Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Immunity ; 44(4): 889-900, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27084119

ABSTRACT

Metagenomic studies show that diverse resident viruses inhabit the healthy gut; however, little is known about the role of these viruses in the maintenance of gut homeostasis. We found that mice treated with antiviral cocktail displayed more severe dextran sulfate sodium (DSS)-induced colitis compared with untreated mice. DSS-induced colitis was associated with altered enteric viral abundance and composition. When wild-type mice were reconstituted with Toll-like receptor 3 (TLR3) or TLR7 agonists or inactivated rotavirus, colitis symptoms were significantly ameliorated. Mice deficient in both TLR3 and TLR7 were more susceptible to DSS-induced experimental colitis. In humans, combined TLR3 and TLR7 genetic variations significantly influenced the severity of ulcerative colitis. Plasmacytoid dendritic cells isolated from inflamed mouse colon produced interferon-ß in a TLR3 and TLR7-dependent manner. These results imply that recognition of resident viruses by TLR3 and TLR7 is required for protective immunity during gut inflammation.


Subject(s)
Colitis/immunology , Gastrointestinal Tract/virology , Interferon-beta/immunology , Membrane Glycoproteins/immunology , Rotavirus/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/immunology , Animals , Antiviral Agents/pharmacology , Colitis/chemically induced , Dendritic Cells/immunology , Dextran Sulfate , Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Humans , Inflammation/immunology , Interferon-beta/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 7/genetics
2.
Immunity ; 38(1): 153-65, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23246312

ABSTRACT

Circulatory antigens transit through the small intestine via the fenestrated capillaries in the lamina propria prior to entering into the draining lymphatics. But whether or how this process controls mucosal immune responses remains unknown. Here we demonstrate that dendritic cells (DCs) of the lamina propria can sample and process both circulatory and luminal antigens. Surprisingly, antigen cross-presentation by resident CX3CR1(+) DCs induced differentiation of precursor cells into CD8(+) T cells that expressed interleukin-10 (IL-10), IL-13, and IL-9 and could migrate into adjacent compartments. We conclude that lamina propria CX3CR1(+) DCs facilitate the surveillance of circulatory antigens and act as a conduit for the processing of self- and intestinally absorbed antigens, leading to the induction of CD8(+) T cells, that partake in the control of T cell activation during mucosal immune responses.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Cross-Priming/immunology , Dendritic Cells/metabolism , Enteritis/immunology , Enteritis/prevention & control , Epitopes, T-Lymphocyte/immunology , Intestinal Mucosa/cytology , Intestine, Small/immunology , Mice , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism
3.
Trends Immunol ; 39(9): 677-696, 2018 09.
Article in English | MEDLINE | ID: mdl-29716793

ABSTRACT

The gastrointestinal (GI) tract represents a unique challenge to the mammalian immune system. It must tolerate the presence of the luminal microbiota and thus not respond to their products, but still protect the intestinal mucosa from potentially harmful dietary antigens and invading pathogens. The intestinal epithelium, composed of a single layer of cells, is crucial for preserving gut homeostasis and acts both as a physical barrier and as a coordinating hub for immune defense and crosstalk between bacteria and immune cells. We highlight here recent findings regarding communication between microbes and intestinal epithelial cells (IECs), as well as the immune mechanisms employed by distinct IEC subsets to promote homeostasis, emphasizing the central and active role that these cells play in host enteric defense.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Antigens/immunology , Cell Communication , Dysbiosis , Epithelial Cells/metabolism , Gastrointestinal Microbiome/immunology , Homeostasis , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity, Innate , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Signal Transduction
4.
J Immunol ; 201(4): 1287-1294, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29987162

ABSTRACT

Secretory IgA is a key host defense mechanism that controls the intestinal microbiota. We investigated the role of CD11c+CX3CR1+CD64+ macrophages in IgA production in the intestine. Intestinal CX3CR1+ macrophages directly induced IgA secretion by B cells. Ag delivery to lamina propria (LP) CX3CR1+ macrophages specifically induced intestinal IgA production. The induction of IgA by CX3CR1+ macrophages required BAFF, a proliferation-inducing ligand, and TNF-α, but was surprisingly independent of TLR-mediated microbial recognition and retinoic acid signaling. IgA secretion by CX3CR1+ macrophages was enhanced by LP CD8+ T cells through the secretion of IL-9 and IL-13. CX3CR1+ macrophages and CD8+ T cells induced IgA production by B cells independently of mesenteric lymph nodes and Peyer patches. Our data reveal a previously unrecognized cellular circuitry in which LP CX3CR1+ macrophages, B cells, and CD8+ T cells coordinate the protective Ig secretion in the small intestine upon peripheral Ag delivery.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunoglobulin A, Secretory/biosynthesis , Intestinal Mucosa/immunology , Macrophages/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes/immunology , CX3C Chemokine Receptor 1/immunology , Immunity, Mucosal/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Int J Mol Sci ; 20(11)2019 May 28.
Article in English | MEDLINE | ID: mdl-31141897

ABSTRACT

The skin is an important physiological barrier against external stimuli, such as ultraviolet radiation (UV), xenobiotics, and bacteria. Dermal inflammatory reactions are associated with various skin disorders, including chemical-induced irritation and atopic dermatitis. Modulation of skin inflammatory response is a therapeutic strategy for skin diseases. Here, we synthesized chrysin-derivatives and identified the most potent derivative of Compound 6 (CPD 6). We evaluated its anti-inflammatory effects in vitro cells of macrophages and keratinocytes, and in vivo dermatitis mouse models. In murine macrophages stimulated by lipopolysaccharide (LPS), CPD 6 significantly attenuated the release of inflammatory mediators such as nitric oxide (NO) (IC50 for NO inhibition: 3.613 µM) and other cytokines. In cultured human keratinocytes, CPD 6 significantly attenuated the release of inflammatory cytokines induced by the combination of IFN-γ and TNF-α, UV irradiation, or chemical irritant stimulation. CPD 6 inhibited NFκB and JAK2/STAT1 signaling pathways, and activated Nrf2/HO-1 signaling. In vivo relevancy of anti-inflammatory effects of CPD 6 was observed in acute and chronic skin inflammation models in mice. CPD 6 showed significant anti-inflammatory properties both in vitro cells and in vivo dermatitis animal models, mediated by the inhibition of the NFκB and JAK2-STAT1 pathways and activation of Nrf2/HO-1 signaling. We propose that the novel chrysin-derivative CPD 6 may be a potential therapeutic agent for skin inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dermatitis/drug therapy , Dermatologic Agents/pharmacology , Flavonoids/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Dermatologic Agents/chemistry , Dermatologic Agents/therapeutic use , Heme Oxygenase-1/metabolism , Humans , Janus Kinase 2/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , RAW 264.7 Cells , STAT1 Transcription Factor/metabolism
6.
Clin Immunol ; 190: 1-10, 2018 05.
Article in English | MEDLINE | ID: mdl-29432811

ABSTRACT

Allergic conjunctivitis is mediated by eosinophilic infiltration and Th2 type immune responses. This study aims to elucidate the role of rapamycin, mTOR inhibitor, on OVA-induced experimental allergic conjunctivitis (EAC). Rapamycin administration intraperitoneally markedly reduced clinical signs, total and OVA-specific IgE and IgG1/G2a ratio in serum, and conjunctival eosinophilic infiltration. Infiltrations of CD11c+ dendritic cells and CD4+ T cells, and the expressions of chemokines and adhesion molecules in the conjunctiva were attenuated in rapamycin-treated mice, as well as decreased Th1 and Th2 cytokines in the cervical lymph nodes compared to non-treated mice. The expression of mTOR signaling proteins was increased in EAC and reduced by rapamycin treatment. Topical application of rapamycin was also proved to show reduced clinical signs, eosinophil infiltration, and Th2 type immune responses comparable to those from intraperitoneal injection of rapamycin. These findings suggest the therapeutic implications of rapamycin in the attenuation of allergic conjunctivitis.


Subject(s)
Conjunctivitis, Allergic/immunology , Conjunctivitis, Allergic/prevention & control , Sirolimus/pharmacology , Th2 Cells/immunology , Animals , Conjunctiva/drug effects , Conjunctiva/immunology , Conjunctiva/metabolism , Conjunctivitis, Allergic/genetics , Female , Gene Expression/drug effects , Gene Expression/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunosuppressive Agents/pharmacology , Mice, Inbred BALB C , Ovalbumin/immunology
7.
J Nat Prod ; 81(12): 2647-2653, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30540183

ABSTRACT

A concise synthesis of sericetin (1) was performed in four steps from readily available 3- O-benzylgalangin (4), featuring electrocyclization to produce the tricyclic core and a sequential aromatic Claisen/Cope rearrangement to incorporate the 8-prenyl group of 1. In addition, the therapeutic potential of sericetin (1), isosericetin (2), and three prenylated tetracyclic synthetic intermediates (11, 12, and 14) against cisplatin-induced nephrotoxicity using renal tubular cells were evaluated. Compound 14 showed therapeutic potential against cisplatin-induced kidney damage.


Subject(s)
Acute Kidney Injury/drug therapy , Pyrans/pharmacology , Acute Kidney Injury/chemically induced , Animals , Cell Line , Cisplatin , Fabaceae/chemistry , Molecular Structure , Protective Agents/chemical synthesis , Protective Agents/therapeutic use , Pyrans/therapeutic use , Rats , Rats, Sprague-Dawley
9.
Toxicol Appl Pharmacol ; 307: 62-71, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27450019

ABSTRACT

Impaired immune responses in skin play a pivotal role in the development and progression of chemical-associated inflammatory skin disorders. In this study, we synthesized new flavonoid derivatives from macakurzin C, and identified in vitro and in vivo efficacy of a potent anti-inflammatory flavonoid, Compound 14 (CPD 14), with its underlying mechanisms. In lipopolysaccharide (LPS)-stimulated murine macrophages and IFN-γ/TNF-α-stimulated human keratinocytes, CPD 14 significantly inhibited the release of inflammatory mediators including nitric oxide (NO), prostaglandins, and cytokines (IC50 for NO inhibition in macrophages: 4.61µM). Attenuated NF-κB signaling and activated Nrf2/HO-1 pathway were responsible for the anti-inflammatory effects of CPD 14. The in vivo relevance was examined in phorbol 12-myristate 13-acetate (TPA)-induced acute skin inflammation and oxazolone-induced atopic dermatitis models. Topically applied CPD 14 significantly protected both irritation- and sensitization-associated skin inflammation by suppressing the expression of inflammatory mediators. In summary, we demonstrated that a newly synthesized flavonoid, CPD 14, has potent inhibitory effects on skin inflammation, suggesting it is a potential therapeutic candidate to treat skin disorders associated with excessive inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Flavonoids , Acute Disease , Animals , Cell Line , Chronic Disease , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Dinoprostone/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonoids/therapeutic use , Heme Oxygenase-1/metabolism , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Oxazolone , RAW 264.7 Cells , Tetradecanoylphorbol Acetate , Tumor Necrosis Factor-alpha/pharmacology
10.
Clin Immunol ; 161(2): 110-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26307432

ABSTRACT

Allergic conjunctivitis from an allergen-driven Th2 response is characterized by conjunctival eosinophilic infiltration. Although CCL20-CCR6 axis has been reported to play a proinflammatory role in several murine models of autoimmune diseases including allergic diseases, their underlying mechanism needs to be investigated. We here examined whether CCL20-CCR6 axis could play a role in the development of allergic conjunctival inflammation using murine experimental allergic conjunctivitis (EAC) model induced by ovalbumin (OVA) allergen. Mice were challenged with consecutive 10days of OVA via conjunctival sac after systemic challenge with OVA and cholera toxin in alum. Several indicators for allergy were comparatively evaluated in wild-type and CCR6 KO EAC mice. Wild-type mice challenged with OVA via conjunctival sac following systemic challenge with OVA in alum had severe allergic conjunctivitis. The absence of CCR6 suppressed IgE secretion and allergic conjunctival inflammation. Reduced allergic inflammation was ascribable to reduced cytokine responses from Th-2 type in draining lymph node although Th17, regulatory T cells and dendritic cell subsets are not affected by the absence of CCR6. In addition, neutralization of CCR6 ligand, CCL20 could repress allergic conjunctival inflammation. Our findings suggested that CCR6 might be crucial for optimal development of Th2 immune responses and further allergic conjunctival inflammation in EAC model.


Subject(s)
Allergens/immunology , Chemokines, CC/immunology , Conjunctivitis, Allergic/immunology , Receptors, CCR6/immunology , Th2 Cells/immunology , Alum Compounds/pharmacology , Animals , Chemokine CCL20/immunology , Cholera Toxin/pharmacology , Cytokines/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Immunoglobulin E/immunology , Inflammation/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
11.
J Infect Dis ; 209(2): 203-15, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23904297

ABSTRACT

In human and nonhuman primates, Shigella spp. cause bacillary dysentery by invading colon epithelium and promoting a strong inflammatory response; however, adult mice are resistant to oral Shigella infection. In this study, intraperitoneal challenge with virulent S. flexneri 2a (YSH6000) resulted in diarrhea and severe body weight loss in adult B6 mice. Of note, virulent S. flexneri 2a could invade and colonize not only systemic tissues but also the serosa and lamina propria region of the large intestine. In addition, epithelial shedding, barrier integrity, and goblet cell hyperplasia were found in the large intestine by 24 hours post-intraperitoneal Shigella infection. Of note, predominant expression of proinflammatory cytokines and chemokines were found in the large intestine after intraperitoneal challenge. Monocytes played a critical role in attenuating diarrhea and in providing protective efficacy against intraperitoneal Shigella infection. Most importantly, mice prevaccinated with attenuated S. flexneri 2a (SC602) strain were protected against intraperitoneal challenge with YSH6000. When taken together, these findings show that intraperitoneal challenge with virulent S. flexneri 2a can provoke bacillary dysentery and severe pathogenesis in adult mice. This model may be helpful for understanding the induction mechanism of bacillary dysentery and for evaluating Shigella vaccine candidates.


Subject(s)
Dysentery, Bacillary/pathology , Shigella flexneri/growth & development , Animal Structures/microbiology , Animal Structures/pathology , Animals , Body Weight , Cytokines/analysis , Diarrhea/microbiology , Diarrhea/pathology , Disease Models, Animal , Dysentery, Bacillary/microbiology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Analysis
12.
Immunol Rev ; 234(1): 233-46, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20193022

ABSTRACT

Dendritic cells (DCs) are key regulators of the immune system. They act as professional antigen-presenting cells and are capable of activating naive T cells and stimulating the growth and differentiation of B cells. According to their molecular expression, DCs can be divided into several subsets with different functions. We focus on DC subsets expressing langerin, a C-type lectin. Langerin expression is predominant in skin DCs, but langerin-expressing DCs also exist in mucosal tissue and can be induced by immunization and sometimes by nutrient deficiency. Topical transcutaneous immunization induces langerin(+)CD8 alpha(-) DCs in mesenteric lymph nodes (MLNs), which mediate the production of antigen-specific immunoglobulin A antibody in the intestine. Yet, in one recent study, langerin(+) DCs were generated in gut-associated lymphoid tissue and contributed to the suppressive intestinal immune environment in the absence of retinoic acid. In this review, we focus on the phenotypic and functional characteristics of langerin(+) DCs in the mucosal tissues, especially MLNs.


Subject(s)
Antigens, CD/immunology , Antigens, Surface/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Lymph Nodes/immunology , Mannose-Binding Lectins/immunology , Peyer's Patches/immunology , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , Cholecalciferol/immunology , Humans , Immunoglobulin A/biosynthesis , Langerhans Cells/immunology , Lymph Nodes/cytology , Mice , Peyer's Patches/cytology , Phenotype , Signal Transduction , Tretinoin/immunology
13.
Int J Cancer ; 132(12): 2839-48, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23184679

ABSTRACT

Myd88 is an important adaptor molecule for the activation of NADPH oxidase and arginase-1, which are responsible for the suppressive function of myeloid-derived suppressor cells (MDSCs). When wild-type and Myd88(-/-) mice were subcutaneously injected with CT26 colon cancer cells expressing human Her-2/neu, tumor growth was retarded in Myd88(-/-) mice than in wild-type mice. Although the generation of CD11b(+) Gr-1(+) MDSCs was less in Myd88(-/-) mice than in wild-type mice, Myd88(-/-) mice having tumor masses still had significant quantities of MDSCs, suggesting that MDSC generation might be independent of Myd88 signaling. However, MDSCs obtained from tumor-bearing Myd88(-/-) mice failed to suppress antigen-specific proliferation of CD8(+) T cells and CD4(+) T cells, whereas MDSCs from wild-type mice significantly suppressed both types of T cells. Consistent with this, we found that the levels of costimulatory molecules and MHC class II were significantly increased in MDSCs obtained from Myd88(-/-) mice compared with wild-type mice after tumor challenge. Furthermore, CD4(+) T cells residing in tumor-draining lymph nodes of Myd88(-/-) mice secreted more TNF-α than those of wild-type mice. Finally, the blockade of Myd88 signaling by treatment with Myd88 inhibitory peptide, during later tumor stages, significantly inhibited the growth of immunogenic tumors. Overall, these data suggest that signaling through the Myd88 adaptor molecule is critical for the direct suppressive function of MDSCs and approaches to block Myd88-mediated signaling in MDSCs might be effective to inhibit the immunosuppressive function of MDSCs.


Subject(s)
Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Humans , Immunosuppression Therapy , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism
14.
Eur J Immunol ; 42(3): 618-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22105301

ABSTRACT

The role of TLR signaling in linking the innate and adaptive immune systems has been a controversial issue that remains to be solved. Here, we determined whether MyD88-dependent TLR signals are required for the generation of B-cell responses during chronic Salmonella infection. Oral administration of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine (RASV) strain in MyD88(-/-) mice resulted in chronic infection. Infection was accompanied by enlarged germinal centers and hypergammaglobulinemia with anti-double-stranded DNA (dsDNA)-specific Ab in sera, and the deposition of immune complexes in the kidneys, suggesting onset of autoimmunity. CD4(+) T cells expressing PD-1, CXCR5, ICOS, and IL-21 were dramatically increased in chronically infected mice, indicating the expansion of follicular helper T (Tfh)-like cells. Of note, the depletion of CD4(+) T cells completely blocked the generation of polyclonal IgG Ab in sera after oral RASV challenge. Inflammatory myeloid cells expressing CD11b and Gr-1 accumulated in high numbers in the spleen of MyD88(-/-) mice. Interestingly, the blockade of PD-1 or ICOS significantly reduced the hypergammaglobulinemia and dsDNA-specific autoantibody production. Overall, these results suggest that Tfh-like cells in chronic bacterial infection trigger autoimmune hypergammaglobulinemia in a PD-1- and ICOS-dependent manner.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Hypergammaglobulinemia/microbiology , Myeloid Differentiation Factor 88/deficiency , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Hypergammaglobulinemia/immunology , Immunoglobulin G/blood , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukins/economics , Interleukins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Programmed Cell Death 1 Receptor , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Salmonella Infections/microbiology , Specific Pathogen-Free Organisms
15.
J Immunol ; 187(6): 3044-52, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21832166

ABSTRACT

Previous studies demonstrated cross talk between mucosal and reproductive organs during secretory IgA Ab induction. In this study, we aimed to clarify the underlying mechanisms of this cross talk. We found significantly higher titers of Ag-specific secretory IgA Ab in the vaginal wash after mucosal vaccination by both the intranasal (i.n.) and the intravaginal routes but not by the s.c. route. Interestingly, Ag-specific IgA Ab-secreting cells (ASCs) were found mainly in the uterus but not in the cervix and vaginal canal after i.n. vaccination. The fact that most Ag-specific IgA ASCs isolated from the uteri of vaccinated mice migrated toward mucosa-associated epithelial chemokine (MEC)/CCL28 suggests dominant expression of CCR10 on the IgA ASCs. Further, IgA ASCs in the uteri of vaccinated mice were reduced drastically in mice treated with neutralizing anti-MEC/CCL28 Ab. Most intriguingly, the female sex hormone estrogen directly regulated MEC/CCL28 expression and was augmented by i.n. vaccination with cholera toxin or stimulators for innate immunity. Further, blockage of estrogen function in the uterus by oral administration of the estrogen antagonist raloxifene significantly inhibited migration of Ag-specific IgA ASCs after i.n. vaccination with OVA plus cholera toxin. Taken together, these data strongly suggest that CCR10(+) IgA ASCs induced by mucosal vaccination via the i.n. route migrate into the uterus in a MEC/CCL28-dependent manner and that estrogen might have a crucial role in the protection against genital infection by regulating MEC/CCL28 expression in the uterus.


Subject(s)
Chemokines, CC/biosynthesis , Estrogens/immunology , Immunity, Mucosal/immunology , Plasma Cells/immunology , Receptors, CCR10/metabolism , Uterus/immunology , Administration, Intranasal , Animals , BALB 3T3 Cells , Blotting, Western , Chemokines, CC/immunology , Chemotaxis, Leukocyte/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Enzyme-Linked Immunosorbent Assay , Estrogens/metabolism , Female , Immunoglobulin A, Secretory/immunology , Immunoglobulin A, Secretory/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Plasma Cells/metabolism , Receptors, CCR10/immunology , Reverse Transcriptase Polymerase Chain Reaction , Uterus/metabolism , Vaccination
16.
J Immunol ; 186(12): 6999-7005, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21572021

ABSTRACT

Although the induction mechanism of secretory IgA has been well studied, that of IgG in the mucosal compartments is not well understood. In this study, vitamin A deficiency was convincingly shown to be associated with increased IgG in serum and intestinal fluid. We found increased numbers of IgG-secreting B cells in the lamina propria of the small intestine and mesenteric lymph node (MLN) of vitamin A-deficient (VAD) mice. Of note, IFN-γ secreted by MLN dendritic cells (DCs) was significantly augmented in VAD mice, unlike control mice, and CD103(+) DCs were the main subsets to secrete IFN-γ. The aberrant increase of IgG in VAD mice can be ascribable to IFN-γ, because IFN-γ(-/-) VAD mice have normal IgG levels and the addition of rIFN-γ increased IgG production by B cells cocultured with MLN DCs from IFN-γ(-/-) VAD mice. Oral feeding of antibiotics resulted in significant reduction of IgG in VAD mice, indicating a critical role for altered commensal bacteria for IgG class-switching recombination in the absence of vitamin A. Collectively, vitamin A deficiency provokes the generation of IFN-γ-secreting CD103(+) DCs, which may be a critical regulator for IgG generation in the MLN.


Subject(s)
Dendritic Cells/immunology , Immunoglobulin G/biosynthesis , Interferon-gamma/immunology , Lymph Nodes/immunology , Vitamin A Deficiency/immunology , Animals , Antigens, CD/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Integrin alpha Chains/immunology , Interferon-gamma/metabolism , Intestinal Mucosa/immunology , Mesentery/cytology , Mesentery/immunology , Mice
17.
Vaccine ; 41(13): 2243-2252, 2023 03 24.
Article in English | MEDLINE | ID: mdl-36863926

ABSTRACT

Mucosal surfaces are in contact with the external environment and protect the body from infection by various microbes. To prevent infectious diseases at the first line of defense, the establishment of pathogen-specific mucosal immunity by mucosal vaccine delivery is needed. Curdlan, a 1,3-ß-glucan has a strong immunostimulatory effect when delivered as a vaccine adjuvant. Here, we investigated whether intranasal administration of curdlan and antigen (Ag) could induce sufficient mucosal immune responses and protect against viral infections. Intranasal co-administration of curdlan and OVA increased OVA-specific IgG and IgA Abs in both serum and mucosal secretions. In addition, intranasal co-administration of curdlan and OVA induced the differentiation of OVA-specific Th1/Th17 cells in the draining lymph nodes. To investigate the protective immunity of curdlan against viral infection, intranasal co-administration of curdlan with recombinant VP1 of EV71 C4a was administered and showed enhanced protection against enterovirus 71 in a passive serum transfer model using neonatal hSCARB2 mice, although intranasal administration of VP1 plus curdlan increased VP1-specific helper T cells responses but not mucosal IgA. Next, Mongolian gerbils were intranasally immunized with curdlan plus VP1, and they had effective protection against EV71 C4a infection, while decreasing viral infection and tissue damage by inducing Th17 responses. These results indicated that intranasal curdlan with Ag improved Ag-specific protective immunity by enhancing mucosal IgA and Th17 against viral infection. Our results suggest that curdlan is an advantageous candidate as a mucosal adjuvant and delivery vehicle for the development of mucosal vaccines.


Subject(s)
Enterovirus A, Human , Enterovirus , Mice , Animals , Administration, Intranasal , Th17 Cells , Immunization , Adjuvants, Immunologic , Immunity, Mucosal , Antigens , Immunoglobulin A , Mice, Inbred BALB C
18.
Vaccine ; 41(41): 6055-6063, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37648607

ABSTRACT

Hand, foot, and mouth disease (HFMD) is a highly contagious viral infection that is mainly caused by enterovirus 71 (EV71) and coxsackievirus 16 (CVA16). As there are no specific therapeutics for HFMD, the development of a bivalent vaccine is required to cover a broad range of infections. In this study, the effectiveness of novel monovalent and bivalent vaccines targeting EV71 C4a and CVA16 was investigated for their ability to prevent viral infections in neonatal human scavenger receptor class B member 2 (hSCARB2) transgenic mice. As hSCARB2 serves as a key viral receptor for EV71, these transgenic mice are susceptible to EV71 strains and facilitate viral binding, internalization, and uncoating processes. Antisera prepared by vaccine immunization were transferred to 2-day-old hSCARB2 transgenic mice, which were then infected with EV71 C4a or CVA16 virus. The antisera generated by each monovalent or bivalent vaccine effectively protected against EV71 C4a and CVA16 infections. The examination of tissue damage and viral contents in various organs indicated that both monovalent and bivalent antisera reduced EV71 C4a viral load in the brainstem, and no significant tissue damage was observed. During CVA16 infection, the monovalent and bivalent antisera significantly reduced viral contents in both the brainstem and muscles. These results suggest that passive immunity by monovalent and bivalent antisera can effectively protect against EV71 C4a and CVA16 infections. Thus, the development of a bivalent vaccine that can provide broad protection against both CV and EV infections may be a promising strategy in preventing HFMD.


Subject(s)
Enterovirus A, Human , Hand, Foot and Mouth Disease , Humans , Animals , Mice , Enterovirus A, Human/genetics , Vaccines, Combined , Hand, Foot and Mouth Disease/prevention & control , Immune Sera , Mice, Transgenic
19.
Biomol Ther (Seoul) ; 31(3): 350-358, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37041034

ABSTRACT

Hand-foot-and-mouth disease (HFMD) is a viral infectious disease that occurs in children under 5 years of age. Its main causes are coxsackievirus (CV) and enterovirus (EV). Since there are no efficient therapeutics for HFMD, vaccines are effective in preventing the disease. To develop broad coverage against CV and EV, the development of a bivalent vaccine form is needed. The Mongolian gerbil is an efficient and suitable animal model of EV71 C4a and CVA16 infection used to investigate vaccine efficacy following direct immunization. In this study, Mongolian gerbils were immunized with a bivalent inactivated EV71 C4a and inactivated CVA16 vaccine to test their effectiveness against viral infection. Bivalent vaccine immunization resulted in increased Ag-specific IgG antibody production; specifically, EV71 C4a-specific IgG was increased with medium and high doses and CVA16-specific IgG was increased with all doses of immunization. When gene expression of T cell-biased cytokines was analysed, Th1, Th2, and Th17 responses were found to be highly activated in the high-dose immunization group. Moreover, bivalent vaccine immunization mitigated paralytic signs and increased the survival rate following lethal viral challenges. When the viral RNA content was determined from various organs, all three doses of bivalent vaccine immunization were found to significantly decrease viral amplification. Upon histologic examination, EV71 C4a and CVA16 induced tissue damage to the heart and muscle. However, bivalent vaccine immunization alleviated this in a dose-dependent manner. These results suggest that the bivalent inactivated EV71 C4a/CVA16 vaccine could be a safe and effective candidate HFMD vaccine.

20.
Exp Mol Med ; 55(2): 401-412, 2023 02.
Article in English | MEDLINE | ID: mdl-36759578

ABSTRACT

Endoplasmic reticulum stress is closely associated with the onset and progression of inflammatory bowel disease. ERdj5 is an endoplasmic reticulum-resident protein disulfide reductase that mediates the cleavage and degradation of misfolded proteins. Although ERdj5 expression is significantly higher in the colonic tissues of patients with inflammatory bowel disease than in healthy controls, its role in inflammatory bowel disease has not yet been reported. In the current study, we used ERdj5-knockout mice to investigate the potential roles of ERdj5 in inflammatory bowel disease. ERdj5 deficiency causes severe inflammation in mouse colitis models and weakens gut barrier function by increasing NF-κB-mediated inflammation. ERdj5 may not be indispensable for goblet cell function under steady-state conditions, but its deficiency induces goblet cell apoptosis under inflammatory conditions. Treatment of ERdj5-knockout mice with the chemical chaperone ursodeoxycholic acid ameliorated severe colitis by reducing endoplasmic reticulum stress. These findings highlight the important role of ERdj5 in preserving goblet cell viability and function by resolving endoplasmic reticulum stress.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , HSP40 Heat-Shock Proteins/metabolism , Protein Folding , Goblet Cells/metabolism , Inflammation , Mice, Knockout , Endoplasmic Reticulum Stress , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Apoptosis , Molecular Chaperones/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL