Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Microorganisms ; 11(10)2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37894182

ABSTRACT

A sero-epidemiology study was conducted in Dhaka, Bangladesh between January 2020 and February 2021 to assess the immune responses to ETEC infection in adults and children. (1) Background: Enterotoxigenic Escherichia coli infection is a main cause of diarrheal disease in endemic countries. The characterization of the immune responses evoked by natural infection can guide vaccine development efforts. (2) Methods: A total of 617 adult and 480 pediatric diarrheal patients were screened, and 43 adults and 46 children (below 5 years of age) with an acute ETEC infection completed the study. The plasma samples were analyzed for antibody responses against the ETEC toxins. (3) Results: Heat-stable toxin (ST)-positive ETEC is the main cause of ETEC infection in adults, unlike in children in an endemic setting. We detected very low levels of anti-ST antibodies, and no ST-neutralizing activity. However, infection with ETEC strains expressing the heat-labile toxin (LT) induced systemic antibody responses in less than 25% of subjects. The antibody levels against LTA and LTB, as well as cholera toxin (CT), correlated well. The anti-LT antibodies were shown to have LT- and CT- neutralizing activity. The antibody reactivity against linear LT epitopes did not correlate with toxin-neutralizing activity. (4) Conclusions: Unlike LT, ST is a poor antigen and even adults have low anti-ST antibody levels that do not allow for the detection of toxin-neutralizing activity.

2.
Front Immunol ; 14: 1166924, 2023.
Article in English | MEDLINE | ID: mdl-37251407

ABSTRACT

Introduction: The COVID-19 pandemic illustrates the need for serology diagnostics with improved accuracy. While conventional serology based on recognition of entire proteins or subunits thereof has made significant contribution to the antibody assessment space, it often suffers from sub-optimal specificity. Epitope-based, high-precision, serology assays hold potential to capture the high specificity and diversity of the immune system, hence circumventing the cross-reactivity with closely related microbial antigens. Methods: We herein report mapping of linear IgG and IgA antibody epitopes of the SARS-CoV-2 Spike (S) protein in samples from SARS-CoV-2 exposed individuals along with certified SARS-CoV-2 verification plasma samples using peptide arrays. Results: We identified 21 distinct linear epitopes. Importantly, we showed that pre-pandemic serum samples contain IgG antibodies reacting to the majority of protein S epitopes, most likely as a result of prior infection with seasonal coronaviruses. Only 4 of the identified SARS-CoV-2 protein S linear epitopes were specific for SARS-CoV-2 infection. These epitopes are located at positions 278-298 and 550-586, just proximal and distal to the RBD, as well as at position 1134-1156 in the HR2 subdomain and at 1248-1271 in the C-terminal subdomain of protein S. To substantiate the applicability of our findings, we tested three of the high-accuracy protein S epitopes in a Luminex assay, using a certified validation plasma sample set from SARS-CoV-2 infected individuals. The Luminex results were well aligned with the peptide array results, and correlated very well with in-house and commercial immune assays for RBD, S1 and S1/S2 domains of protein S. Conclusion: We present a comprehensive mapping of linear B-cell epitopes of SARS-CoV-2 protein S, that identifies peptides suitable for a precision serology assay devoid of cross-reactivity. These results have implications for development of highly specific serology test for exposure to SARS-CoV-2 and other members of the coronaviridae family, as well as for rapid development of serology tests for future emerging pandemic threats.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2 , Epitopes, B-Lymphocyte , Protein S , Spike Glycoprotein, Coronavirus , Pandemics , Antibodies, Viral , Immunoglobulin G , COVID-19 Testing
3.
Infect Immun ; 85(10)2017 10.
Article in English | MEDLINE | ID: mdl-28694295

ABSTRACT

Emerging evidence shows that the human microbiota plays a larger role in disease progression and health than previously anticipated. Helicobacter pylori, the causative agent of gastric cancer and duodenal and gastric ulcers, was early associated with gastric disease, but it has also been proposed that the accompanying microbiota in Helicobacter pylori-infected individuals might affect disease progression and gastric cancer development. In this study, the composition of the transcriptionally active microbial community and H. pylori gene expression were determined using metatranscriptomic RNA sequencing of stomach biopsy specimens from individuals with different H. pylori infection statuses and premalignant tissue changes. The results show that H. pylori completely dominates the microbiota not only in infected individuals but also in most individuals classified as H. pylori uninfected using conventional methods. Furthermore, H. pylori abundance is positively correlated with the presence of Campylobacter, Deinococcus, and Sulfurospirillum Finally, we quantified the expression of a large number of Helicobacter pylori genes and found high expression of genes involved in pH regulation and nickel transport. Our study is the first to dissect the viable microbiota of the human stomach by metatranscriptomic analysis, and it shows that metatranscriptomic analysis of the gastric microbiota is feasible and can provide new insights into how bacteria respond in vivo to variations in the stomach microenvironment and at different stages of disease progression.


Subject(s)
Carcinogenesis , Gastrointestinal Microbiome , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Stomach Neoplasms/microbiology , Stomach/microbiology , Transcriptome , Adult , Aged , Bacteria/genetics , Bacteria/isolation & purification , Disease Progression , Female , Gastric Mucosa/microbiology , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Gene Expression Profiling , Helicobacter Infections/pathology , Humans , Male , Microbial Viability , Middle Aged , Stomach/pathology , Young Adult
4.
Cytokine ; 99: 30-34, 2017 11.
Article in English | MEDLINE | ID: mdl-28683359

ABSTRACT

BACKGROUND: Chronic infection with Helicobacter pylori leads to gastritis and in a subpopulation of infected individuals to ulcers and cancer. Bacterial virulence factors and host immune inflammatory responses are risk factors related to disease. CD4+ T cells secrete cytokines that promote inflammation and an anti-bacterial response in the gastric mucosa during infection. The aim of the study was to investigate the pattern of expression of CD4+ T cell derived cytokines, IL-17A and IFNγ in paired antrum and corpus biopsies and correlate it to H. pylori infection outcome. METHODS: Gene and protein expression of IL-17A and IFNγ was analyzed in gastric biopsies from H. pylori infected subjects with non-ulcer dyspepsia (NUD) or gastric ulcer; and for comparison uninfected individuals. RESULTS: Upregulation of IL-17A and IFNγ gene expression was seen in corpus and antrum biopsies of H. pylori infected individuals with NUD compared to in uninfected controls. The expression of these cytokines correlated significantly with each other. Immunofluorescence staining revealed increased frequencies of IL-17A+ and IFNγ+ cells in antrum biopsies of gastric ulcer patients compared to of H. pylori infected NUD individuals; positive cells were not detected in any of the biopsies of uninfected controls. The frequencies of IFNγ and IL-17A+ cells correlated positively with inflammation in the antrum, but not the corpus, of H. pylori infected individuals. In the antrum, while there was no significant evidence of correlation between IFNγ and bacterial score, a positive correlation between bacterial score and IL-17A+ cells was seen. CONCLUSIONS: In H. pylori infected individuals, the frequencies of IFNγ and IL-17A+ cells were increased in the antrum, particularly in patients with H. pylori induced gastric ulcers. Even though H. pylori colonized both the corpus and antrum regions of the stomach, the cytokine responses and subsequent pathology were mainly detected in the antrum.


Subject(s)
Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Aged , Biopsy , Female , Helicobacter Infections/blood , Humans , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-17/blood , Interleukin-17/genetics , Male , Middle Aged , Stomach/pathology , Stomach Ulcer/pathology , Up-Regulation/genetics
5.
Gastric Cancer ; 20(1): 116-125, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26782287

ABSTRACT

BACKGROUND: An increase of regulatory T cells, defined as CD25high- and/or FOXP3+-expressing CD4+ T cells, within tumors has been reported in several studies. Tregs promote tumor growth by modulating the antitumor immune response, mainly through inhibition of T-cell-mediated tumor cell killing: this has been suggested to be dependent on IL-10 and/or TGF-ß. In stomach cancer, the mechanisms behind the accumulation of Tregs in tumor tissue has not been fully elucidated, and neither has Treg gene expression in situ. MATERIALS AND METHODS: Stomach tissue from gastric cancer patients undergoing gastric resection was analyzed using flow cytometry and cell sorting, followed by RT-PCR. RESULTS: We observed that stomach CD4+ FOXP3+ T cells proliferated to a higher degree than CD4+ FOXP3- T cells, which may contribute to Treg accumulation in the mucosa. By analyzing DNA methylation, we demonstrated that both proliferating and nonproliferating FOXP3+ T cells exhibited complete demethylation of the FOXP3 gene, indicating a stable FOXP3 expression in both cell populations. Furthermore, analysis of T-cell populations isolated directly from the tumor and tumor-free mucosa demonstrated that CD4+ CD25high T cells have a higher IL-10/IFN-γ gene expression ratio but express lower levels of TGF-ß than CD4+ CD25low/- T cells. CONCLUSION: We demonstrate strong proliferation among regulatory CD4+ FOXP3+ CD25high T cells in the gastric cancer mucosa. These local Treg express a suppressive cytokine profile characterized by high IL-10 and low TGF-ß and IFN-γ production.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Gastric Mucosa/immunology , Interleukin-10/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/genetics , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , DNA Methylation , Female , Flow Cytometry , Follow-Up Studies , Forkhead Transcription Factors/metabolism , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Tumor Cells, Cultured
6.
BMC Evol Biol ; 16: 53, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26928576

ABSTRACT

BACKGROUND: Helicobacter pylori (H. pylori) is one of the most common bacterial infections in humans and this infection can lead to gastric ulcers and gastric cancer. H. pylori is one of the most genetically variable human pathogens and the ability of the bacterium to bind to the host epithelium as well as the presence of different virulence factors and genetic variants within these genes have been associated with disease severity. Nicaragua has particularly high gastric cancer incidence and we therefore studied Nicaraguan clinical H. pylori isolates for factors that could contribute to cancer risk. METHODS: The complete genomes of fifty-two Nicaraguan H. pylori isolates were sequenced and assembled de novo, and phylogenetic and virulence factor analyses were performed. RESULTS: The Nicaraguan isolates showed phylogenetic relationship with West African isolates in whole-genome sequence comparisons and with Western and urban South- and Central American isolates using MLSA (Multi-locus sequence analysis). A majority, 77 % of the isolates carried the cancer-associated virulence gene cagA and also the s1/i1/m1 vacuolating cytotoxin, vacA allele combination, which is linked to increased severity of disease. Specifically, we also found that Nicaraguan isolates have a blood group-binding adhesin (BabA) variant highly similar to previously reported BabA sequences from Latin America, including from isolates belonging to other phylogenetic groups. These BabA sequences were found to be under positive selection at several amino acid positions that differed from the global collection of isolates. CONCLUSION: The discovery of a Latin American BabA variant, independent of overall phylogenetic background, suggests hitherto unknown host or environmental factors within the Latin American population giving H. pylori isolates carrying this adhesin variant a selective advantage, which could affect pathogenesis and risk for sequelae through specific adherence properties.


Subject(s)
Adhesins, Bacterial/genetics , Helicobacter pylori/classification , Helicobacter pylori/genetics , Adhesins, Bacterial/chemistry , Adolescent , Adult , Aged , Amino Acid Sequence , Antigens, Bacterial/genetics , Female , Genetic Variation , Genome, Bacterial , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Middle Aged , Molecular Sequence Data , Multilocus Sequence Typing , Nicaragua , Phylogeny , Virulence Factors/chemistry , Virulence Factors/genetics , Young Adult
7.
PLoS One ; 9(4): e93943, 2014.
Article in English | MEDLINE | ID: mdl-24714675

ABSTRACT

Both Th1 and Th17 cells are important components of the immune response to Helicobacter pylori (Hp) in adults, but less is known about T cell responses to Hp during early childhood, when the infection is often acquired. We investigated Th1 and Th17 type responses to Hp in adults, children and infants in Bangladesh, where Hp is highly endemic. IL-17 and IFN-γ mRNA levels in gastric biopsies from Hp-infected Bangladeshi adults were analyzed and compared to levels in infected and uninfected Swedish controls. Since biopsies could not be collected from infants and children, cytokine responses in Bangladeshi infants (6-12 months), children (3-5 years) and adults (>19 years) were instead compared by stimulating peripheral blood mononuclear cells (PBMCs) with a Hp membrane preparation (MP) and analyzing culture supernatants by ELISA and cytometric bead array. We found significantly higher expression of IL-17 and IFN-γ mRNA in gastric mucosa of Hp-infected Bangladeshi and Swedish adults compared to uninfected Swedish controls. PBMCs from all age groups produced IL-17 and IFN-γ after MP stimulation, but little Th2 cytokines. IL-17 and IFN-γ were primarily produced by CD4+ T cells, since CD4+ T cell depleted PBMCs produced reduced amounts of these cytokines. Infant cells produced significantly more IL-17, but similar levels of IFN-γ, compared to adult cells after MP stimulation. In contrast, polyclonal stimulation induced lower levels IL-17 and IFN-γ in infant compared to adult PBMCs and CD4+ T cells. The strong IL-17 production in infants after MP stimulation was paralleled by significantly higher production of the IL-17 promoting cytokine IL-1ß from infant compared to adult PBMCs and monocytes. In conclusion, these results show that T cells can produce high levels of IL-17 and IFN-γ in response to Hp from an early age and indicate a potential role for IL-1ß in promoting Th17 responses to Hp during infancy.


Subject(s)
Helicobacter Infections/immunology , Helicobacter pylori/immunology , Th1 Cells/metabolism , Th17 Cells/metabolism , Adult , Aged , Aged, 80 and over , Bangladesh , Child, Preschool , Female , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Humans , Infant , Interferon-gamma/metabolism , Interleukin-17/metabolism , Male , Middle Aged , Sweden , Th1 Cells/pathology , Th17 Cells/pathology , Young Adult
8.
BMC Med Genomics ; 6: 41, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24119614

ABSTRACT

BACKGROUND: The majority of gastric cancer cases are believed to be caused by chronic infection with the bacterium Helicobacter pylori, and atrophic corpus gastritis is a predisposing condition to gastric cancer development. We aimed to increase understanding of the molecular details of atrophy by performing a global transcriptome analysis of stomach tissue. METHODS: Biopsies from patients with different stages of H. pylori infection were taken from both the antrum and corpus mucosa and analyzed on microarrays. The stages included patients without current H. pylori infection, H. pylori-infected without corpus atrophy and patients with current or past H. pylori-infection with corpus-predominant atrophic gastritis. RESULTS: Using clustering and integrated analysis, we found firm evidence for antralization of the corpus mucosa of atrophy patients. This antralization harbored gain of gastrin expression, as well as loss of expression of corpus-related genes, such as genes associated with acid production, energy metabolism and blood clotting. The analyses provided detailed molecular evidence for simultaneous intestinal metaplasia (IM) and spasmolytic polypeptide expressing metaplasia (SPEM) in atrophic corpus tissue. Finally, acidic mammalian chitinase, a chitin-degrading enzyme produced by chief cells, was shown to be strongly down-regulated in corpus atrophy. CONCLUSIONS: Transcriptome analysis revealed several gene groups which are related to development of corpus atrophy, some of which were increased also in H. pylori-infected non-atrophic patients. Furthermore, loss of acidic chitinase expression is a promising marker for corpus atrophy.


Subject(s)
Chitinases/genetics , Gastric Mucosa/microbiology , Gastritis, Atrophic/enzymology , Gastritis, Atrophic/genetics , Helicobacter pylori/physiology , Transcriptome , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Blood Vessels/physiopathology , Chitinases/deficiency , Energy Metabolism/genetics , Female , Gastric Mucosa/blood supply , Gastric Mucosa/metabolism , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/physiopathology , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
9.
PLoS One ; 8(5): e65124, 2013.
Article in English | MEDLINE | ID: mdl-23741469

ABSTRACT

This study aimed to clarify whether Gram-positive (G+) and Gram-negative (G-) bacteria affect antigen-presenting cells differently and thereby influence the immunogenicity of proteins they express. Lactobacilli, lactococci and Escherichia coli strains were transformed with plasmids conferring intracellular ovalbumin (OVA) production. Murine splenic antigen presenting cells (APCs) were pulsed with washed and UV-inactivated OVA-producing bacteria, control bacteria, or soluble OVA. The ability of the APCs to activate OVA-specific DO11.10 CD4(+) T cells was assessed by measurments of T cell proliferation and cytokine (IFN-γ, IL-13, IL-17, IL-10) production. OVA expressed within E. coli was strongly immunogenic, since 500 times higher concentrations of soluble OVA were needed to achieve a similar level of OVA-specific T cell proliferation. Furthermore, T cells responding to soluble OVA produced mainly IL-13, while T cells responding to E. coli-expressed OVA produced high levels of both IFN-γ and IL-13. Compared to E. coli, G+ lactobacilli and lactococci were poor inducers of OVA-specific T cell proliferation and cytokine production, despite efficient intracellular expression and production of OVA and despite being efficiently phagocytosed. These results demonstrate a pronounced difference in immunogenicity of intracellular antigens in G+ and G- bacteria and may be relevant for the use of bacterial carriers in vaccine development.


Subject(s)
Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Ovalbumin/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens/biosynthesis , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Escherichia coli/immunology , Escherichia coli/metabolism , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Intracellular Space/metabolism , Lactobacillus/immunology , Lactobacillus/metabolism , Lactococcus/immunology , Lactococcus/metabolism , Lymphocyte Activation/immunology , Mice , Ovalbumin/metabolism , Phagocytosis/immunology , Spleen/immunology , Spleen/microbiology
10.
Physiol Genomics ; 45(15): 667-83, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23715263

ABSTRACT

In a screen for genes expressed specifically in gastric mucous neck cells, we identified GKN3, the recently discovered third member of the gastrokine family. We present confirmatory mouse data and novel porcine data showing that mouse GKN3 expression is confined to mucous cells of the corpus neck and antrum base and is prominently expressed in metaplastic lesions. GKN3 was proposed originally to be expressed in some human populations and a pseudogene in others. To investigate that hypothesis, we studied human GKN3 evolution in the context of its paralogous genomic neighbors, GKN1 and GKN2. Haplotype analysis revealed that GKN3 mimics GKN2 in patterns of exonic SNP allocation, whereas GKN1 appeared to be more stringently selected. GKN3 showed signatures of both directional selection and population based selective sweeps in humans. One such selective sweep includes SNP rs10187256, originally identified as an ancestral tryptophan to premature STOP codon mutation. The derived (nonancestral) allele went to fixation in Asia. We show that another SNP, rs75578132, identified 5 bp downstream of rs10187256, exhibits a second selective sweep in almost all Europeans, some Latinos, and some Africans, possibly resulting from a reintroduction of European genes during African colonization. Finally, we identify a mutation that would destroy the splice donor site in the putative exon3-intron3 boundary, which occurs in all human genomes examined to date. Our results highlight a stomach-specific human genetic locus, which has undergone various selective sweeps across European, Asian, and African populations and thus reflects geographic and ethnic patterns in genome evolution.


Subject(s)
Carrier Proteins/genetics , Evolution, Molecular , Genetic Loci/genetics , Membrane Proteins/genetics , Pseudogenes/genetics , Racial Groups/genetics , Selection, Genetic/genetics , Animals , Carrier Proteins/metabolism , Computational Biology , DNA Primers/genetics , Fluorescent Antibody Technique , Gastric Mucosa/metabolism , Genetics, Population , Genotype , Haplotypes/genetics , Humans , Likelihood Functions , Macaca mulatta/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL/genetics , Microarray Analysis , Microscopy, Confocal , Models, Genetic , Mutation/genetics , Phylogeny , Polymorphism, Single Nucleotide/genetics , Species Specificity , Sus scrofa/genetics
11.
Helicobacter ; 18(1): 73-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23067268

ABSTRACT

BACKGROUND: It has previously been reported that weak serum IgG but elevated IgA antibody responses against H. pylori may be associated with risk of gastric cancer (GC) development. To search for potential immunologic markers for GC, we analyzed antibody responses against H. pylori in risk groups of cancer development. MATERIAL AND METHODS: Sera and stomach biopsies collected from H. pylori-infected GC patients as well as from patients with gastric ulcer (GU), atrophic gastritis, intestinal metaplasia (IM) and duodenal ulcer and from H. pylori-infected control subjects without atrophy or IM, and in addition from H. pylori-negative subjects were analyzed for IgG and IgA antibodies against three different H. pylori antigen preparations, that is, membrane protein (MP), urease, and CagA. RESULTS: We observed an increased serum IgA/IgG titer ratio against H. pylori anti-MP in GC and GU patients, and against CagA in Hp-infected GC patients and risk groups. Female patients with GC had a higher serum anti-MP IgA/IgG titer ratio and a higher proportion of poorly differentiated cancer compared with male patients. As earlier observed, the non-tumorous mucosa of H. pylori-infected GC patients contained considerably lower levels of total IgA and H. pylori-specific IgA compared with H. pylori-infected controls. Similarly, we observed decreased specific mucosal anti-MP IgA response in patients with IM. CONCLUSION: We observed several differences in local and systemic immunologic responses against H. pylori in H. pylori-infected GC patients and putative GC risk group patients compared with H. pylori-infected controls. These findings may be of importance in efforts to identify risk groups of GC or early stages of GC.


Subject(s)
Antibodies, Bacterial/analysis , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Stomach Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Female , Gastric Mucosa/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Male , Middle Aged , Serum/immunology
12.
PLoS One ; 7(5): e36378, 2012.
Article in English | MEDLINE | ID: mdl-22563496

ABSTRACT

Helicobacter pylori colonizes the mucus niche of the gastric mucosa and is a risk factor for gastritis, ulcers and cancer. The main components of the mucus layer are heavily glycosylated mucins, to which H. pylori can adhere. Mucin glycosylation differs between individuals and changes during disease. Here we have examined the H. pylori response to purified mucins from a range of tumor and normal human gastric tissue samples. Our results demonstrate that mucins from different individuals differ in how they modulate both proliferation and gene expression of H. pylori. The mucin effect on proliferation varied significantly between samples, and ranged from stimulatory to inhibitory, depending on the type of mucins and the ability of the mucins to bind to H. pylori. Tumor-derived mucins and mucins from the surface mucosa had potential to stimulate proliferation, while gland-derived mucins tended to inhibit proliferation and mucins from healthy uninfected individuals showed little effect. Artificial glycoconjugates containing H. pylori ligands also modulated H. pylori proliferation, albeit to a lesser degree than human mucins. Expression of genes important for the pathogenicity of H. pylori (babA, sabA, cagA, flaA and ureA) appeared co-regulated in response to mucins. The addition of mucins to co-cultures of H. pylori and gastric epithelial cells protected the viability of the cells and modulated the cytokine production in a manner that differed between individuals, was partially dependent of adhesion of H. pylori to the gastric cells, but also revealed that other mucin factors in addition to adhesion are important for H. pylori-induced host signaling. The combined data reveal host-specific effects on proliferation, gene expression and virulence of H. pylori due to the gastric mucin environment, demonstrating a dynamic interplay between the bacterium and its host.


Subject(s)
Gastric Mucins/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Helicobacter pylori/drug effects , Helicobacter pylori/genetics , Adhesins, Bacterial/genetics , Antigens, Bacterial/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Cell Line, Tumor , Cytokines/metabolism , Dose-Response Relationship, Drug , Gastric Mucins/isolation & purification , Gastric Mucins/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Glycosylation , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/physiology , Host-Pathogen Interactions , Humans , Interleukin-6/metabolism , Microbial Viability/drug effects , Microscopy, Fluorescence , Mucin 5AC/isolation & purification , Mucin 5AC/metabolism , Mucin 5AC/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
13.
PLoS One ; 7(2): e30695, 2012.
Article in English | MEDLINE | ID: mdl-22319577

ABSTRACT

BACKGROUND: Colorectal cancer usually gives rise to a specific anti-tumor immune response, but for unknown reasons the resulting immunity is not able to clear the tumor. Recruitment of activated effector lymphocytes to the tumor is important for efficient anti-tumor responses, while the presence of regulatory T cells (Treg) down-modulate tumor-specific immunity. We therefore aimed to determine homing mechanisms and activation stage of Treg and effector T cell infiltrating colon tumors compared to cells from the unaffected mucosa in patients suffering from colon adenocarcinoma. METHODOLOGY/PRINCIPAL FINDINGS: Lymphocytes were isolated from unaffected and tumor mucosa from patients with colon adenocarcinoma, and flow cytometry, immunohistochemistry, and quantitative PCR was used to investigate the homing mechanisms and activation stage of infiltrating Treg and conventional lymphocytes. We detected significantly higher frequencies of CD25(high)FOXP3⁺CD127(low) putative Treg in tumors than unaffected mucosa, which had a complete demethylation in the FOXP3 promotor. Tumor-associated Treg had a high expression of CTLA-4, and some appeared to be antigen experienced effector/memory cells based on their expression of αEß7 (CD103). There were also significantly fewer activated T cells and more CTLA-4⁺ conventional T cells susceptible to immune regulation in the tumor-associated mucosa. In contrast, CD8⁺granzyme B⁺ putative cytotoxic cells were efficiently recruited to the tumors. The frequencies of cells expressing α4ß7 and the Th1 associated chemokine receptor CXCR3 were significantly decreased among CD4⁺ T cells in the tumor, while frequencies of CD4⁺CCR4⁺ lymphocytes were significantly increased. CONCLUSIONS/SIGNIFICANCE: This study shows that CCR4⁺CTLA4(hi) Treg accumulate in colon tumors, while the frequencies of activated conventional Th1 type T cells are decreased. The altered lymphocyte composition in colon tumors will probably diminish the ability of the immune system to effectively attack tumor cells, and reducing the Treg activity is an important challenge for future immunotherapy protocols.


Subject(s)
Adenocarcinoma, Mucinous/immunology , Colonic Neoplasms/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , CTLA-4 Antigen , Humans , Immunity , Interleukin-2 Receptor alpha Subunit , Mucous Membrane , Receptors, CCR4 , T-Lymphocytes/immunology
14.
Exp Cell Res ; 317(6): 849-58, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21255568

ABSTRACT

Gastric adenocarcinoma is a major health problem world-wide, as this is the second most common cause of cancer death in the world. It has been estimated that infection by Helicobacter pylori cause at least half of the gastric cancers. Previously, we have demonstrated that H. pylori antigens directly activate NK cells to secrete IFN-γ. There is also a marked synergistic effect in NK cells stimulated with bacterial lysate and low levels of IL-12, a cytokine which is produced by macrophages and dendritic cells in the H. pylori-infected stomach. The present study was designed to investigate whether NK cells from gastric cancer patients display an altered ability to respond to components from H. pylori and other bacteria. The results show that NK cells from peripheral blood of gastric cancer patients have a severely suppressed ability to produce IFN-γ after stimulation with H. pylori lysate and the synthetic bacterial lipoprotein FSL-1. Furthermore, the synergistic effect of IL-12 and lysate is absent in gastric cancer patients, unless the concentration of IL-12 is increased 10-fold. We also demonstrate that there is a similar lack of IFN-γ production from NK cells isolated from the gastric mucosa of cancer patients. In addition, we propose that the observed suppression is due to tumour-derived TGF-ß and that increased expression of the transcription factor GATA-3 may be responsible for the TGF-ß induced suppression.


Subject(s)
Antigens, Bacterial/pharmacology , Gastric Mucosa/immunology , Gene Expression Regulation/drug effects , Helicobacter pylori/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Aged , Cytotoxicity, Immunologic/drug effects , Down-Regulation/drug effects , Female , GATA3 Transcription Factor/metabolism , Helicobacter Infections/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Middle Aged , Stomach Neoplasms/immunology , T-Box Domain Proteins/metabolism
15.
Innate Immun ; 17(2): 191-203, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20130107

ABSTRACT

Helicobacter pylori induce a chronic inflammation in the human gastric mucosa characterized by increased production of interferon-gamma (IFN-γ). The presence of natural killer (NK) cells in the human gastric mucosa and the ability of NK cells to produce IFN-γ suggest an important role of NK cells in the immune response directed towards H. pylori infection. Since NK cells previously have been shown to respond to bacterial components with IFN-γ production, we investigated the mechanisms for the recognition of H. pylori. We found that inhibition of MyD88 homodimerization resulted in decreased production of IFN-γ and that inhibition of the p38 MAPK decreased the production as well as the secretion of IFN-γ. Further studies indicated an involvement of Toll-like receptors (TLRs), in particular TLR2. Finally, we showed that the H. pylori specific membrane bound lipoprotein HpaA induced IFN-γ production from NK cells through recognition by TLR2. In conclusion, we suggest an involvement of TLR2 in the recognition of H. pylori by human NK cells and that HpaA is a TLR2 ligand important for recognition.


Subject(s)
Adhesins, Bacterial/metabolism , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Toll-Like Receptor 2/metabolism , Adhesins, Bacterial/immunology , Gastric Mucosa/microbiology , Gene Expression Regulation/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Host-Pathogen Interactions , Humans , Inflammation , Interleukin-12/immunology , Interleukin-12/metabolism , Ligands , Myeloid Differentiation Factor 88/antagonists & inhibitors , Signal Transduction/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
J Innate Immun ; 2(3): 294-302, 2010.
Article in English | MEDLINE | ID: mdl-20375553

ABSTRACT

Natural killer (NK) cells can be activated to produce IFN-gamma by lysate from Helicobacter pylori in combination with IL-12. Furthermore, NK cells in the gastrointestinal mucosa are likely to encounter H. pylori as well as other bacteria and may play a role in the mucosal innate immune defense. In this report, we show that in marked contrast to peripheral blood, the large majority of NK cells of human gastrointestinal mucosa lack CD8 expression. Importantly, we show that CD8(-) and CD8(+) NK cells have different functional properties; although the cytotoxic capacity of the different NK cell populations was equal, only CD8(-) NK cells were capable of responding by IFN-gamma production to stimulation with lysates from H. pylori and other bacteria - this was not due to an intrinsic defect in IFN-gamma production by CD8(+) NK cells. We propose that CD8(-) CD16(-) CD56(bright) NK cells constitute a subset of NK cells that is present in the gastrointestinal mucosa and is especially adapted to responding to bacterial infection by production of cytokines. These findings may have important implications for the understanding of NK cell subsets and the innate defense against gastrointestinal bacterial infections.


Subject(s)
Gastrointestinal Tract/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunity, Mucosal , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Adult , Antigens, CD/biosynthesis , Cell Separation , Cytotoxicity, Immunologic , Flow Cytometry , Helicobacter pylori/pathogenicity , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Intestinal Mucosa/pathology , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/pathology
17.
Infect Immun ; 77(5): 1888-93, 2009 May.
Article in English | MEDLINE | ID: mdl-19237532

ABSTRACT

Considerable effort is being made to understand the acute and memory antibody responses in natural cholera infection, while rather less is known about the roles of cellular immune responses involving T and B lymphocytes. We studied responses in adult patients hospitalized with cholera caused by Vibrio cholerae O1. Peripheral blood mononuclear cells from patients (n = 15) were analyzed by flow cytometry after stimulation with V. cholerae O1 membrane protein (MP) or toxin-coregulated pilus antigen (TcpA). The gamma interferon (IFN-gamma) and interleukin-13 (IL-13) responses in stimulated-lymphocyte supernatants were studied. The responses were compared with those of healthy controls (n = 10). Patients responded with increased frequencies of gut-homing CD4(+) T cells (CD4(+) beta7(+)), gut-homing CD8(+) T cells (CD8(+) beta7(+)), and gut-homing B cells (CD19(+) beta7(+)) at the early and/or late convalescent stages compared to the acute stage. After stimulation with MP or TcpA, proliferation of CD4(+) and CD8(+) T cells was increased at the acute stage and/or early convalescent stage compared to healthy controls. Increased IL-13 and IFN-gamma responses were observed after antigenic stimulation at the acute and convalescent stages compared to healthy controls. Thus, increases in the levels of gut-homing T and B cells, as well as involvement of CD8 and CD4 Th1-mediated (IFN-gamma) and CD4 Th2-mediated (IL-13) cytokine responses, take place in acute dehydrating disease caused by V. cholerae O1. Further studies are needed to determine if such responses are also stimulated after immunization with oral cholera vaccines and if these responses play a role in protection following exposure to cholera.


Subject(s)
Cholera/immunology , T-Lymphocytes/immunology , Vibrio cholerae O1/immunology , Adolescent , Adult , B-Lymphocyte Subsets/immunology , Cells, Cultured , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-13/metabolism , Male , Middle Aged , T-Lymphocyte Subsets/immunology , Young Adult
18.
Eur J Immunol ; 38(12): 3327-38, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19003934

ABSTRACT

Organ-specific homing of lymphoid cells depends on the expression of tissue-specific adhesion molecules and production of specific chemokines. CCL25 (TECK) and CCL28 (MEC) have been reported to direct circulating memory/effector B cells to mucosal tissues. Here, we examined if differential responsiveness to mucosal and systemic chemokines could explain the differential migration pattern of circulating human antibody-secreting cells (ASC), induced by mucosal and systemic immunization. There was a robust migration of specific IgA- and IgM-ASC induced by Salmonella vaccination toward the mucosal chemokines CCL25 and CCL28. In contrast, tetanus-specific ASC migrated to the systemic chemokine CXCL12 (SDF-1alpha) and showed no response to CCL25 or CCL28, not even tetanus-specific IgA-ASC. Cell sorting experiments demonstrated that Salmonella-specific ASC co-expressed CCR9 and CCR10. Our results show that induction site, rather than isotype commitment, determines the chemokine responsiveness and migration pattern of human effector B cells.


Subject(s)
Chemokines, CC/immunology , Immunization , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Intestines/immunology , Adult , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Movement/immunology , Female , Humans , Intestines/cytology , Male , Middle Aged , Receptors, CCR10/immunology , Receptors, CCR3/immunology , Salmonella/immunology , Tetanus Toxin/immunology
19.
J Immunol ; 177(11): 7634-44, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17114433

ABSTRACT

Oral administration of Ag coupled to cholera toxin B subunit (CTB) efficiently induces peripheral immunological tolerance. We investigated the extent to which this oral tolerance is mediated by CD25+CD4+ regulatory T cells (T(reg)). We found that total T(reg), KJ1-26+ T(reg) and CTLA-4+ T(reg) were all increased in Peyer's patches, mesenteric lymph nodes, and, to a lesser extent, in spleen of mice after intragastric administration of OVA/CTB conjugate, which also increased TGF-beta in serum. This could be abolished by co-administering cholera toxin or by treatment with anti-TGF-beta mAb. CD25+ T(reg), but also CD25-CD4+ T cells from OVA/CTB-treated BALB/c or DO11.10 mice efficiently suppressed effector T cell proliferation and IL-2 production in vitro. Following adoptive transfer, both T cell populations also suppressed OVA-specific T cell and delayed-type hypersensitivity responses in vivo. Foxp3 was strongly expressed by CD25+ T(reg) from OVA/CTB-treated mice, and treatment also markedly expanded CD25+Foxp3+ T(reg). Furthermore, in Rag1(-/-) mice that had adoptively received highly purified Foxp3-CD25-CD4+ OT-II T cells OVA/CTB feeding efficiently induced CD25+ T(reg) cells, which expressed Foxp3 more strongly than naturally developing T(reg) and also had stronger ability to suppress effector OT-II T cell proliferation. A remaining CD25- T cell population, which also became suppressive in response to OVA/CTB treatment, did not express Foxp3. Our results demonstrate that oral tolerance induced by CTB-conjugated Ag is associated with increase in TGF-beta and in both the frequency and suppressive capacity of Foxp3+ and CTLA-4+ CD25+ T(reg) together with the generation of both Foxp3+ and Foxp3-CD25- CD4+ T(reg).


Subject(s)
Cholera Toxin/immunology , Forkhead Transcription Factors/metabolism , Immune Tolerance , Interleukin-2 Receptor alpha Subunit/metabolism , Ovalbumin/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Adoptive Transfer , Animals , Cell Proliferation , Cholera Toxin/administration & dosage , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage
20.
Inflamm Bowel Dis ; 12(6): 447-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16775488

ABSTRACT

BACKGROUND: Factors determining the extension and degree of inflammation in the colonic mucosa of patients with ulcerative colitis (UC) are largely unknown, but CD4+CD25high regulatory T cells (Tregs) have been implicated to play an important role in suppressing inflammation. Therefore, the aims of this study were to determine whether colonic Tregs have suppressive effects on colonic effector T cells in UC and to analyze the association between segmental colonic Treg distribution and disease activity. MATERIALS AND METHODS: The suppressive activity of colonic CD4+CD25high Tregs from patients with active UC was determined in coculture assays measuring proliferation and cytokine production. The frequency of Tregs and the expression of the Treg marker FOXP3 were analyzed with flow cytometry and RT-PCR in isolated cells and the whole mucosa from patients with active and inactive disease, as well as healthy mucosa. RESULTS: Colonic CD4+CD25high T cells from patients with UC suppressed the proliferation and cytokine secretion of colonic effector CD4+ T cells. Healthy controls but not patients with UC had lower Treg frequencies in the sigmoid than in the ascending colon. Patients with UC with active disease had increased frequency of colonic Tregs. The frequency of Tregs was positively correlated with colonic disease activity and serum C-reactive protein. CONCLUSIONS: Colonic CD4+CD25high Tregs are able to suppress colonic effector T cell activity in vitro, and the Treg frequency in the inflamed intestine increases with disease activity in patients with active UC. This suggests that Tregs may be outnumbered by other inflammatory cells or that their suppressive activity may be influenced by the in vivo environment.


Subject(s)
Colitis, Ulcerative/immunology , Colon/immunology , Intestinal Mucosa/immunology , Receptors, Interleukin-2/analysis , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Biopsy , Colitis, Ulcerative/pathology , Colon/pathology , Female , Humans , Intestinal Mucosa/pathology , Male , Middle Aged , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...