Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 24(24): 14481-14490, 2020 12.
Article in English | MEDLINE | ID: mdl-33140535

ABSTRACT

Macrophages are fundamental components of inflammation in post-myocardial infarction (MI) and contribute to adverse cardiac remodelling and heart failure. However, the regulatory mechanisms in macrophage activation have not been fully elucidated. Previous studies showed that myeloid-associated immunoglobulin-like receptor II (MAIR-II) is involved in inflammatory responses in macrophages. However, its role in MI is unknown. Thus, this study aimed to determine a novel role and mechanism of MAIR-II in MI. We first identified that MAIR-II-positive myeloid cells were abundant from post-MI days 3 to 5 in infarcted hearts of C57BL/6J (WT) mice induced by permanent left coronary artery ligation. Compared to WT, MAIR-II-deficient (Cd300c2-/- ) mice had longer survival, ameliorated cardiac remodelling, improved cardiac function and smaller infarct sizes. Moreover, we detected lower pro-inflammatory cytokine and fibrotic gene expressions in Cd300c2-/- -infarcted hearts. These mice also had less infiltrating pro-inflammatory macrophages following MI. To elucidate a novel molecular mechanism of MAIR-II, we considered macrophage activation by Toll-like receptor (TLR) 9-mediated inflammation. In vitro, we observed that Cd300c2-/- bone marrow-derived macrophages stimulated by a TLR9 agonist expressed less pro-inflammatory cytokines compared to WT. In conclusion, MAIR-II may enhance inflammation via TLR9-mediated macrophage activation in MI, leading to adverse cardiac remodelling and poor prognosis.


Subject(s)
Macrophage Activation/genetics , Macrophages/metabolism , Myocardial Infarction/complications , Receptors, Polymeric Immunoglobulin/deficiency , Toll-Like Receptor 9/metabolism , Ventricular Remodeling/genetics , Animals , Biomarkers , Biopsy , Cytokines/metabolism , DNA, Mitochondrial , Disease Models, Animal , Disease Susceptibility , Echocardiography , Inflammation Mediators/metabolism , Kaplan-Meier Estimate , Macrophages/immunology , Mice , Mice, Knockout , Myocardial Infarction/etiology , Myocardial Infarction/mortality , Myocardium/pathology , Prognosis
2.
PLoS One ; 13(6): e0198434, 2018.
Article in English | MEDLINE | ID: mdl-29856838

ABSTRACT

BACKGROUND: Polymeric immunoglobulin receptor (pIgR) transport of secretory immunoglobulin A (SIgA) to mucosal surfaces is thought to promote gut integrity and immunity to Salmonella enterica serovar Typhimurium (S. Typhimurium), an invasive pathogen in mice. To elucidate potential mechanisms, we assessed intestinal barrier function and both oral and systemic S. Typhimurium virulence in pIgR knockout (KO) and wildtype (WT) mice. METHODS: In uninfected animals, we harvested jejunal segments for Ussing chamber analyses of transepithelial resistance (TER); mesenteric lymph nodes (mLN) for bacterial culture; and serum and stool for IgA. Separately, we infected mice either orally or intravenously (IV) with S. Typhimurium to compare colonization, tissue dynamics, and inflammation between KOs and WTs. RESULTS: Uninfected KOs displayed decreased TER and dramatically increased serum IgA and decreased fecal IgA vs. WT; however, KO mLNs yielded fewer bacterial counts. Remarkably, WTs challenged orally with S. Typhimurium exhibited increased splenomegaly, tissue colonization, and pro-inflammatory cytokines vs. pIgR KOs, which showed increased survival following either oral or IV infection. CONCLUSIONS: Absence of pIgR compromises gut integrity but does not exacerbate bacterial translocation nor S. Typhimurium infection. These findings raise the possibility that immune adaptation to increased gut permeability and elevated serum IgA in the setting of SIgA deficiency provides compensatory protection against invasive gut pathogens.


Subject(s)
Receptors, Polymeric Immunoglobulin/genetics , Salmonella Infections, Animal/pathology , Salmonella enterica/pathogenicity , Administration, Oral , Animals , Cytokines/blood , Feces/chemistry , Immunoglobulin A/analysis , Immunoglobulin A/blood , Injections, Intravenous , Intestines/pathology , Lymph Nodes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Polymeric Immunoglobulin/deficiency , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella enterica/physiology , Splenomegaly/etiology , Survival Rate
3.
Nat Commun ; 7: 11240, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27046438

ABSTRACT

Mechanisms driving persistent airway inflammation in chronic obstructive pulmonary disease (COPD) are incompletely understood. As secretory immunoglobulin A (SIgA) deficiency in small airways has been reported in COPD patients, we hypothesized that immunobarrier dysfunction resulting from reduced SIgA contributes to chronic airway inflammation and disease progression. Here we show that polymeric immunoglobulin receptor-deficient (pIgR(-/-)) mice, which lack SIgA, spontaneously develop COPD-like pathology as they age. Progressive airway wall remodelling and emphysema in pIgR(-/-) mice are associated with an altered lung microbiome, bacterial invasion of the airway epithelium, NF-κB activation, leukocyte infiltration and increased expression of matrix metalloproteinase-12 and neutrophil elastase. Re-derivation of pIgR(-/-) mice in germ-free conditions or treatment with the anti-inflammatory phosphodiesterase-4 inhibitor roflumilast prevents COPD-like lung inflammation and remodelling. These findings show that pIgR/SIgA deficiency in the airways leads to persistent activation of innate immune responses to resident lung microbiota, driving progressive small airway remodelling and emphysema.


Subject(s)
Aging/immunology , Aminopyridines/pharmacology , Benzamides/pharmacology , Microbiota/immunology , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Emphysema/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Aging/pathology , Airway Remodeling/immunology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/immunology , Cyclopropanes/pharmacology , Disease Models, Animal , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Immunity, Innate , Immunoglobulin A, Secretory/genetics , Leukocyte Elastase/genetics , Leukocyte Elastase/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/genetics , Pulmonary Emphysema/microbiology , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/immunology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology
4.
PLoS One ; 10(4): e0121979, 2015.
Article in English | MEDLINE | ID: mdl-25835383

ABSTRACT

Selective breeding to introduce a gene mutation from one mouse strain onto the genetic background of another strain invariably produces "hitchhiking" (i.e. flanking) genomic intervals, which may independently affect a disease trait of interest. To investigate a role for the polymeric Ig receptor in autoimmune diabetes, a congenic nonobese diabetic (NOD) mouse strain was generated that harbors a Pigr null allele derived from C57BL/6 (B6) mice. These pIgR-deficient NOD mice exhibited increased serum IgA along with an increased diabetes incidence. However, the Pigr null allele was encompassed by a relatively large "hitchhiking" genomic interval that was derived from B6 mice and overlaps Idd5.4, a susceptibility locus for autoimmune diabetes. Additional congenic NOD mouse strains, harboring smaller B6-derived intervals, confirmed Idd5.4 independently of the other three known susceptibility loci on chromosome 1, and further localized Idd5.4 to an interval proximal to Pigr. Moreover, these congenic NOD mice showed that B6 mice harbor a more diabetogenic allele than NOD mice for this locus. The smallest B6-derived interval encompassing the Pigr null allele may, however, confer a small degree of protection against diabetes, but this protection appears to be dependent on the absence of the diabetogenic B6 allele for Idd5.4. This study provides another example of the potential hidden effects of "hitchhiking" genomic intervals and how such intervals can be used to localize disease susceptibility loci.


Subject(s)
Chromosomes, Mammalian/chemistry , Diabetes Mellitus, Type 1/genetics , Genetic Loci , Genetic Predisposition to Disease , Genome , Receptors, Polymeric Immunoglobulin/genetics , Age Factors , Alleles , Animals , Chromosome Mapping , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Humans , Immunoglobulin A/blood , Immunoglobulin A/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/immunology
5.
Nat Commun ; 5: 4710, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25134989

ABSTRACT

Inflammatory monocytes play an important role in host defense against infections. However, the regulatory mechanisms of transmigration into infected tissue are not yet completely understood. Here we show that mice deficient in MAIR-II (also called CLM-4 or LMIR2) are more susceptible to caecal ligation and puncture (CLP)-induced peritonitis than wild-type (WT) mice. Adoptive transfer of inflammatory monocytes from WT mice, but not from MAIR-II, TLR4 or MyD88-deficient mice, significantly improves survival of MAIR-II-deficient mice after CLP. Migration of inflammatory monocytes into the peritoneal cavity after CLP, which is dependent on VLA-4, is impaired in above mutant and FcRγ chain-deficient mice. Lipopolysaccharide stimulation induces association of MAIR-II with FcRγ chain and Syk, leading to enhancement of VLA-4-mediated adhesion to VCAM-1. These results indicate that activation of MAIR-II/FcRγ chain by TLR4/MyD88-mediated signalling is essential for the transmigration of inflammatory monocytes from the blood to sites of infection mediated by VLA-4.


Subject(s)
Cell Movement/physiology , Inflammation/pathology , Integrin alpha4beta1/physiology , Monocytes/pathology , Receptors, Immunologic/physiology , Receptors, Polymeric Immunoglobulin/physiology , Toll-Like Receptor 4/physiology , Animals , Cecum , Cell Adhesion/physiology , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/physiopathology , Ligation , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/physiology , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Peritonitis/etiology , Peritonitis/pathology , Peritonitis/physiopathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
6.
Infect Immun ; 82(2): 522-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478068

ABSTRACT

Clostridium difficile rivals methicillin-resistant Staphylococcus aureus as the primary hospital-acquired infection. C. difficile infection (CDI) caused by toxins A and/or B can manifest as mild diarrhea to life-threatening pseudomembranous colitis. Although most patients recover fully from CDI, ~20% undergo recurrent disease. Several studies have demonstrated a correlation between anti-toxin antibody (Ab) and decreased recurrence; however, the contributions of the systemic and mucosal Ab responses remain unclear. Our goal was to use the CDI mouse model to characterize the protective immune response to C. difficile. C57BL/6 mice infected with epidemic C. difficile strain BI17 developed protective immunity against CDI and did not develop CDI upon rechallenge; they generated systemic IgG and IgA as well as mucosal IgA Ab to toxin. To determine if protective immunity to C. difficile could be generated in immunodeficient individuals, we infected CD4(-/-) mice and found that they generated both mucosal and serum IgA anti-toxin Abs and were protected from CDI upon rechallenge, with protection dependent on major histocompatibility complex class II (MHCII) expression; no IgG anti-toxin Ab was found. We found that protection was likely due to neutralizing mucosal IgA Ab. In contrast, pIgR(-/-) mice, which lack the receptor to transcytose polymeric Ab across the epithelium, were also protected from CDI, suggesting that although mucosal anti-toxin Ab may contribute to protection, it is not required. We conclude that protection from CDI can occur by several mechanisms and that the mechanism of protection is determined by the state of immunocompetence of the host.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Receptors, Polymeric Immunoglobulin/immunology , Animals , Antibodies, Bacterial/blood , Clostridium Infections/microbiology , Clostridium Infections/prevention & control , Disease Models, Animal , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Polymeric Immunoglobulin/deficiency
7.
J Biochem ; 154(6): 541-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089533

ABSTRACT

Fucosylation of GA1 in murine intestinal epithelia occurs through transcriptional induction of α1,2-fucosyltransferase along with bacterial infection, but the mechanism has not been clearly characterized as to whether it is induced as a result of an immune response to bacteria or of genetic manipulation of the host by bacteria. Accordingly, we analysed the expression of fucosyl GA1 (FGA1) and fucosyltransferase activity in the digestive tracts of immune-deficient scid, nude and pIgR(-/-) mice. In comparison with those in control mice bred under the same SPF circumstances, the amount of FGA1 and the α1,2-fucosyltransferase activity were significantly increased in the immune-deficient mice, indicating that the immune system is not involved in induction of the α1,2-fucosyltransferase gene. Reflecting the enhanced synthesis of FGA1, the total amounts of FGA1 in the intestinal contents of immune-deficient mice were higher than those in control mice. Also, the major faecal bacteria grown on a MRS agar plate were different in immune-deficient and control mice as follows, Lactobacillus murinus for scid and pIgR(-/-) mice, and Lactobacillus johnsonii for their control, and Enterococcus faecalis for nude mice and Lactococcus garvieae for the control, indicating that an alteration in the intestinal lactobacilli is partly involved in the induction of α1,2-fucosyltransferase.


Subject(s)
Gastrointestinal Tract/enzymology , Gastrointestinal Tract/immunology , Glycolipids/biosynthesis , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/immunology , Animals , Female , Fucosyltransferases/metabolism , Gastrointestinal Tract/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mice, SCID
8.
Viral Immunol ; 26(2): 133-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23509985

ABSTRACT

Intranasal immunization is currently used to deliver live virus vaccines such as influenza. However, to develop an intranasal vaccine to deliver inactivated virus, a safe and effective adjuvant is necessary to enhance the mucosal immune response. Here, we demonstrate the effectiveness of a chitosan microparticle (1-20 µm, 50 kDa, degree of deacetylation=85%) and a cationized chitosan (1000 kDa, degree of deacetylation=85%) derived from natural crab shells as adjuvants for an intranasal vaccine candidate. We examined the effectiveness of chitosan derivatives as an adjuvant by co-administering them with ovalbumin (OVA) intranasally in BALB/c mice, polymeric Ig receptor knockout (pIgR-KO) mice, and cynomolgus monkeys (Macaca fascicularis). pIgR-KO mice were used to evaluate S-IgA production on the mucosal surface without nasal swab collection. Administration of OVA with chitosan microparticles or cationized chitosan induced a high OVA-specific IgA response in the serum of pIgR-KO mice and a high IgG response in the serum of BALB/c mice and cynomolgus monkeys. We also found that administration of chitosan derivatives did not have a detrimental effect on cynomolgus monkeys as determined by complete blood count, blood chemistries, and gross pathology results. These results suggest that chitosan derivatives are safe and effective mucosal adjuvants for intranasal vaccination.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Chitosan/administration & dosage , Chitosan/adverse effects , Administration, Intranasal , Animals , Chitosan/chemistry , Female , Immunoglobulin A, Secretory/analysis , Immunoglobulin A, Secretory/blood , Immunoglobulin G/blood , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Weight , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Polymeric Immunoglobulin/deficiency
9.
Eur J Immunol ; 42(11): 2959-70, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22865203

ABSTRACT

Innate and adaptive mucosal defense mechanisms ensure a homeostatic relationship with the large and complex mutualistic gut microbiota. Dimeric IgA and pentameric IgM are transported across the intestinal epithelium via the epithelial polymeric Ig receptor (pIgR) and provide a significant portion of the first line of natural or adaptive antibody-mediated immune defense of the intestinal mucosa. We found that colonic epithelial cells from pIgR KO mice differentially expressed (more than twofold change) more than 200 genes compared with cells from WT mice, and upregulated the expression of antimicrobial peptides in a commensal-dependent manner. Detailed profiling of microbial communities based on 16S rRNA genes revealed differences in the commensal microbiota between pIgR KO and WT mice. Furthermore, we found that pIgR KO mice showed increased susceptibility to dextran sulfate sodium-induced colitis, and that this was driven by their conventional intestinal microbiota. Thus, in the absence of pIgR, the stability of the commensal microbiota is disturbed, gut homeostasis is compromised, and the outcome of colitis is significantly worsened.


Subject(s)
Adaptive Immunity/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Metagenome/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/immunology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Colitis/immunology , Colitis/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Intestinal Mucosa/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA/chemistry , RNA/genetics , Random Allocation , Receptors, Polymeric Immunoglobulin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
10.
J Exp Med ; 208(8): 1661-71, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21727189

ABSTRACT

DAP12, an immunoreceptor tyrosine-based activation motif-bearing adapter protein, is involved in innate immunity mediated by natural killer cells and myeloid cells. We show that DAP12-deficient mouse B cells and B cells from a patient with Nasu-Hakola disease, a recessive genetic disorder resulting from loss of DAP12, showed enhanced proliferation after stimulation with anti-IgM or CpG. Myeloid-associated immunoglobulin-like receptor (MAIR) II (Cd300d) is a DAP12-associated immune receptor. Like DAP12-deficient B cells, MAIR-II-deficient B cells were hyperresponsive. Expression of a chimeric receptor composed of the MAIR-II extracellular domain directly coupled to DAP12 into the DAP12-deficient or MAIR-II-deficient B cells suppressed B cell receptor (BCR)-mediated proliferation. The chimeric MAIR-II-DAP12 receptor recruited the SH2 domain-containing protein tyrosine phosphatase 1 (SHP-1) after BCR stimulation. DAP12-deficient mice showed elevated serum antibodies against self-antigens and enhanced humoral immune responses against T cell-dependent and T cell-independent antigens. Thus, DAP12-coupled MAIR-II negatively regulates B cell-mediated adaptive immune responses.


Subject(s)
Adaptive Immunity/immunology , Adaptor Proteins, Signal Transducing/immunology , B-Lymphocytes/immunology , Lipodystrophy/immunology , Osteochondrodysplasias/immunology , Receptors, Natural Killer Cell/immunology , Recombinant Fusion Proteins/immunology , Subacute Sclerosing Panencephalitis/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Genetic Vectors , Humans , Lentivirus , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Natural Killer Cell/deficiency , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/immunology , Receptors, Polymeric Immunoglobulin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Allergy ; 65(5): 561-70, 2010 May.
Article in English | MEDLINE | ID: mdl-19886928

ABSTRACT

BACKGROUND: Mucosal immunity protects the epithelial barrier by immune exclusion of foreign antigens and by anti-inflammatory tolerance mechanisms, but there is a continuing debate about the role of secretory immunoglobulins (SIgs), particularly SIgA, in the protection against allergy and other inflammatory diseases. Lack of secretory antibodies may cause immune dysfunction and affect mucosally induced (oral) tolerance against food antigens. METHODS: We used polymeric Ig receptor (pIgR) knockout (KO) mice, which cannot export SIgA or SIgM, to study oral tolerance induction by ovalbumin (OVA) feeding and for parenteral antigen sensitization in the same animal. RESULTS: Remarkable systemic hyperreactivity was observed in pIgR KO mice, as 50% died after intradermal OVA challenge, which was not seen in similarly sensitized and challenged wild-type (WT) mice. Oral tolerance induced by OVA completely protected the sensitized pIgR KO mice against anaphylaxis and suppressed antibody levels (particularly IgG1) as well as delayed-type hypersensitivity (DTH) to OVA. Delayed-type hypersensitivity to a bystander antigen, human serum albumin, was also suppressed and T-cell proliferation against OVA in vitro was reduced in tolerized compared with non-tolerized pIgR KO mice. This effect was largely mediated by CD25+ T cells. Adoptive transfer of splenic putative regulatory T cells (CD4+ CD25+) obtained from OVA-fed pIgR KO mice to naïve WT mice mediated suppression of DTH against OVA after sensitization of the recipients. CONCLUSION: Compensatory regulatory T-cell function becomes critical in pIgR-deficient mice to avoid the potentially catastrophic effects of systemic immune hyperreactivity, presumably resulting from defective secretory antibody-mediated immune exclusion of microbial components.


Subject(s)
Hypersensitivity, Delayed/immunology , Immune Tolerance/immunology , Immunity, Mucosal/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Adoptive Transfer , Animals , Enzyme-Linked Immunosorbent Assay , Female , Food Hypersensitivity/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/immunology , T-Lymphocytes, Regulatory/immunology
12.
Int Immunol ; 19(3): 257-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17255112

ABSTRACT

The humoral response to the gastrointestinal (GI) flora was analyzed in secretory Ig (sIg)-deficient polymeric IgR (pIgR)(-/-) mice and otherwise congenic C57BL/6 mice. While both strains carried an ileal flora of similar size and composition, increased bacterial translocation to mesenteric lymph node was demonstrated in pIgR(-/-) mice. Serum IgA was greatly increased in pIgR(-/-) mice compared with C57BL/6 mice and reacted with commensal organisms and food. Serum IgG levels in pIgR(-/-) mice were increased to 6-fold above that of C57BL/6 mice and included specificities that bound to selected flora antigens. The enhanced recognition of flora antigens in pIgR(-/-) mice was explored using ovalbumin (OVA)-specific CD4(+) T cells and feeding of low concentrations of OVA. Increased proliferation of transgenic T cells was observed in pIgR(-/-) mice, relative to C57BL/6 mice, suggesting elevated net uptake of protein antigens from the GI tract in the absence of sIg. These studies suggest that there is increased recognition of GI flora antigens by systemic antibodies in pIgR(-/-) mice, most probably as a result of increased access of antigens from the GI flora to the systemic immune compartment, and support the hypothesis that a major function of the secretory immune system is to return environmental antigens to mucosal surfaces.


Subject(s)
Antigens, Bacterial/immunology , Bacteria/immunology , Gastrointestinal Tract/microbiology , Immunoglobulin A, Secretory/blood , Immunoglobulin G/blood , Receptors, Polymeric Immunoglobulin/immunology , Administration, Oral , Adoptive Transfer , Animals , Antibody Formation , Antigens, Bacterial/metabolism , Bacteria/classification , Bacterial Translocation , Female , Gastrointestinal Tract/metabolism , Ileum/microbiology , Intestinal Absorption , Lymph Nodes/microbiology , Lymphocyte Activation , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
13.
J Immunol ; 177(9): 6281-90, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056558

ABSTRACT

The polymeric Ig receptor (pIgR) is conserved in mammals and has an avian homologue, suggesting evolutionarily important functions in vertebrates. It transports multimeric IgA and IgM across polarized epithelia and is highly expressed in the intestine, yet little direct evidence exists for its importance in defense against common enteric pathogens. In this study, we demonstrate that pIgR can play a critical role in intestinal defense against the lumen-dwelling protozoan parasite Giardia, a leading cause of diarrheal disease. The receptor was essential for the eradication of Giardia when high luminal IgA levels were required. Clearance of Giardia muris, in which IgA plays a dominant role, was severely compromised in pIgR-deficient mice despite significant fecal IgA output at 10% of normal levels. In contrast, eradication of the human strain Giardia lamblia GS/M, for which adaptive immunity is less IgA dependent in mice, was unaffected by pIgR deficiency, indicating that pIgR had no physiologic role when lower luminal IgA levels were sufficient for parasite elimination. Immune IgA was greatly increased in the serum of pIgR-deficient mice, conferred passive protection against Giardia, and recognized several conserved giardial Ags, including ornithine carbamoyltransferase, arginine deiminase, alpha-enolase, and alpha- and beta-giardins, that are also detected in human giardiasis. Corroborative observations were made in mice lacking the J chain, which is required for pIgR-dependent transepithelial IgA transport. These results, together with prior data on pIgR-mediated immune neutralization of luminal cholera toxin, suggest that pIgR is essential in intestinal defense against pathogenic microbes with high-level and persistent luminal presence.


Subject(s)
Giardia , Giardiasis/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Receptors, Polymeric Immunoglobulin/physiology , Animals , Antigens, Protozoan/analysis , Antigens, Protozoan/immunology , Feces/chemistry , Giardiasis/genetics , Immunity/genetics , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin A/metabolism , Intestines/immunology , Intestines/parasitology , Mice , Mice, Mutant Strains , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics
14.
Int Immunol ; 18(5): 807-16, 2006 May.
Article in English | MEDLINE | ID: mdl-16569672

ABSTRACT

It is generally accepted that cellular, and not humoral immunity, plays the crucial role in defense against intracellular bacteria. However, accumulating data indicate the importance of humoral immunity for the defense against a number of intracellular bacteria, including mycobacteria. We have investigated the role of secretory IgA, the main isotype found in mucosal tissues, in protection against mycobacterial infection, using polymeric IgR (pIgR)-deficient mice. Characterization of the humoral response induced after intra-nasal immunizations with the mycobacterial antigen PstS-1 revealed a loss of antigen-specific IgA response in saliva from the knockout mice. IgA level in the bronchoalveolar lavage of knockout mice was similar to wild-type level, although the IgA antibodies must have reached the lumen by other means than pIgR-mediated transport. Infection with Mycobacterium bovis bacillus Calmette-Guérin (BCG) demonstrated that the immunized pIgR-/- mice were more susceptible to BCG infection than immunized wild-type mice, based on higher bacterial loads in the lungs. This was accompanied by a reduced production of both IFN-gamma and tumor necrosis factor-alpha (TNF-alpha) in the lungs. Additionally, the pIgR-/- mice displayed reduced natural resistance to mycobacterial infection proved by significantly higher bacterial growth in their lungs compared with wild-type mice after infection with virulent Mycobacterium tuberculosis. The knockout mice appeared to have a delayed mycobacteria-induced immune response with reduced expression of protective mediators, such as IFN-gamma, TNF-alpha, inducible nitric oxide synthase and regulated upon activation normal T cell sequence, during early infection. Collectively, our results show that actively secreted IgA plays a role in protection against mycobacterial infections in the respiratory tract, by blocking entrance of bacilli into the lungs, in addition to modulation of the mycobacteria-induced pro-inflammatory response.


Subject(s)
Immunoglobulin A, Secretory/immunology , Mycobacterium tuberculosis/immunology , Receptors, Polymeric Immunoglobulin/immunology , Respiratory Tract Infections/immunology , Tuberculosis/immunology , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/genetics , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Respiratory Tract Infections/genetics , Respiratory Tract Infections/microbiology , Secretory Component/immunology , Tuberculosis/genetics , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
15.
J Exp Med ; 203(1): 21-6, 2006 Jan 23.
Article in English | MEDLINE | ID: mdl-16390940

ABSTRACT

The production of IgA is induced in an antigen-unspecific manner by commensal flora. These secretory antibodies (SAbs) may bind multiple antigens and are thought to eliminate commensal bacteria and self-antigens to avoid systemic recognition. In this study, we addressed the role of "innate" SAbs, i.e., those that are continuously produced in normal individuals, in protection against infection of the gastrointestinal tract. We used polymeric immunoglobulin receptor (pIgR-/-) knock-out mice, which are unable to bind and actively transport dimeric IgA and pentameric IgM to the mucosae, and examined the role of innate SAbs in protection against the invasive pathogen Salmonella typhimurium. In vitro experiments suggested that innate IgA in pIgR-/- serum bound S. typhimurium in a cross-reactive manner which inhibited epithelial cell invasion. Using a "natural" infection model, we demonstrated that pIgR-/- mice are profoundly sensitive to infection with S. typhimurium via the fecal-oral route and, moreover, shed more bacteria that readily infected other animals. These results imply an important evolutionary role for innate SAbs in protecting both the individual and the herd against infections, and suggest that the major role of SAbs may be to prevent the spread of microbial pathogens throughout the population, rather than protection of local mucosal surfaces.


Subject(s)
Antibodies, Bacterial/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Salmonella Infections, Animal/immunology , Salmonella typhimurium/pathogenicity , Animals , Cell Line , Colony Count, Microbial , Dogs , Feces/microbiology , Immunity, Innate , Immunoglobulin A/blood , Intestine, Small/immunology , Intestine, Small/microbiology , Lethal Dose 50 , Mice , Mice, Inbred Strains , Mice, Knockout , Peyer's Patches/immunology , Peyer's Patches/microbiology , Receptors, Polymeric Immunoglobulin/blood , Receptors, Polymeric Immunoglobulin/genetics , Salmonella Infections, Animal/mortality , Salmonella Infections, Animal/transmission
16.
Eur J Immunol ; 35(1): 180-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15593123

ABSTRACT

Secretory IgA (SIgA) is widely held to be responsible for the defense of the mucosae against pathogenics and other potentially harmful agents. In this study, polymeric Ig receptor (pIgR) knockout mice, which lack secretory antibodies (SAb), were used to investigate the role of vaccine-elicited SAb in protection against gastrointestinal bacterial infections. An essential role for specific SAb in protection against Vibrio cholerae was evident from experiments showing that vaccinated pIgR(-/-) mice, but not vaccinated C57BL/6 mice, were susceptible to cholera toxin challenge. Vaccination of C57BL/6 mice with Salmonella typhimurium elicited strong antigen-specific, mucosal responses, which blocked in vitro invasion of epithelia. However, vaccinated C57BL/6 and pIgR(-/-) mice were equally resistant to challenge infection with virulent S. typhimurium. Finally, we investigated the importance of SIgA in protection against recurrent infections with Citrobacter rodentium. Although higher numbers of bacteria were detected early after challenge infection in feces of vaccinated pIgR(-/-) mice compared with vaccinated C57BL/6 mice, both mouse strains showed complete clearance after 9 days. These results suggested that, in immune animals, SIgA is crucial for the protection of gastrointestinal surfaces against secreted bacterial toxins, may inhibit early colonization by C. rodentium, but is not essential for protection against re-infection with S. typhimurium or C. rodentium.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/prevention & control , Bacterial Vaccines/pharmacology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/prevention & control , Immunoglobulin A, Secretory/immunology , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibody Specificity , Bacterial Vaccines/administration & dosage , Cholera Toxin/immunology , Cholera Toxin/toxicity , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Immunity, Mucosal , Immunization , Immunoglobulin A, Secretory/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity
17.
J Immunol ; 173(7): 4576-81, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383591

ABSTRACT

The importance of IgA for protection at mucosal surfaces remains unclear, and in fact, it has been reported that IgA-deficient mice have fully functional vaccine-induced immunity against several bacterial and viral pathogens. The role of respiratory Ab in preventing colonization by Streptococcus pneumoniae has now been examined using polymeric IgR knockout (pIgR(-/-)) mice, which lack the ability to actively secrete IgA into the mucosal lumen. Intranasal vaccination with a protein conjugate vaccine elicited serotype-specific anti-capsular polysaccharide Ab locally and systemically, and pIgR(-/-) mice produced levels of total serum Ab after vaccination that were similar to wild-type mice. However, pIgR(-/-) mice had approximately 5-fold more systemic IgA and 6-fold less nasal IgA Ab than wild-type mice due to defective transport into mucosal tissues. Wild-type, but not pIgR(-/-) mice were protected against infection with serotype 14 S. pneumoniae, which causes mucosal colonization but does not induce systemic inflammatory responses in mice. The relative importance of secretory IgA in host defense was further shown by the finding that intranasally vaccinated IgA gene-deficient mice were not protected from colonization. Although secretory IgA was found to be important for protection against nasal carriage, it does not appear to have a crucial role in immunity to systemic pneumococcus infection, because both vaccinated wild-type and pIgR(-/-) mice were fully protected from lethal systemic infection by serotype 3 pneumococci. The results demonstrate the critical role of secretory IgA in protection against pneumococcal nasal colonization and suggest that directed targeting to mucosal tissues will be needed for effective vaccination in humans.


Subject(s)
Immunoglobulin A, Secretory/metabolism , Nasopharynx/immunology , Nasopharynx/microbiology , Receptors, Polymeric Immunoglobulin/physiology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Streptococcal Infections/prevention & control , Streptococcus pneumoniae/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibodies, Bacterial/physiology , Bacterial Capsules/administration & dosage , Bacterial Capsules/immunology , Immunity, Mucosal , Immunoglobulin A, Secretory/genetics , Immunoglobulin A, Secretory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasopharynx/metabolism , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/immunology , Protein Transport/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Respiratory Mucosa/metabolism , Serotyping , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/growth & development , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
18.
Appl Environ Microbiol ; 69(4): 2100-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12676689

ABSTRACT

Terminal restriction fragment length polymorphism (T-RFLP) analysis was conducted on the 16S rRNA genes of the bacterial communities colonizing the epithelial surfaces of the terminal ilea of open conventionally housed mice in an institutional small-animal facility. Polymeric-immunoglobulin-receptor-deficient (pIgR(-/-)) mice that were unable to secrete antibodies across mucosal surfaces were cohoused with normal and otherwise genetically identical wild-type (C57BL/6) mice for 4 weeks. If secretory antibodies played a role in modeling the gastrointestinal microbiota, C57BL/6 mice would have had a more distinct and uniform microbiota than their pIgR(-/-) cage mates. The T-RFLP profiles of the bacterial communities were compared by using Sorensen's pairwise similarity coefficient, a newly developed weighted pairwise similarity coefficient, and on the basis of Shannon's and Simpson's diversity indices. No systematic differences were observed between the dominant components of the mucosa-associated bacterial communities of the terminal ileal walls of the two types of mice, indicating that secretory antibodies do not control the composition of this microbiota. Similar analyses of experiments conducted at two different times, between which the bacterial community composition of the mouse colony in the small-animal facility appeared to have changed, showed that differences could have been detected, had they existed.


Subject(s)
Antibodies, Bacterial/immunology , Bacteria/classification , Ecosystem , Ileum/immunology , Ileum/microbiology , Immunoglobulin A, Secretory/immunology , Animals , Bacteria/genetics , Bacteria/isolation & purification , DNA, Ribosomal/analysis , Housing, Animal , Immunity, Mucosal , Mice , Mice, Inbred C57BL , Polymorphism, Restriction Fragment Length , RNA, Ribosomal, 16S/genetics , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics
19.
J Immunol ; 170(5): 2531-9, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12594279

ABSTRACT

Secretory IgA (SIgA) is the most characteristic component of the mucosal immune system and has long been considered the major protective factor that prevents pathogens from invading hosts through the mucosae. Recent studies, however, have suggested that complete immunity against a range of mucosal bacterial and viral pathogens can be achieved in the absence of IgA. Therefore, to further dissect the role of SIgA, we generated mice deficient in the polymeric Ig receptor (pIgR(-/-) mice). As a result of an inability to transport dimeric IgA to the secretions, pIgR(-/-) mice are deficient in SIgA and accumulate circulating dimeric IgA, with serum levels 100-fold greater than those observed in normal mice. Examination of lamina propria mononuclear cells showed that pIgR(-/-) mice had approximately 3 times as many IgA-secreting cells as C57BL/6 mice. Further analysis showed that these cells displayed the differentiated IgA(+) B220(-) phenotype and accounted for a 2-fold increase in the number of lamina propria blast cells in the pIgR(-/-) mice. Subsequent experiments showed that OVA-specific CD4(+) T cell expansion following OVA feeding was not elevated in pIgR(-/-) mice. Furthermore, no differences in CD8(+) T cell tolerance or induction of influenza virus-specific CD8(+) T cells were detected in pIgR(-/-) mice compared with controls. Therefore, while SIgA is clearly involved in maintaining some parameters of mucosal homeostasis in the intestine, the mechanisms associated with its barrier function and the clinical consequences of its deficiency are yet to be identified.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Homeostasis/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptors, Polymeric Immunoglobulin/physiology , Administration, Oral , Animals , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocyte Subsets/cytology , Dimerization , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Homeostasis/genetics , IgA Deficiency/genetics , IgA Deficiency/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/blood , Immunoglobulin A, Secretory/genetics , Intestinal Mucosa/cytology , Lymphocyte Activation/genetics , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Up-Regulation/genetics , Up-Regulation/immunology
20.
J Immunol ; 168(6): 2930-8, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11884464

ABSTRACT

The role of secretory IgA in conferring cross-protective immunity was examined in polymeric (p)IgR knockout (KO) mice immunized intranasally with different inactivated vaccines prepared from A/PR/8/34 (H1N1), A/Yamagata/120/86 (H1N1), A/Beijing/262/95 (H1N1), and B/Ibaraki/2/85 viruses and infected with the A/PR/8/34 virus in the upper respiratory tract (RT)-restricting volume. In wild-type mice, immunization with A/PR/8/34 or its variant (A/Yamagata/120/86 and A/Beijing/262/95) vaccines conferred complete protection or partial cross-protection against infection, while the B-type virus vaccine failed to provide protection. The protection or cross-protection was accompanied by an increase in the nasal A/PR/8/34 hemagglutinin-reactive IgA concentration, which was estimated to be >30 times the serum IgA concentration and much higher than the nasal IgG concentration. In contrast, the blockade of transepithelial transport of dimeric IgA in pIgR-KO mice reduced the degree of protection or cross-protection, in parallel with the marked increase in serum IgA concentration and the decrease in nasal IgA concentration (about 20 and 0.3 times those in wild-type mice, respectively). The degree of the reduction of protection or cross-protection was moderately reversed by the low but non-negligible level of nasal IgA, transudates from the accumulated serum IgA. These results, together with the absence of the IgA-dependent cross-protection in the lower RT and the unaltered level of nasal or serum IgG in wild-type and pIgR-KO mice, confirm that the actively secreted IgA plays an important role in cross-protection against variant virus infection in the upper RT, which cannot be substituted by serum IgG.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Administration, Intranasal , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/chemistry , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization, Secondary , Immunoglobulin A/biosynthesis , Immunoglobulin A/chemistry , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/prevention & control , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...