Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.140
Filter
1.
Am J Hum Biol ; 35(8): e23897, 2023 08.
Article in English | MEDLINE | ID: mdl-36951242

ABSTRACT

INTRODUCTION: Multiple studies have reported that milk immune content increases for infants experiencing infectious disease (ID) episodes, suggesting that the immune system of milk (ISOM) offers enhanced protection when needed to combat ID. METHODS: To test the hypothesis that ISOM content and/or activity increases during an infant's ID episode, we characterized milk secretory immunoglobulin A (sIgA; a major ISOM constituent) and in vitro interleukin-6 (IL-6) responses to Salmonella enterica and Escherichia coli, as system-level biomarkers of ISOM activity, in a prospective study among 96 mother-infant dyads in Kilimanjaro, Tanzania. RESULTS: After control for covariates, no milk immune variables (sIgA, Coef: 0.03; 95% CI -0.25, 0.32; in vitro IL-6 response to S. enterica, Coef: 0.23; 95% CI: -0.67, 1.13; IL-6 response to E. coli, Coef: -0.11; 95% CI: -0.98, 0.77) were associated with prevalent ID (diagnosed at the initial participation visit). Among infants experiencing an incident ID (diagnosed subsequent to the initial participation), milk immune content and responses were not substantially higher or lower than the initial visit (sIgA, N: 61; p: 0.788; IL-6 response to S. enterica, N: 56; p: 0.896; IL-6 response to E. coli, N: 36; p: 0.683); this was unchanged by exclusion of infants with ID at the time of initial participation. CONCLUSION: These findings are not consistent with the hypothesis that milk delivers enhanced immune protection when infants experience ID. In environments with a high burden of ID, dynamism may be less valuable to maternal reproductive success than stability in the ISOM.


Subject(s)
Escherichia coli Infections , Escherichia coli , Immunoglobulin A, Secretory , Interleukin-6 , Milk, Human , Salmonella Infections , Salmonella enterica , Humans , Female , Milk, Human/chemistry , Interleukin-6/analysis , Interleukin-6/immunology , Salmonella enterica/physiology , Salmonella Infections/immunology , Escherichia coli/physiology , Escherichia coli Infections/immunology , Infant, Newborn , Infant , Tanzania , Prospective Studies , Adult , Cross-Sectional Studies , Immunoenzyme Techniques , Immunoglobulin A, Secretory/analysis , Immunoglobulin A, Secretory/immunology , Longitudinal Studies
2.
Lung ; 200(1): 119-128, 2022 02.
Article in English | MEDLINE | ID: mdl-34825965

ABSTRACT

PURPOSE: Transport of secretory immunoglobulin A (SIgA) through the airway epithelial cell barrier into the mucosal lumen by the polymeric immunoglobulin receptor (pIgR) is an important mechanism of respiratory mucosal host defense. Identification of immunomodulating substances that regulate secretory immunity might have therapeutic implications with regard to an improved immune exclusion. Thus, we sought to analyze secretory immunity under homeostatic and immunomodulating conditions in different compartments of the murine upper and lower respiratory tract (URT&LRT). METHODS: Pigr gene expression in lung, trachea, and nasal-associated lymphoid tissue (NALT) of germ-free mice, specific pathogen-free mice, mice with an undefined microbiome, as well as LPS- and IFN-γ-treated mice was determined by quantitative real-time PCR. IgA levels in bronchoalveolar lavage (BAL), nasal lavage (NAL), and serum were determined by ELISA. LPS- and IFN-γ-treated mice were colonized with Streptococcus pneumoniae and bacterial CFUs were determined in URT and LRT. RESULTS: Respiratory Pigr expression and IgA levels were dependent on the degree of exposure to environmental microbial stimuli. While immunostimulation with LPS and IFN-γ differentially impacts respiratory Pigr expression and IgA in URT vs. LRT, only prophylactic IFN-γ treatment reduces nasal colonization with S. pneumoniae. CONCLUSION: Airway-associated secretory immunity can be partly modulated by exposure to microbial ligands and proinflammatory stimuli. Prophylactic IFN-γ-treatment modestly improves antibacterial immunity in the URT, but this does not appear to be mediated by SIgA or pIgR.


Subject(s)
Immunoglobulin A, Secretory , Receptors, Polymeric Immunoglobulin , Respiratory Mucosa , Animals , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Immunoglobulin A, Secretory/immunology , Immunoglobulin A, Secretory/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, Polymeric Immunoglobulin/immunology , Receptors, Polymeric Immunoglobulin/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism
3.
Front Immunol ; 12: 789748, 2021.
Article in English | MEDLINE | ID: mdl-34938298

ABSTRACT

Recently, a mass spectrometry-based approach was introduced to directly assess the IgG1 immunoglobulin clonal repertoires in plasma. Here we expanded upon this approach by describing a mass spectrometry-based technique to assess specifically the clonal repertoire of another important class of immunoglobulin molecules, IgA1, and show it is efficiently and robustly applicable to either milk or plasma samples. Focusing on two individual healthy donors, whose milk was sampled longitudinally during the first 16 weeks of lactation, we demonstrate that the total repertoire of milk sIgA1 is dominated by only 50-500 clones, even though the human body theoretically can generate several orders of magnitude more clones. We show that in each donor the sIgA1 repertoire only changes marginally and quite gradually over the monitored 16-week period of lactation. Furthermore, the observed overlap in clonal repertoires between the two individual donors is close to non-existent. Mothers provide protection to their newborn infants directly by the transfer of antibodies via breastfeeding. The approach introduced here, can be used to visualize the clonal repertoire transferred from mother to infant and to detect changes in-time in that repertoire adapting to changes in maternal physiology.


Subject(s)
Immunoglobulin A, Secretory/immunology , Mass Spectrometry , Milk, Human/immunology , Proteome/immunology , Proteomics , Breast Milk Expression , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Colostrum/immunology , Colostrum/metabolism , Female , Humans , Immunoglobulin A, Secretory/blood , Lactation , Milk, Human/metabolism
4.
Sci Immunol ; 6(66): eabj5129, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34890255

ABSTRACT

Antibodies secreted at the mucosal surface play an integral role in immune defense by serving to neutralize the pathogen and promote its elimination at the site of entry. Secretory immunoglobulin A (IgA) is a predominant Ig isotype at mucosal surfaces whose epithelial cells express polymeric Ig receptor capable of transporting dimeric IgA to the lumen. Although the role of IgA in intestinal mucosa has been extensively studied, the cell types responsible for secreting the IgA that protects the host against pathogens in the lower respiratory tract are less clear. Here, using a mouse model of influenza virus infection, we demonstrate that intranasal, but not systemic, immunization induces local IgA secretion in the bronchoalveolar space. Using single-cell RNA sequencing, we found a heterogeneous population of IgA-expressing cells within the respiratory mucosa, including tissue-resident memory B cells, plasmablasts, and plasma cells. IgA-secreting cell establishment within the lung required CXCR3. An intranasally administered protein-based vaccine also led to the establishment of IgA-secreting cells in the lung, but not when given intramuscularly or intraperitoneally. Last, local IgA secretion correlated with superior protection against secondary challenge with homologous and heterologous virus infection than circulating antibodies alone. These results provide key insights into establishment of protective immunity in the lung based on tissue-resident IgA-secreting B cells and inform vaccine strategies designed to elicit highly effective immune protection against respiratory virus infections.


Subject(s)
Antiviral Agents/immunology , B-Lymphocytes/immunology , Immunity, Mucosal/immunology , Immunoglobulin A, Secretory/immunology , Influenza Vaccines/immunology , Lung/immunology , Administration, Intranasal , Animals , Antiviral Agents/administration & dosage , Female , Immunoglobulin A, Secretory/administration & dosage , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Lung/virology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout
5.
Cell Rep Med ; 2(12): 100468, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34873588

ABSTRACT

In view of the scarcity of data to guide decision making, we evaluated how BNT162b2 and mRNA-1273 vaccines affect the immune response in lactating women and the protective profile of breastmilk. Compared with controls, lactating women had a higher frequency of circulating RBD memory B cells and higher anti-RBD antibody titers but similar neutralizing capacity. We show that upon vaccination, immune transfer to breastmilk occurs through a combination of anti-spike secretory IgA (SIgA) antibodies and spike-reactive T cells. Although we found that the concentration of anti-spike IgA in breastmilk might not be sufficient to directly neutralize SARS-CoV-2, our data suggest that cumulative transfer of IgA might provide the infant with effective neutralization capacity. Our findings put forward the possibility that breastmilk might convey both immediate (through anti-spike SIgA) and long-lived (via spike-reactive T cells) immune protection to the infant. Further studies are needed to address this possibility and to determine the functional profile of spike T cells.


Subject(s)
COVID-19 Vaccines/immunology , Immunoglobulin A, Secretory/immunology , Milk, Human/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocytes/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , Female , Humans , Immunity, Maternally-Acquired , Lactation/immunology , Memory B Cells/immunology , Vaccination , mRNA Vaccines/immunology
6.
Front Immunol ; 12: 697725, 2021.
Article in English | MEDLINE | ID: mdl-34804004

ABSTRACT

The intestinal mucosal immune environment requires multiple immune cells to maintain homeostasis. Although intestinal B cells are among the most important immune cells, little is known about the mechanism that they employ to regulate immune homeostasis. In this study, we found that CD11b+ B cells significantly accumulated in the gut lamina propria and Peyer's patches in dextran sulfate sodium-induced colitis mouse models and patients with ulcerative colitis. Adoptive transfer of CD11b+ B cells, but not CD11b-/- B cells, effectively ameliorated colitis and exhibited therapeutic effects. Furthermore, CD11b+ B cells were found to produce higher levels of IgA than CD11b- B cells. CD11b deficiency in B cells dampened IgA production, resulting in the loss of their ability to ameliorate colitis. Mechanistically, CD11b+ B cells expressed abundant TGF-ß and TGF-ß receptor II, as well as highly activate phosphorylated Smad2/3 signaling pathway, consequently promoting the class switch to IgA. Collectively, our findings demonstrate that CD11b+ B cells are essential intestinal suppressive immune cells and the primary source of intestinal IgA, which plays an indispensable role in maintaining intestinal homeostasis.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/immunology , Colitis, Ulcerative/immunology , Colitis/immunology , Immunoglobulin A, Secretory/immunology , Peyer's Patches/immunology , Adoptive Transfer , Animals , B-Lymphocytes/pathology , CD11b Antigen/genetics , Colitis/chemically induced , Colitis/pathology , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Immunoglobulin Class Switching , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/pathology , Signal Transduction , Smad2 Protein/metabolism
7.
Nat Microbiol ; 6(12): 1493-1504, 2021 12.
Article in English | MEDLINE | ID: mdl-34811531

ABSTRACT

Secretory immunoglobulin A (sIgA) plays an important role in gut barrier protection by shaping the resident microbiota community, restricting the growth of bacterial pathogens and enhancing host protective immunity via immunological exclusion. Here, we found that a portion of the microbiota-driven sIgA response is induced by and directed towards intestinal fungi. Analysis of the human gut mycobiota bound by sIgA revealed a preference for hyphae, a fungal morphotype associated with virulence. Candida albicans was a potent inducer of IgA class-switch recombination among plasma cells, via an interaction dependent on intestinal phagocytes and hyphal programming. Characterization of sIgA affinity and polyreactivity showed that hyphae-associated virulence factors were bound by these antibodies and that sIgA influenced C. albicans morphotypes in the murine gut. Furthermore, an increase in granular hyphal morphologies in patients with Crohn's disease compared with healthy controls correlated with a decrease in antifungal sIgA antibody titre with affinity to two hyphae-associated virulence factors. Thus, in addition to its importance in gut bacterial regulation, sIgA targets the uniquely fungal phenomenon of hyphal formation. Our findings indicate that antifungal sIgA produced in the gut can play a role in regulating intestinal fungal commensalism by coating fungal morphotypes linked to virulence, thereby providing a protective mechanism that might be dysregulated in patients with Crohn's disease.


Subject(s)
Crohn Disease/microbiology , Fungi/physiology , Gastrointestinal Microbiome , Immunoglobulin A, Secretory/immunology , Symbiosis , Animals , Candida albicans/genetics , Candida albicans/physiology , Crohn Disease/genetics , Crohn Disease/immunology , Female , Fungi/genetics , Host-Pathogen Interactions , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Phagocytes/immunology , Phagocytes/microbiology
8.
Nature ; 598(7882): 657-661, 2021 10.
Article in English | MEDLINE | ID: mdl-34646015

ABSTRACT

Dimeric IgA secreted across mucous membranes in response to nonpathogenic taxa of the microbiota accounts for most antibody production in mammals. Diverse binding specificities can be detected within the polyclonal mucosal IgA antibody response1-10, but limited monoclonal hybridomas have been studied to relate antigen specificity or polyreactive binding to functional effects on microbial physiology in vivo11-17. Here we use recombinant dimeric monoclonal IgAs (mIgAs) to finely map the intestinal plasma cell response to microbial colonization with a single microorganism in mice. We identify a range of antigen-specific mIgA molecules targeting defined surface and nonsurface membrane antigens. Secretion of individual dimeric mIgAs targeting different antigens in vivo showed distinct alterations in the function and metabolism of intestinal bacteria, largely through specific binding. Even in cases in which the same microbial antigen is targeted, microbial metabolic alterations differed depending on IgA epitope specificity. By contrast, bacterial surface coating generally reduced motility and limited bile acid toxicity. The overall intestinal IgA response to a single microbe therefore contains parallel components with distinct effects on microbial carbon-source uptake, bacteriophage susceptibility, motility and membrane integrity.


Subject(s)
Immunoglobulin A, Secretory/immunology , Intestines/immunology , Microbiota/immunology , Plasma Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Escherichia coli , Germ-Free Life , Mice , Mice, Inbred C57BL , Porins/immunology
9.
Cell Rep ; 36(10): 109655, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34496253

ABSTRACT

The evolutionary strategy of transferring maternal antibodies via milk profoundly impacts the survival, lifelong health, and wellbeing of all neonates, including a pronounced impact on human breastfeeding success and infant development. While there has been increased recognition that interorgan connectivity influences the quality of a mother's milk, potentially to personalize it for her offspring, the underlying bases for these processes are incompletely resolved. Here, we define an essential role of Peyer's patches (PPs) for the generation of plasma cells that secrete maternal immunoglobulin A (IgA) into milk. Our metagenomic analysis reveals that the presence of certain residential microorganisms in the gastrointestinal (GI) tract, such as Bacteroides acidifaciens and Prevotella buccalis, is indispensable for the programming of maternal IgA synthesis prior to lactational transfer. Our data provide important insights into how the microbiome of the maternal GI environment, specifically through PPs, can be communicated to the next generation via milk.


Subject(s)
Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Milk, Human/immunology , Plasma Cells/cytology , Animals , Humans , Immunoglobulin A/immunology , Immunoglobulin A, Secretory/immunology , Mice , Peyer's Patches/immunology
10.
Mucosal Immunol ; 14(6): 1347-1357, 2021 11.
Article in English | MEDLINE | ID: mdl-34417548

ABSTRACT

Efficient IgA transcytosis is critical for the maintenance of a homeostatic microbiota. In the canonical model, locally-secreted dimeric (d)IgA reaches the polymeric immunoglobulin receptor (pIgR) on intestinal epithelium via simple diffusion. A role for integrin αE(CD103)ß7 during transcytosis has not been described, nor its expression by intestinal B cell lineage cells. We found that αE-deficient (αE-/-) mice have a luminal IgA deficit, despite normal antibody-secreting cells (ASC) recruitment, local IgA production and increased pIgR expression. This deficit was not due to dendritic cell (DC)-derived retinoic acid (RA) nor class-switching defects, as stool from RAG-/- mice reconstituted with αE-/- B cells was also IgA deficient. Flow cytometric, ultrastructural and transcriptional profiling showed that αEß7-expressing ASC represent an undescribed subset of terminally-differentiated intestinal plasma cells (PC) that establishes direct cell to cell contact with intestinal epithelium. We propose that IgA not only reaches pIgR through diffusion, but that αEß7+ PC dock with E-cadherin-expressing intestinal epithelium to directly relay IgA for transcytosis into the intestinal lumen.


Subject(s)
Immunoglobulin A/immunology , Integrins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Transcytosis/immunology , Animals , Cell Differentiation/immunology , Gene Expression , Gene Expression Regulation , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/immunology , Integrins/deficiency , Integrins/metabolism , Intestinal Mucosa/ultrastructure , Lymphocyte Activation , Mice , Mice, Knockout , Models, Biological , Plasma Cells/cytology , Plasma Cells/ultrastructure
12.
Eur J Immunol ; 51(11): 2641-2650, 2021 11.
Article in English | MEDLINE | ID: mdl-34398472

ABSTRACT

Helicobacter pylori is a Gram-negative bacterium found on the luminal surface of the gastric mucosa in at least 50% of the world's human population. The protective effect of breastfeeding against H. pylori infection has been extensively reported; however, the mechanisms behind this protection remain poorly understood. Human IgA from colostrum has reactivity against H. pylori antigens. Despite that IgA1 and IgA2 display structural and functional differences, their reactivity against H. pylori had not been previously determined. We attested titers and reactivity of human colostrum-IgA subclasses by ELISA, immunoblot, and flow cytometry. Colostrum samples from healthy mothers had higher titers of IgA; and IgA1 mostly recognized H. pylori antigens. Moreover, we found a correlation between IgA1 reactivity and their neutralizing effect determined by inhibition of cytoskeletal changes in AGS cells infected with H. pylori. In conclusion, colostrum-IgA reduces H. pylori infection of epithelial gastric cells, suggesting an important role in preventing the bacteria establishment during the first months of life. As a whole, these results suggest that IgA1 from human colostrum provides protection that may help in the development of the mucosal immune system of newborn children.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Colostrum/immunology , Helicobacter pylori/immunology , Immunoglobulin A, Secretory/immunology , Cytoskeleton , Epithelial Cells , Female , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Humans , Pregnancy
13.
Viruses ; 13(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-34062811

ABSTRACT

It is well known that approximately 50% of cattle infected with foot-and-mouth disease (FMD) virus (FMDV) may become asymptomatic carrier (persistently infected) animals. Although transmission of FMDV from carrier cattle to naïve cattle has not been demonstrated experimentally, circumstantial evidence from field studies has linked FMDV-carrier cattle to cause subsequent outbreaks. Therefore, the asymptomatic carrier state complicates the control and eradication of FMD. Current serological diagnosis using tests for antibodies to the viral non-structural proteins (NSP-ELISA) are not sensitive enough to detect all carrier animals, if persistently infected after vaccination and do not distinguish between carriers and non-carriers. The specificity of the NSP ELISA may also be reduced after vaccination, in particular after multiple vaccination. FMDV-specific mucosal antibodies (IgA) are not produced in vaccinated cattle but are elevated transiently during the acute phase of infection and can be detected at a high level in cattle persistently infected with FMDV, irrespective of their vaccination status. Therefore, detection of IgA by ELISA may be considered a diagnostic alternative to RT-PCR for assessing FMDV persistent infection in ruminants in both vaccinated and unvaccinated infected populations. This study reports on the development and validation of a new mucosal IgA ELISA for the detection of carrier animals using nasal, saliva, and oro-pharyngeal fluid (OPF) samples. The diagnostic performance of the IgA ELISA using nasal samples from experimentally vaccinated and infected cattle demonstrated a high level of specificity (99%) and an improved level of sensitivity (76.5%). Furthermore, the detection of carrier animals reached 96.9% when parallel testing of samples was carried out using both the IgA-ELISA and NSP-ELISA.


Subject(s)
Cattle Diseases/immunology , Cattle Diseases/virology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/virology , Immunoglobulin A, Secretory/immunology , Mucous Membrane/immunology , Animals , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/epidemiology , Disease Outbreaks , Enzyme-Linked Immunosorbent Assay , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/epidemiology , Mucous Membrane/metabolism , ROC Curve , Vaccines/immunology
14.
Sci Rep ; 11(1): 12939, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34155266

ABSTRACT

We previously reported that dietary supplementation with cholic acid (CA), the primary 12α-hydroxylated (12αOH) bile acid (BA), reduces plasma adiponectin concentration in rats. The aim of this study was to examine the distribution of adiponectin in the body of CA-fed rats and its influence on mucosal immunoglobulin A concentration in the intestine. Rats were fed a diet supplemented with or without CA (0.5 g CA/kg diet) for 13 weeks. A reduction in plasma adiponectin level was observed from week 3. At the end of the experiment, the CA diet reduced plasma adiponectin concentration both in the portal and aortic plasma. Accumulation of adiponectin was accompanied by an increase in cadherin-13 mRNA expression in the ileal mucosa of CA-fed rats. No increase was observed in adiponectin mRNA expression in the ileal and adipose tissues of the CA-fed rats. Immunoglobulin A concentration in the ileal mucosa was elevated in the CA-fed rats and was correlated with the ileal adiponectin concentration. 12αOH BAs may modulate mucosal immune response that are involved in the accumulation of adiponectin in the ileum.


Subject(s)
Adiponectin/biosynthesis , Bile Acids and Salts/metabolism , Ileum/immunology , Ileum/metabolism , Immunoglobulin A, Secretory/immunology , Animal Feed , Animals , Biomarkers , Feces/chemistry , Male , Rats
15.
Mucosal Immunol ; 14(5): 1067-1076, 2021 09.
Article in English | MEDLINE | ID: mdl-34108594

ABSTRACT

The upper respiratory tract is highly exposed to airborne pathogens and serves as an important inductive site for protective antibody responses, including mucosal IgA and systemic IgG. However, it is currently unknown to what extent inhaled environmental toxins, such as a cigarette smoke, affect the ability to induce antibody-mediated immunity at this site. Using a murine model of intranasal lipopolysaccharide and ovalbumin (LPS/OVA) immunization, we show that cigarette smoke exposure compromises the induction of antigen-specific IgA in the upper airways and systemic circulation. Deficits in OVA-IgA were observed in conjunction with a reduced accumulation of OVA-specific IgA antibody-secreting cells (ASCs) in the nasal mucosa, inductive tissues (NALT, cervical lymph nodes, spleen) and the blood. Nasal OVA-IgA from smoke-exposed mice also demonstrated reduced avidity during the acute post-immunization period in association with an enhanced mutational burden in the cognate nasal Igha repertoire. Mechanistically, smoke exposure attenuated the ability of the nasal mucosa to upregulate VCAM-1 and pIgR, suggesting that cigarette smoke may inhibit both nasal ASC homing and IgA transepithelial transport. Overall, these findings demonstrate the immunosuppressive nature of tobacco smoke and illustrate the diversity of mechanisms through which this noxious stimulus can interfere with IgA-mediated immunity in the upper airways.


Subject(s)
Antibody Formation/immunology , Antigens/immunology , Immunity, Mucosal , Immunoglobulin A, Secretory/immunology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Tobacco Smoking/adverse effects , Animals , Biomarkers , Chemokines, CC/metabolism , Immunization , Immunophenotyping , Lipopolysaccharides/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Ovalbumin/immunology , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/immunology , Somatic Hypermutation, Immunoglobulin , Vascular Cell Adhesion Molecule-1/metabolism
16.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 707-718, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33963824

ABSTRACT

The major etiologic agent that causes acute gastroenteritis worldwide in young animals and children is Group A rotavirus. Currently, commercially available vaccines do not often prevent porcine rotavirus (PRV) infection. In this study, we evaluated the efficacy of oral recombinant Lactobacillus vaccine against PRV in a mouse model. Lactobacillus plantarum NC8 was used as the host strain, and bacterial vectors were constructed, because the NC8 isolated has shown the capability to survive gastric transit and to colonize the intestinal tract of humans and other mammals. To explore the immunological mechanisms, lactic acid bacterial vectors were used to express VP7 antigen from PRV. We constructed an L. plantarum strain with surface-displayed VP7, named NC8-pSIP409-pgsA-VP7-DCpep. The expressed recombinant protein had a molecular weight of ∼37 kDa. The strain was used to immunize BALB/c mice to evaluate their immunomodulatory characteristics. Mice were orally immunized with recombinant L. plantarum NC8-pSIP409-pgsA-VP7-DCpep at a dose of 2 × 109 colony forming units/200 µl. The results showed that NC8-pSIP409-pgsA-VP7-DCpep significantly stimulated the differentiation of dendritic cells (DCs) in Peyer's patches (PPs) and increased the serum levels of IL-4 and IFN-γ, as measured by enzyme-linked immunosorbent assay in mice treated with NC8-pSIP409-pgsA-VP7-DCpep. Compared to the empty vector group, NC8-pSIP409-pgsA-VP7-DCpep significantly increased the production of B220+ B cells in mesenteric lymph nodes (MLNs) and PPs and also increased the titer levels of the VP7-specific antibodies, including IgG and sIgA. The administration of NC8-pSIP409-pgsA-VP7-DCpep mediated relatively broad cellular responses. This study reveals that clear alternatives exist for PRV control strategies and provides information on PRV infection.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Engineering/methods , Immunization/methods , Immunogenicity, Vaccine , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Vaccines, Synthetic/administration & dosage , Animals , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Antigens, Heterophile/metabolism , Antigens, Viral/metabolism , B-Lymphocytes/immunology , Capsid Proteins/metabolism , Cytokines/blood , Female , Genes, Viral , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Rotavirus/immunology , Rotavirus/metabolism , Swine , Vaccines, Synthetic/immunology
17.
Nutrients ; 13(4)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920202

ABSTRACT

Consumption of indigestible dietary fiber increases immunoglobulin A (IgA) levels in saliva. The purpose of this study is to clarify the synergistic effect of the intake of a high amount of fats and indigestible dietary fiber on IgA levels in saliva and submandibular glands (SMG). Seven-week-old Wistar rats were fed a low-fat (60 g/kg) fiberless diet, low-fat fructo-oligosaccharide (FOS, 30 g/kg) diet, high-fat (220 g/kg) fiberless diet, or high-fat FOS diet for 70 days. The IgA flow rate of saliva (IgA FR-saliva) was higher in the low-fat FOS group than in the other groups (p < 0.05). Furthermore, the concentration of tyrosine hydroxylase (a marker of sympathetic nerve activation) in the SMG was higher in the low-fat FOS group (p < 0.05) and positively correlated with the IgA FR-saliva (rs = 0.68. p < 0.0001. n = 32) in comparison to that in the other groups. These findings suggest that during low-fat FOS intake, salivary IgA levels may increase through sympathetic nerve activation.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Fiber/administration & dosage , Immunoglobulin A, Secretory/analysis , Oligosaccharides/administration & dosage , Respiratory Tract Infections/prevention & control , Animal Feed , Animals , Humans , Immunoglobulin A, Secretory/immunology , Male , Models, Animal , Rats , Rats, Wistar , Respiratory Tract Infections/immunology , Saliva/chemistry , Saliva/immunology , Submandibular Gland/chemistry , Submandibular Gland/immunology , Submandibular Gland/innervation , Submandibular Gland/metabolism , Sympathetic Nervous System/immunology , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/metabolism
18.
Front Immunol ; 12: 657679, 2021.
Article in English | MEDLINE | ID: mdl-33815419

ABSTRACT

Human Immunodeficiency Virus (HIV) and Simian Immunodeficiency Virus (SIV) are associated with severe perturbations in the gut mucosal environment characterized by massive viral replication and depletion of CD4 T cells leading to dysbiosis, breakdown of the epithelial barrier, microbial translocation, immune activation and disease progression. Multiple mechanisms play a role in maintaining homeostasis in the gut mucosa and protecting the integrity of the epithelial barrier. Among these are the secretory IgA (sIgA) that are produced daily in vast quantities throughout the mucosa and play a pivotal role in preventing commensal microbes from breaching the epithelial barrier. These microbe specific, high affinity IgA are produced by IgA+ plasma cells that are present within the Peyer's Patches, mesenteric lymph nodes and the isolated lymphoid follicles that are prevalent in the lamina propria of the gastrointestinal tract (GIT). Differentiation, maturation and class switching to IgA producing plasma cells requires help from T follicular helper (Tfh) cells that are present within these lymphoid tissues. HIV replication and CD4 T cell depletion is accompanied by severe dysregulation of Tfh cell responses that compromises the generation of mucosal IgA that in turn alters barrier integrity leading to commensal bacteria readily breaching the epithelial barrier and causing mucosal pathology. Here we review the effect of HIV infection on Tfh cells and mucosal IgA responses in the GIT and the consequences these have for gut dysbiosis and mucosal immunopathogenesis.


Subject(s)
Gastrointestinal Microbiome/immunology , HIV Infections/immunology , HIV/immunology , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Immunoglobulin A/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Communication/immunology , HIV Infections/metabolism , HIV Infections/virology , Humans , Immunity, Mucosal , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Plasma Cells/immunology , Plasma Cells/metabolism , Signal Transduction , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Helper-Inducer/metabolism
19.
Molecules ; 26(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808410

ABSTRACT

Background: The study examined the oral microbiota, physiological and immunological changes in patients using thermoplastic retainers during three months of use. Methods: The study included several steps. Firstly, 10 swabs were collected from the buccal and palatal surfaces of the teeth of the patients, approximately 2 mL of saliva was collected from the same patients and 2 mL of saliva was collected from 10 healthy people to measure the pH and secretory IgA level. This was followed by the isolation and identfication of the bacterial isolates in the patient samples. Then, isolate susceptibility toward chlorhexidine (CHX) and their adhesion ability to thermoplastic retainer surfaces was measured. In addition to that the study estimated the numbers of Lactobacillus and Streptooccus mutans colonies during three months and finally, a comparsion of pH acidity and IgA level between the patients and healthy people was performed. The results showed the predominant bacteria during the three months were Lactobacillus spp. and Streptococcus spp. followed by different rates of other bacteria. Raoultella ornithinolytica showed more resistance to CHX while Lactobacillus spp. showed more sensitivity. Streptococcus mutans colony levels were higher than Lactobacillus spp. colonies during the three months, also S. mutans had the highest value in adherence to retainer thermoplastic. Finally, pH acidity showed a highly significant difference (p ≤ 0.05) in the third month, like IgA levels (p ≤ 0.05). Conclusions: According to the results obtained from the current study, the researchers noted that the thermoplastic retainers helped change the oral cavity environment.


Subject(s)
Hydrogen-Ion Concentration , Immunoglobulin A, Secretory/immunology , Microbiota , Mouth , Orthodontic Retainers , Saliva , Humans , Lactobacillus/isolation & purification , Mouth/immunology , Mouth/microbiology , Saliva/immunology , Saliva/microbiology , Streptococcus mutans/isolation & purification
20.
Res Vet Sci ; 136: 310-317, 2021 May.
Article in English | MEDLINE | ID: mdl-33756379

ABSTRACT

Oral probiotics are used to induce immune responses in the intestines to protect against infection. However, oral probiotics may also affect immune responses in other mucosal tissues such as in the respiratory tract. To examine this possibility, we explored the potential of immunocytes to home to the respiratory system after oral administration of Bacillus subtilis. The results showed that B. subtilis could promote intestinal development and not cause pathological changes in the respiratory tract. Following the oral administration with B. subtilis, the number of IgA-secreting cells and CD3+ T cells not only significantly increased in the intestinal tracts but also in respiratory tracts (P < 0.01). Moreover, the levels of secretory IgA were significantly higher in the trachea, lungs, ileum, and jejunum after oral B. subtilis administration than in the control groups (P < 0.05). The mRNA expression of interleukin (IL)-1ß, IL-5, IL-6, tumor necrosis factor-α, B cell activating factor, and IgA-inducing protein increased following B. subtilis administration (P < 0.01) in the trachea, lungs, ileum, and jejunum. These data suggest that B. subtilis administration regulates the immune response not only in the intestine but also in the respiratory tract of piglets. Our work highlights a potentially new strategy for promoting respiratory mucosal immunity and may contribute to the design of vaccines with B. subtilis as a mucosal adjuvant.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Vaccines/immunology , CD3 Complex/immunology , Immunity, Mucosal , Respiratory System/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Animals, Newborn , Immunoglobulin A, Secretory/immunology , Male , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...