to severe illness and death in the elderly. Thus, it is important to induce safe and effective mucosal immunity against this pathogen in order to prevent pnuemocaccal infection. However, this is a very difficult task to elicit protective mucosal IgA antibody responses in older individuals. A combind nasal adjuvant consisting of a plasmid encoding the Flt3 ligand cDNA (pFL) and CpG oligonucleotide (CpG ODN) successfully enhanced S. pneumoniae-specific mucosal immunity in aged mice. In particular, a pneumococcal surface protein A-based nasal vaccine given with pFL and CpG ODN induced complete protection from S. pneumoniae infection. These results show that nasal delivery of a combined DNA adjuvant offers an attractive potential for protection against the pneumococcus in the elderly.

5.
PLoS One ; 8(4): e60453, 2013.
Article in English | MEDLINE | ID: mdl-23565250

ABSTRACT

We assessed the role of CCR5(+)/CCR6(+)/CD11b(+)/CD11c(+) dendritic cells (DCs) for induction of ovalbumin (OVA)-specific antibody (Ab) responses following mucosal immunization. Mice given nasal OVA plus an adenovirus expressing Flt3 ligand (Ad-FL) showed early expansion of CCR5(+)/CCR6(+)/CD11b(+)/CD11c(+) DCs in nasopharyngeal-associated lymphoid tissue (NALT) and cervical lymph nodes (CLNs). Subsequently, this DC subset became resident in submandibular glands (SMGs) and nasal passages (NPs) in response to high levels of CCR-ligands produced in these tissues. CD11b(+)/CD11c(+) DCs were markedly decreased in both CCR5(-/-) and CCR6(-/-) mice. Chimera mice reconstituted with bone marrow cells from CD11c-diphtheria toxin receptor (CD11c-DTR) and CCR5(-/-) or CD11c-DTR and CCR6(-/-) mice given nasal OVA plus Ad-FL had elevated plasma IgG, but reduced IgA as well as low anti-OVA secretory IgA (SIgA )Ab responses in saliva and nasal washes. These results suggest that CCR5(+)CCR6(+) DCs play an important role in the induction of Ag-specific SIgA Ab responses.


Subject(s)
Dendritic Cells/metabolism , Immunoglobulin A/metabolism , Membrane Proteins/metabolism , Mucous Membrane/immunology , Ovalbumin/pharmacology , Receptors, CCR5/metabolism , Receptors, CCR6/metabolism , Adenoviridae , Administration, Intranasal , Animals , Membrane Proteins/genetics , Mice , Mucous Membrane/drug effects
6.
Biochem Biophys Res Commun ; 418(1): 6-11, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22200492

ABSTRACT

Our previous studies showed that an adenovirus (Ad) serotype 5 vector expressing Flt3 ligand (Ad-FL) as nasal adjuvant activates CD11c(+) dendritic cells (DCs) for the enhancement of antigen (Ag)-specific IgA antibody (Ab) responses. In this study, we examined the molecular mechanism for activation of CD11c(+) DCs and their roles in induction of Ag-specific Th1- and Th2-cell responses. Ad-FL activated CD11c(+) DCs expressed increased levels of the Notch ligand (L)-expression and specific mRNA. When CD11c(+) DCs from various mucosal and systemic lymphoid tissues of mice given nasal OVA plus Ad-FL were cultured with CD4(+) T cells isolated from non-immunized OVA TCR-transgenic (OT II) mice, significantly increased levels of T cell proliferative responses were noted. Furthermore, Ad-FL activated DCs induced IFN-γ, IL-2 and IL-4 producing CD4(+) T cells. Of importance, these APC functions by Ad-FL activated DCs were down-regulated by blocking Notch-Notch-L pathway. These results show that Ad-FL induces CD11c(+) DCs to the express Notch-ligands and these activated DCs regulate the induction of Ag-specific Th1- and Th2-type cytokine responses.


Subject(s)
Dendritic Cells/immunology , Receptors, Notch/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing , Animals , CD11c Antigen/immunology , Calcium-Binding Proteins/immunology , Cell Proliferation , Immunity, Active , Immunity, Mucosal , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Jagged-2 Protein , Ligands , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/immunology , Serrate-Jagged Proteins
7.
Vaccine ; 30(4): 803-12, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22100889

ABSTRACT

Since a combination of flt3 ligand plasmid (pFL) and CpG-oligodeoxynucleotides (ODN)(3) as a dendritic cell (DC)-targeting double mucosal adjuvant elicited ovalbumin-specific secretory IgA (S-IgA) antibody (Ab) responses, we examined whether this double adjuvant could induce influenza-specific protective immunity in aged mice. A double adjuvant plus A/Puerto Rico/8/34 (PR8) hemagglutinin (HA) induced increased numbers of CD11b(+) CD11c(+) DCs and both CD4(+) Th1- and Th2-type responses in the nasopharyngeal-associated lymphoreticular tissue, nasal passages and cervical lymph nodes. Further, increased levels of PR8 HA-specific S-IgA Ab responses were detected in the upper respiratory tact (URT) of aged and young adult mice given nasal PR8 HA with this double adjuvant. Thus, when mice were challenged with PR8 virus via the nasal route, both aged and young adult mice given nasal vaccine exhibited complete protection. Further, IgA-deficient mice nasally immunized with a double adjuvant influenza vaccine failed to provide protection against PR8 challenge. These results indicate that a nasal double adjuvant successfully induces PR8 HA-specific IgA Ab responses in both young adult and aged mice, which are essential for the prevention of influenza infection in the murine URT.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunity, Mucosal , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Membrane Proteins/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Dendritic Cells/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin A/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Th1 Cells/immunology , Th2 Cells/immunology
8.
PLoS One ; 6(9): e25396, 2011.
Article in English | MEDLINE | ID: mdl-21980444

ABSTRACT

Native cholera toxin (nCT) as a nasal adjuvant was shown to elicit increased levels of T-independent S-IgA antibody (Ab) responses through IL-5- IL-5 receptor interactions between CD4+ T cells and IgA+ B-1 B cells in murine submandibular glands (SMGs) and nasal passages (NPs). Here, we further investigate whether oral-nasopharyngeal dendritic cells (DCs) play a central role in the induction of B-1 B cell IgA class switch recombination (CSR) for the enhancement of T cell-independent (TI) mucosal S-IgA Ab responses. High expression levels of activation-induced cytidine deaminase, Iα-Cµ circulation transcripts and Iµ-Cα transcripts were seen on B-1 B cells purified from SMGs and NPs of both TCRß⁻/⁻ mice and wild-type mice given nasal trinitrophenyl (TNP)-LPS plus nCT, than in the same tissues of mice given nCT or TNP-LPS alone. Further, DCs from SMGs, NPs and NALT of mice given nasal TNP-LPS plus nCT expressed significantly higher levels of a proliferation-inducing ligand (APRIL) than those in mice given TNP-LPS or nCT alone, whereas the B-1 B cells in SMGs and NPs showed elevated levels of transmembrane activator and calcium modulator cyclophilin ligand interactor (TACI) expression. Interestingly, high frequencies of IgA+ B-1 B cells were induced when peritoneal IgA⁻ IgM+ B cells were stimulated with mucosal DCs from mice given nasal TNP-LPS plus nCT. Taken together, these findings show that nasal nCT plays a key role in the enhancement of mucosal DC-mediated TI IgA CSR by B-1 B cells through their interactions with APRIL and TACI.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Immunoglobulin A/immunology , Immunoglobulin Class Switching/immunology , Mouth Mucosa/immunology , Nasopharynx/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Cell Activation Factor Receptor/genetics , B-Cell Maturation Antigen/genetics , Cholera Toxin/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation/immunology , Immunoglobulin A/chemistry , Lipopolysaccharides/immunology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Submandibular Gland/immunology , T-Lymphocytes , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
9.
Adv Otorhinolaryngol ; 72: 25-7, 2011.
Article in English | MEDLINE | ID: mdl-21865682

ABSTRACT

Pneumococcal diseases such as otitis media, pneumonia, and meningitis are invariably preceded by nasopharyngeal colonization, and herd immunity against pneumococcal disease requires protection against colonization. An early study in mice demonstrated that mucosal immunization with cholera toxin B subunit as adjuvant could elicit solid mucosal immunity. Recent data from several laboratories provides support for three different mechanisms by which adaptive immunity can provide protection against colonization. (1) IL-17-dependent T cell immunity can recruit PMN to sites of colonization. This IL-17-dependent immunity can be elicited by immunization with antigen plus a mucosal adjuvant, or can be elicited by colonization itself. (2) Immunity against colonization can be mediated by mucosal IgA and at the mucosal surface passive mucosal IgA antibody provides much better protection against carriage than passive IgG antibody. (3) Complement-fixing IgG antibody can protect against colonization and may act by protecting against colonization of bacteria.


Subject(s)
Bacterial Proteins/immunology , Immunity, Mucosal/immunology , Pneumococcal Infections/immunology , Respiratory Mucosa/immunology , Respiratory Tract Infections/immunology , Streptococcus pneumoniae/metabolism , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/metabolism , Colony Count, Microbial , Humans , Pneumococcal Infections/microbiology , Respiratory Mucosa/microbiology , Respiratory Tract Infections/microbiology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/isolation & purification
10.
Infect Immun ; 79(7): 2819-28, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21536790

ABSTRACT

We have previously shown that a pneumococcal surface protein A (PspA)-based vaccine containing DNA plasmid encoding the Flt3 ligand (FL) gene (pFL) as a nasal adjuvant prevented nasal carriage of Streptococcus pneumoniae. In this study, we further investigated the safety and efficacy of this nasal vaccine for the induction of PspA-specific antibody (Ab) responses against lung infection with S. pneumoniae. C57BL/6 mice were nasally immunized with recombinant PspA/Rx1 (rPspA) plus pFL three times at weekly intervals. When dynamic translocation of pFL was initially examined, nasal pFL was taken up by nasal dendritic cells (DCs) and epithelial cells (nECs) but not in the central nervous systems, including olfactory nerve and epithelium. Of importance, nasal pFL induced FL protein synthesis with minimum levels of inflammatory cytokines in the nasal washes (NWs) and bronchoalveolar lavage fluid (BALF). NWs and BALF as well as plasma of mice given nasal rPspA plus pFL contained increased levels of rPspA-specific secretory IgA and IgG Ab responses that were correlated with elevated numbers of CD8(+) and CD11b(+) DCs and interleukin 2 (IL-2)- and IL-4-producing CD4(+) T cells in the nasal mucosa-associated lymphoid tissues (NALT) and cervical lymph nodes (CLNs). The in vivo protection by rPspA-specific Abs was evident in markedly reduced numbers of CFU in the lungs, airway secretions, and blood when mice were nasally challenged with Streptococcus pneumoniae WU2. Our findings show that nasal pFL is a safe and effective mucosal adjuvant for the enhancement of bacterial antigen (Ag) (rPspA)-specific protective immunity through DC-induced Th2-type and IL-2 cytokine responses.


Subject(s)
Adjuvants, Immunologic , Antibodies, Bacterial/biosynthesis , Bacterial Proteins/immunology , Dendritic Cells/immunology , Membrane Proteins/immunology , Nasal Mucosa/immunology , Pneumococcal Vaccines/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Bronchoalveolar Lavage Fluid/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/analysis , Epithelial Cells/metabolism , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nasal Cavity/immunology , Nasal Sprays , Plasmids , Pneumococcal Vaccines/administration & dosage , Pneumonia, Pneumococcal/prevention & control , Recombinant Proteins , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
11.
J Immunol ; 186(4): 2454-61, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242514

ABSTRACT

Our previous study showed that a combination of a plasmid-expressing Flt3 ligand (pFL) and CpG oligodeoxynucleotides (CpG ODN) as a combined nasal adjuvant elicited mucosal immune responses in aged (2-y-old) mice. In this study, we investigated whether a combination of pFL and CpG ODN as a nasal adjuvant for a pneumococcal surface protein A (PspA) would enhance PspA-specific secretory-IgA Ab responses, which could provide protective mucosal immunity against Streptococcus pneumoniae infection in aged mice. Nasal immunization with PspA plus a combination of pFL and CpG ODN elicited elevated levels of PspA-specific secretory-IgA Ab responses in external secretions and plasma in both young adult and aged mice. Significant levels of PspA-specific CD4(+) T cell proliferative and PspA-induced Th1- and Th2- type cytokine responses were noted in nasopharyngeal-associated lymphoreticular tissue, cervical lymph nodes, and spleen of aged mice, which were equivalent to those in young adult mice. Additionally, increased numbers of mature-type CD8, CD11b-expressing dendritic cells were detected in mucosal inductive and effector lymphoid tissues of aged mice. Importantly, aged mice given PspA plus a combination of pFL and CpG ODN showed protective immunity against nasal S. pneumoniae colonization. These results demonstrate that nasal delivery of a combined DNA adjuvant offers an attractive possibility for protection against S. pneumoniae in the elderly.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aging/immunology , DNA, Complementary/administration & dosage , Immunoglobulin A, Secretory/biosynthesis , Membrane Proteins/genetics , Nasal Mucosa/immunology , Oligodeoxyribonucleotides/administration & dosage , Pneumococcal Infections/immunology , Adjuvants, Immunologic/blood , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Cells, Cultured , CpG Islands/immunology , DNA, Complementary/blood , DNA, Complementary/immunology , Drug Combinations , Humans , Immunoglobulin A, Secretory/physiology , Membrane Proteins/administration & dosage , Membrane Proteins/blood , Mice , Nasal Mucosa/metabolism , Nasal Mucosa/microbiology , Oligodeoxyribonucleotides/blood , Oligodeoxyribonucleotides/immunology , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Streptococcus pneumoniae/immunology
12.
J Immunol ; 185(3): 1755-62, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20585031

ABSTRACT

This study was designed to investigate whether secretory-IgA (S-IgA) Abs induced by a pneumococcal surface protein A (PspA)-based nasal vaccine are necessary for prevention of streptococcal colonization. Mice nasally immunized with PspA plus a plasmid expressing Flt3 ligand (pFL) cDNA as a mucosal adjuvant showed significantly higher levels of PspA-specific S-IgA and IgG Ab responses in both plasma and nasal washes when compared with naive mice. Although IgA(-/-) mice given nasal PspA plus pFL had significantly high levels of PspA-specific IgG Abs, high numbers of CFUs were detected in nasal washes and nasal passages. In contrast, vaccinated wild-type mice showed essentially no bacteria in the nasal cavity. Further, a nasal vaccine consisting of PspA plus pFL effectively reduced pre-existing Streptococcus pneumoniae in the nasal cavity. These results show that PspA-based vaccine-induced specific S-IgA Abs play a necessary role in the regulation of S. pneumoniae colonization in the nasal cavity.


Subject(s)
Antibodies, Bacterial/physiology , Bacterial Proteins/immunology , Immunity, Innate , Immunoglobulin A, Secretory/physiology , Streptococcal Infections/prevention & control , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Cells, Cultured , Colony Count, Microbial , Female , Immunity, Innate/genetics , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin A, Secretory/genetics , Membrane Proteins/administration & dosage , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Streptococcal Infections/immunology , Streptococcal Infections/microbiology
13.
Proc Natl Acad Sci U S A ; 107(16): 7419-24, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20360558

ABSTRACT

The indigenous bacteria create natural cohabitation niches together with mucosal Abs in the gastrointestinal (GI) tract. Here we report that opportunistic bacteria, largely Alcaligenes species, specifically inhabit host Peyer's patches (PPs) and isolated lymphoid follicles, with the associated preferential induction of antigen-specific mucosal IgA Abs in the GI tract. Alcaligenes were identified as the dominant bacteria on the interior of PPs from naïve, specific-pathogen-free but not from germ-free mice. Oral transfer of intratissue uncultured Alcaligenes into germ-free mice resulted in the presence of Alcaligenes inside the PPs of recipients. This result was further supported by the induction of antigen-specific Ab-producing cells in the mucosal (e.g., PPs) but not systemic compartment (e.g., spleen). The preferential presence of Alcaligenes inside PPs and the associated induction of intestinal secretory IgA Abs were also observed in both monkeys and humans. Localized mucosal Ab-mediated symbiotic immune responses were supported by Alcaligenes-stimulated CD11c(+) dendritic cells (DCs) producing the Ab-enhancing cytokines TGF-beta, B-cell-activating factor belonging to the TNF family, and IL-6 in PPs. These CD11c(+) DCs did not migrate beyond the draining mesenteric lymph nodes. In the absence of antigen-specific mucosal Abs, the presence of Alcaligenes in PPs was greatly diminished. Thus, indigenous opportunistic bacteria uniquely inhabit PPs, leading to PP-DCs-initiated, local antigen-specific Ab production; this may involve the creation of an optimal symbiotic environment on the interior of the PPs.


Subject(s)
Antibodies/chemistry , Bacteria/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Peyer's Patches/immunology , Animals , Humans , In Situ Hybridization, Fluorescence , Lymph Nodes/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , RNA, Ribosomal, 16S/metabolism , Spleen/immunology
14.
J Immunol ; 180(12): 8126-34, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523277

ABSTRACT

Previously, we showed that nasal administration of a naked cDNA plasmid expressing Flt3 ligand (FL) cDNA (pFL) enhanced CD4(+) Th2-type, cytokine-mediated mucosal immunity and increased lymphoid-type dendritic cell (DC) numbers. In this study, we investigated whether targeting nasopharyngeal-associated lymphoreticular tissue (NALT) DCs by a different delivery mode of FL, i.e., an adenovirus (Ad) serotype 5 vector expressing FL (Ad-FL), would provide Ag-specific humoral and cell-mediated mucosal immunity. Nasal immunization of mice with OVA plus Ad-FL as mucosal adjuvant elicited high levels of OVA-specific Ab responses in external secretions and plasma as well as significant levels of OVA-specific CD4(+) T cell proliferative responses and OVA-induced IFN-gamma and IL-4 production in NALT, cervical lymph nodes, and spleen. We also observed higher levels of OVA-specific CTL responses in the spleen and cervical lymph nodes of mice given nasal OVA plus Ad-FL than in mice receiving OVA plus control Ad. Notably, the number of CD11b(+)CD11c(+) DCs expressing high levels of costimulatory molecules was preferentially increased. These DCs migrated from the NALT to mucosal effector lymphoid tissues. Taken together, these results suggest that the use of Ad-FL as a nasal adjuvant preferentially induces mature-type NALT CD11b(+)CD11c(+) DCs that migrate to effector sites for subsequent CD4(+) Th1- and Th2-type cytokine-mediated, Ag-specific Ab and CTL responses.


Subject(s)
Adenoviridae/genetics , Cell Movement/immunology , Dendritic Cells/immunology , Lymphoid Tissue/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mononuclear Phagocyte System/immunology , Nasopharynx/immunology , Adjuvants, Immunologic/genetics , Administration, Intranasal , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Genetic Vectors/administration & dosage , Humans , Immunity, Mucosal/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/virology , Membrane Proteins/administration & dosage , Mice , Mice, Inbred C57BL , Mononuclear Phagocyte System/cytology , Mononuclear Phagocyte System/virology , Nasopharynx/cytology , Nasopharynx/virology , fms-Like Tyrosine Kinase 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...