Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Biol Chem ; 295(32): 11082-11098, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32518160

ABSTRACT

Infection with the Gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and oxidative DNA damage in gastric epithelial cells that can lead to gastric cancer (GC). However, the underlying pathogenic mechanism is largely unclear. Here, we report that the suppression of Nei-like DNA glycosylase 2 (NEIL2), a mammalian DNA glycosylase that specifically removes oxidized bases, is one mechanism through which H. pylori infection may fuel the accumulation of DNA damage leading to GC. Using cultured cell lines, gastric biopsy specimens, primary cells, and human enteroid-derived monolayers from healthy human stomach, we show that H. pylori infection greatly reduces NEIL2 expression. The H. pylori infection-induced downregulation of NEIL2 was specific, as Campylobacter jejuni had no such effect. Using gastric organoids isolated from the murine stomach in coculture experiments with live bacteria mimicking the infected stomach lining, we found that H. pylori infection is associated with the production of various inflammatory cytokines. This response was more pronounced in Neil2 knockout (KO) mouse cells than in WT cells, suggesting that NEIL2 suppresses inflammation under physiological conditions. Notably, the H. pylori-infected Neil2-KO murine stomach exhibited more DNA damage than the WT. Furthermore, H. pylori-infected Neil2-KO mice had greater inflammation and more epithelial cell damage. Computational analysis of gene expression profiles of DNA glycosylases in gastric specimens linked the reduced Neil2 level to GC progression. Our results suggest that NEIL2 downregulation is a plausible mechanism by which H. pylori infection impairs DNA damage repair, amplifies the inflammatory response, and initiates GC.


Subject(s)
DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Down-Regulation , Gastric Mucosa/metabolism , Genome , Helicobacter Infections/metabolism , Helicobacter pylori/isolation & purification , Inflammation/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , DNA Glycosylases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Progression , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/metabolism , Humans , Mice , RNA, Messenger/genetics
2.
Curr Top Microbiol Immunol ; 421: 229-265, 2019.
Article in English | MEDLINE | ID: mdl-31123892

ABSTRACT

Helicobacter pylori is a prevalent human pathogen that successfully establishes chronic infection, which leads to clinically significant gastric diseases including chronic gastritis, peptic ulcer disease (PUD), and gastric cancer (GC). H. pylori is able to produce a persistent infection due in large part to its ability to hijack the host immune response. The host adaptive immune response is activated to strategically and specifically attack pathogens and normally clears them from the infected host. Since B and T lymphocytes are central mediators of adaptive immunity, in this chapter we review their development and the fundamental mechanisms regulating their activation in order to understand how some of the normal processes are subverted by H. pylori. In this review, we place particular emphasis on the CD4+ T cell responses, their subtypes, and regulatory mechanisms because of the expanding literature in this area related to H. pylori. T lymphocyte differentiation and function are finely orchestrated through a series of cell-cell interactions, which include immune checkpoint receptors. Among the immune checkpoint receptor family, there are some with inhibitory properties that are exploited by tumor cells to facilitate their immune evasion. Gastric epithelial cells (GECs), which act as antigen-presenting cells (APCs) in the gastric mucosa, are induced by H. pylori to express immune checkpoint receptors known to sway T lymphocyte function and thus circumvent effective T effector lymphocyte responses. This chapter reviews these and other mechanisms used by H. pylori to interfere with host immunity in order to persist.


Subject(s)
B-Lymphocytes/pathology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immune Evasion , T-Lymphocytes/pathology , B-Lymphocytes/immunology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Humans , T-Lymphocytes/immunology
3.
J Immunol ; 193(5): 2218-29, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070848

ABSTRACT

Signaling via programmed death ligand-1 (PD-L1) and PD-L2 is crucial for maintaining peripheral tolerance. CD90(+) myofibroblasts/fibroblasts (CMFs) are major programmed cell death-1 (PD-1) ligand-expressing cells in normal human colonic mucosa. CMFs suppress activated CD4(+) T cell proliferation via PD-1 ligands. It is not known whether signaling through TLRs contribute to the regulation PD-1 ligands on CMFs upon colonic mucosal tolerance. In this study, we demonstrated that stimulation of TLR4 on human CMFs upregulates PD-L1, but not PD-L2, and reinforces CMF-mediated suppression of CD4(+) T cell proliferation and IFN-γ production. TLR4-mediated upregulation of PD-L1 on CMFs involved NF-κB pathways and was JAK2 and MyD88 dependent. MyD88-dependent stimulation of TLR1/2 and TLR5 also upregulated PD-L1 expression on CMFs in culture. PD-L1 expression was drastically decreased in vivo in the colonic mucosa of mice devoid of MyD88. Induction of MyD88 deficiency in CMFs in fibroblast-specific MyD88 conditional knockout mice resulted in a strong increase in a mucosal IFN-γ expression concomitantly with the abrogation of PD-L1 expression in CMFs under homeostasis and epithelial injury induced by dextran sodium sulfate. Together, these data suggest that MyD88-dependent TLR stimulation of CMFs in the normal colonic mucosa may reinforce these cells' anti-inflammatory capacity and thus contribute to the maintenance of mucosal tolerance.


Subject(s)
B7-H1 Antigen/immunology , Colon/immunology , Immune Tolerance/physiology , Intestinal Mucosa/immunology , Thy-1 Antigens/immunology , Toll-Like Receptor 4/immunology , Animals , B7-H1 Antigen/genetics , Colon/cytology , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Mucosa/cytology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myofibroblasts/cytology , Myofibroblasts/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Thy-1 Antigens/genetics , Toll-Like Receptor 4/genetics , Up-Regulation/genetics , Up-Regulation/immunology
4.
J Immunol ; 191(7): 3838-46, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23997227

ABSTRACT

Gastric epithelial cells (GECs) are the primary target for Helicobacter pylori infection and may act as APCs regulating local T cell responses. We previously reported that H. pylori infection of GECs induces the expression of the T cell coinhibitory molecule B7-H1 on GECs. This process contributes to the hyporesponsiveness of CD4(+) effector T cells and accumulation of regulatory T cells. In the present study, we investigated the impact of H. pylori cytotoxin-associated gene A (CagA) on the modulation of the expression of the T cell costimulator B7-H2 by GECs. B7-H2 is involved in promoting Th17 type responses. H. pylori infection downregulates B7-H2 expression by GECs in a CagA-dependent manner. IFN-γ, which is increased in the H. pylori-infected gastric mucosa, synergizes with H. pylori in downregulating B7-H2 expression by GECs. CagA-mediated modulation of B7-H2 on GECs involves p70 S6 kinase phosphorylation. The CagA-dependent B7-H2 downregulation in GECs correlates with a decrease in Th17 type responses in vitro and in vivo. Furthermore, CagA-dependent modulation of Th17 responses was inversely correlated with the H. pylori colonization levels in vivo. Our data suggest that CagA contributes to the ability of H. pylori to evade Th17-mediated clearance by modulating expression of B7-H2 and, thus, to the establishment of the H. pylori chronic infection.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter Infections/immunology , Helicobacter pylori/metabolism , Inducible T-Cell Co-Stimulator Ligand/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Cell Line , Down-Regulation , Female , Gene Expression Regulation/drug effects , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Interferon-gamma/pharmacology , Mice , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 306(5): G361-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24407592

ABSTRACT

Fetal swallowing of amniotic fluid, which contains numerous cytokines and growth factors, plays a key role in gut mucosal development. Preterm birth interrupts this exposure to amniotic fluid-borne growth factors, possibly contributing to the increased risk of necrotizing enterocolitis (NEC) in premature infants. We hypothesized that supplementation of formula feeds with amniotic fluid can provide amniotic fluid-borne growth factors and prevent experimental NEC in rat pups. We compared NEC-like injury in rat pups fed with infant formula vs. formula supplemented either with 30% amniotic fluid or recombinant hepatocyte growth factor (HGF). Cytokines/growth factors in amniotic fluid were measured by immunoassays. Amniotic fluid and HGF effects on enterocyte migration, proliferation, and survival were measured in cultured IEC6 intestinal epithelial cells. Finally, we used an antibody array to investigate receptor tyrosine kinase (RTK) activation and immunoblots to measure phosphoinositide 3-kinase (PI3K) signaling. Amniotic fluid supplementation in oral feeds protected rat pups against NEC-like injury. HGF was the most abundant growth factor in rat amniotic fluid in our panel of analytes. Amniotic fluid increased cell migration, proliferation, and cell survival in vitro. These effects were reproduced by HGF and blocked by anti-HGF antibody or a PI3K inhibitor. HGF transactivated several RTKs in IEC6 cells, indicating that its effects extended to multiple signaling pathways. Finally, similar to amniotic fluid, recombinant HGF also reduced the frequency and severity of NEC-like injury in rat pups. Amniotic fluid supplementation protects rat pups against experimental NEC, which is mediated, at least in part, by HGF.


Subject(s)
Amniotic Fluid/metabolism , Enterocolitis, Necrotizing/prevention & control , Hepatocyte Growth Factor/administration & dosage , Amniotic Fluid/chemistry , Animal Feed , Animals , Cells, Cultured , Cytokines/metabolism , Dietary Supplements , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Enzymologic , Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/metabolism , Humans , Infant , Infant Formula , Intestinal Mucosa/cytology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
6.
Microorganisms ; 11(5)2023 May 17.
Article in English | MEDLINE | ID: mdl-37317287

ABSTRACT

Gastric cancer is a challenging public health concern worldwide and remains a leading cause of cancer-related mortality. The primary risk factor implicated in gastric cancer development is infection with Helicobacter pylori. H. pylori induces chronic inflammation affecting the gastric epithelium, which can lead to DNA damage and the promotion of precancerous lesions. Disease manifestations associated with H. pylori are attributed to virulence factors with multiple activities, and its capacity to subvert host immunity. One of the most significant H. pylori virulence determinants is the cagPAI gene cluster, which encodes a type IV secretion system and the CagA toxin. This secretion system allows H. pylori to inject the CagA oncoprotein into host cells, causing multiple cellular perturbations. Despite the high prevalence of H. pylori infection, only a small percentage of affected individuals develop significant clinical outcomes, while most remain asymptomatic. Therefore, understanding how H. pylori triggers carcinogenesis and its immune evasion mechanisms is critical in preventing gastric cancer and mitigating the burden of this life-threatening disease. This review aims to provide an overview of our current understanding of H. pylori infection, its association with gastric cancer and other gastric diseases, and how it subverts the host immune system to establish persistent infection.

7.
Gastroenterology ; 140(7): 2019-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376048

ABSTRACT

BACKGROUND & AIMS: Regulatory T (Treg) cells (CD4+ CD25high FoxP3+) regulate mucosal tolerance; their adoptive transfer prevents or reduces symptoms of colitis in mouse models of inflammatory bowel disease. Colonic CD90+ mesenchymal myofibroblasts and fibroblasts (CMFs) are abundant, nonprofessional antigen-presenting cells in the normal human colonic mucosa that suppress proliferation of activated CD4+ effector T cells. We studied CMF suppressive capacity and evaluated the ability of CMF to induce Treg cells. METHODS: Allogeneic cocultures of CD4+ T cells and CMFs, derived from normal mucosa of patients undergoing colectomy for colon cancer or inflamed colonic tissues from patients with ulcerative colitis or Crohn's disease, were used to assess activation of the Treg cells. RESULTS: Coculture of normal CMF with resting or naïve CD4+ T cells led to development of cells with a Treg phenotype; it also induced proliferation of a CD25+ CD127- FoxP3+ T cells, which expressed CTLA-4, interleukin-10, and transforming growth factor-ß and had suppressive activities. In contrast to dendritic cells, normal CMFs required exogenous interleukin-2 to induce proliferation of naturally occurring Treg cells. Induction of Treg cells by normal CMFs required major histocompatibility complex class II and prostaglandin E2. CMFs from patients with inflammatory bowel diseases had reduced capacity to induce active Treg cells and increased capacity to transiently generate CD4+CD25+/- CD127+ T cells that express low levels of FoxP3. CONCLUSIONS: CMFs suppress the immune response in normal colon tissue and might therefore help maintain colonic mucosal tolerance. Alterations in CMF-mediated induction of Treg cells might promote pathogenesis of inflammatory bowel diseases.


Subject(s)
Cell Proliferation , Colon/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Myofibroblasts/immunology , Paracrine Communication , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Coculture Techniques , Colonic Neoplasms/immunology , Dinoprostone/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immune Tolerance , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Leukocyte Common Antigens/metabolism , Phenotype , Signal Transduction , Time Factors , Transforming Growth Factor beta/metabolism
8.
Med Res Arch ; 10(12)2022 Dec.
Article in English | MEDLINE | ID: mdl-37936946

ABSTRACT

H. pylori is perhaps the most prevalent human pathogen worldwide and infects almost half of the world's population. Despite the decreasing prevalence of infection overall, it is significant in developing countries. Most infections are acquired in childhood and persist for a lifetime unless treated. Children are often asymptomatic and often develop a tolerogenic immune response that includes T regulatory cells and their products, immunosuppressive cytokines, such as interleukin (IL)-10, and transforming growth factor-ß (TGF-ß). This contrasts to the gastric immune response seen in H. pylori-infected adults, where the response is mainly inflammatory, with predominant Th1 and Th17 cells, as well as, inflammatory cytokines, such as TNF-α, IFN-γ, IL-1, IL-6, IL-8, and IL-17. Therefore, compared to adults, infected children generally have limited gastric inflammation and peptic ulcer disease. H. pylori surreptitiously subverts immune defenses to persist in the human gastric mucosa for decades. The chronic infection might result in clinically significant diseases in adults, such as peptic ulcer disease, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. This review compares the infection in children and adults and highlights the H. pylori virulence mechanisms responsible for the pathogenesis and immune evasion.

9.
Infect Immun ; 79(7): 2737-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482686

ABSTRACT

Gastric epithelial cells (GECs) express the class II major histocompatibility complex (MHC) and costimulatory molecules, enabling them to act as antigen-presenting cells (APCs) and affect local T cell responses. During Helicobacter pylori infection, GECs respond by releasing proinflammatory cytokines and by increasing the surface expression of immunologically relevant receptors, including class II MHC. The CD4(+) T cell response during H. pylori infection is skewed toward a Th1 response, but these cells remain hyporesponsive. Activated T cells show decreased proliferation during H. pylori infection, and CD4(+) CD25(+) FoxP3(+) regulatory T cells (Tregs) are present at the site of infection. In this study, we examined the mechanisms surrounding the CD4(+) T cell responses during H. pylori infection and found that transforming growth factor ß (TGF-ß) plays a major role in these responses. GECs produced TGF-ß1 and TGF-ß2 in response to infection. Activated CD4(+) T cells in culture with H. pylori-treated GECs were decreased in proliferation but increased upon neutralization of TGF-ß. Naïve CD4(+) T cell development into Tregs was also enhanced in the presence of GEC-derived TGF-ß. Herein, we demonstrate a role for GEC-produced TGF-ß in the inhibition of CD4(+) T cell responses seen during H. pylori infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Cell Proliferation , Cytokines/biosynthesis , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Flow Cytometry , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Genes, MHC Class II , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Humans , Interleukin-2 Receptor alpha Subunit , Lymphocyte Activation , Polymerase Chain Reaction , Stomach Neoplasms , T-Lymphocytes, Regulatory/metabolism
10.
Gastroenterology ; 135(4): 1228-1237, 1237.e1-2, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18760278

ABSTRACT

BACKGROUND & AIMS: A prominent role for inhibitory molecules PD-L1 and PD-L2 in peripheral tolerance has been proposed. However, the phenotype and function of PD-L-expressing cells in human gut remains unclear. Recent studies suggest that colonic myofibroblasts (CMFs) and fibroblasts are important in the switch from acute inflammation to adaptive immunity. In the normal human colon, CMFs represent a distinct population of major histocompatibility complex class II(+) cells involved in the regulation of mucosal CD4(+) T-cell responses. METHODS: PD-L1 and PD-L2 expression on human CMFs was determined using Western blot, fluorescence-activated cell sorter analysis and confocal microscopy. Lymphoproliferation assays and cytokine enzyme-linked immunosorbent assays were used to evaluate the role of B7 costimulators expressed by CMFs with regard to the regulation of preactivated T-helper cell responses. RESULTS: We demonstrate here the expression of PD-L1/2 molecules by normal human CMF and fibroblasts in situ and in culture. Both molecules support suppressive functions of CMFs in the regulation of activated CD4(+) T-helper cell proliferative responses; blocking this interaction reverses the suppressive effect of CMFs on T-cell proliferation and leads to increased production of the major T-cell growth factor, interleukin (IL)-2. PD-L1/2-mediated CMF suppressive functions are mainly due to the inhibition of IL-2 production, because supplementation of the coculture media with exogenous IL-2 led to partial recovery of activated T-cell proliferation. CONCLUSIONS: Our data suggest that stromal myofibroblasts and fibroblasts may limit T-helper cell proliferative activity in the gut and, thus, might play a prominent role in mucosal intestinal tolerance.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Colon/immunology , Fibroblasts/physiology , Immune Tolerance/physiology , Antigens, CD/genetics , Antigens, CD/metabolism , B7-H1 Antigen , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cell Division/drug effects , Cell Division/immunology , Cells, Cultured , Colon/cytology , Fibroblasts/cytology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-2/pharmacology , Phenotype , Programmed Cell Death 1 Ligand 2 Protein , RNA, Small Interfering , Stromal Cells/cytology , Stromal Cells/physiology , Thy-1 Antigens/metabolism
11.
Clin Oncol Res ; 2(5)2019.
Article in English | MEDLINE | ID: mdl-31998864

ABSTRACT

Helicobacter pylori (H. pylori) is a gram negative bacterium that infects more than 50% of humanity and is associated with gastritis, peptic ulcer and gastric cancer. Although CD4+ T cells are recruited to the gastric mucosa, the host is unable to clear the bacteria. Previously, we demonstrated that H. pylori infection upregulates the expression of the T cell co-inhibitory molecule B7-H1 while simultaneously downregulating the expression of T cell co-stimulatory molecule B7-H2 on gastric epithelial cells (GEC), which together affect the Treg and Th17 cell balance and foster bacterial persistence. Because B7-H3, another member of the B7 family of co-inhibitory receptors, has been found to have important immunoregulatory roles and in cancer, in this study we examined the expression of B7-H3 molecules on GEC and how the expression is regulated by H. pylori during infection. Our study showed that both human and murine GEC constitutively express B7-H3 molecules, but their expression levels increased during H. pylori infection. We further demonstrated that H. pylori uses its type 4 secretion system (T4SS) components CagA and cell wall peptidoglycan (PG) fragment to upregulate B7-H3. Th17 cells and Treg cells which are increased during H. pylori infection also had an effect on B7-H3 induction. The underlying cell signaling pathway involves modulation of p38MAPK pathway. Since B7-H3 were shown to up-regulate Th2 responses, the phenotype of T cell subpopulations in mice infected with H. pylori PMSS1 or SS1 strains were characterized. A mixed Th1/Th2 response in H. pylori infected mice was observed. Consistent with previous findings, increased Treg cells and decreased Th17 cells in MLN of PMSS1 infected mice compared to SS1 infected mice was observed. Human biopsy samples collected from gastritis biopsies and gastric tumors showed a strong association between increased B7-H3 and Th2 responses in H. pylori strains associated with gastritis. T cell: GEC co-cultures and anti-B7-H3 blocking Ab confirmed that the induction of Th2 is mediated by B7-H3 and associated exclusively with an H. pylori gastritis strain not cancer or ulcer strains. In conclusion, these studies revealed a novel regulatory mechanism employed by H. pylori to influence the type of T cell response that develops within the infected gastric mucosa.

12.
Infect Immun ; 76(7): 3233-40, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18474653

ABSTRACT

While a link between Helicobacter pylori exposure and gastric cancer has been established, the underlying mechanisms remain unclear. H. pylori induces a chronic inflammatory response in infected individuals. A link between chronic inflammation and carcinogenesis has long been suggested but never elucidated. Epidermal growth factor receptor (EGFR) signaling plays an important role in both proinflammatory and procarcinogenic mechanisms and is upregulated on gastric epithelial cells (GECs) during H. pylori exposure. The aim of this study was to examine the effects of two important proinflammatory cytokines released during H. pylori infection, macrophage migration inhibitory factor (MIF) and interleukin-8 (IL-8), on the expression and transactivation of EGFR and on the proliferation of GECs during H. pylori exposure. The expression of EGFR by GECs was increased by exposure to either H. pylori, recombinant MIF, or recombinant IL-8. However, cag pathogenicity island knockout strains of H. pylori had very little effect on expression. MIF and IL-8 also induced phosphorylation of EGFR, signaling events, and proliferation during H. pylori exposure, all of which were decreased when they were neutralized by these cytokines or were blocked from their receptors. The overall role of EGFR in these responses to H. pylori exposure was assessed by knocking down EGFR expression by small interfering RNA.


Subject(s)
Epithelial Cells/metabolism , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Helicobacter pylori/pathogenicity , Interleukin-8/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/microbiology , Epithelial Cells/physiology , Gastric Mucosa/cytology , Humans , Interleukin-8/pharmacology , Macrophage Migration-Inhibitory Factors/pharmacology , Phosphorylation
13.
Front Immunol ; 9: 1125, 2018.
Article in English | MEDLINE | ID: mdl-29910803

ABSTRACT

Background and Aims: The role of programmed cell death protein 1 (PD-1) and its ligands in the dysregulation of T helper immune responses observed in the inflammatory bowel disease (IBD) is unclear. Recently, a novel concept emerged that CD90+ colonic (myo)fibroblasts (CMFs), also known as stromal cells, act as immunosuppressors, and are among the key regulators of acute and chronic inflammation. The objective of this study was to determine if the level of the PD-1 ligands is changed in the IBD inflamed colonic mucosa and to test the hypothesis that changes in IBD-CMF-mediated PD-1 ligand-linked immunosuppression is a mechanism promoting the dysregulation of Th1 cell responses. Methods: Tissues and cells derived from Crohn's disease (CD), ulcerative colitis (UC), and healthy individuals (N) were studied in situ, ex vivo, and in culture. Results: A significant increase in programmed death-ligand 1 (PD-L1) was observed in the inflamed UC colonic mucosa when compared to the non-inflamed matched tissue samples, CD, and healthy controls. UC-CMFs were among the major populations in the colonic mucosa contributing to the enhanced PD-L1 expression. In contrast, PD-L1 expression was decreased in CD-CMFs. When compared to CD-CMFs and N-CMFs, UC-CMFs demonstrated stronger suppression of IL-2, Th1 transcriptional factor Tbet, and IFN-γ expression by CD3/CD28-activated CD4+ T cells, and this process was PD-L1 dependent. Similar observations were made when differentiated Th1 cells were cocultured with UC-CMFs. In contrast, CD-CMFs showed reduced capacity to suppress Th1 cell activity and addition of recombinant PD-L1 Fc to CD-CMF:T cell cocultures partially restored the suppression of the Th1 type responses. Conclusion: We present evidence showing that increased PD-L1 expression suppresses Th1 cell activity in UC. In contrast, loss of PD-L1 expression observed in CD contributes to the persistence of the Th1 inflammatory milieu in CD. Our data suggest that dysregulation of the Th1 responses in the inflamed colonic mucosa of IBD patients is promoted by the alterations in PD-L1 expression in the mucosal mesenchymal stromal cell compartment.


Subject(s)
B7-H1 Antigen/genetics , Colitis, Ulcerative/etiology , Colitis, Ulcerative/metabolism , Crohn Disease/etiology , Crohn Disease/metabolism , Stromal Cells/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Thy-1 Antigens/metabolism , Actins/metabolism , Adolescent , Adult , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis, Ulcerative/pathology , Colitis, Ulcerative/therapy , Crohn Disease/pathology , Crohn Disease/therapy , Cytokines/metabolism , Female , Gene Expression Regulation , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Activation , Male , Mice , Microscopy, Confocal , Middle Aged , Myofibroblasts/metabolism , RNA, Messenger/genetics , Young Adult
14.
World J Gastroenterol ; 12(35): 5599-605, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-17007010

ABSTRACT

H. pylori is probably the most prevalent human pathogen worldwide. Since it was initially suggested in 1983 by Marshall and Warren to be implicated in gastritis and peptic ulcer disease, H. pylori has also been implicated in gastric carcinoma and was classified as a class I carcinogen. In the last two decades, a noteworthy body of research has revealed the multiple processes that this gram negative bacterium activates to cause gastroduodenal disease in humans. Most infections are acquired early in life and may persist for the life of the individual. While infected individuals mount an inflammatory response that becomes chronic, along with a detectable adaptive immune response, these responses are ineffective in clearing the infection. H. pylori has unique features that allow it to reside within the harsh conditions of the gastric environment, and also to evade the host immune response. In this review, we discuss the various virulence factors expressed by this bacterium and how they interact with the host epithelium to influence pathogenesis.


Subject(s)
Gastrointestinal Tract/pathology , Helicobacter Infections/etiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Cell Communication/physiology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastrointestinal Tract/microbiology , Humans , Immunity, Mucosal
15.
World J Gastroenterol ; 12(35): 5593-8, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-17007009

ABSTRACT

The gastric mucosa separates the underlying tissue from the vast array of antigens that traffic through the stomach lumen. While the extreme pH of this environment is essential in aiding the activation of enzymes and food digestion, it also renders the gastric epithelium free from bacterial colonization, with the exception of one important human pathogen, H. pylori. This bacterium has developed mechanisms to survive the harsh environment of the stomach, actively move through the mucosal layer, attach to the epithelium, evade immune responses, and achieve persistent colonization. While a hallmark of this infection is a marked inflammatory response with the infiltration of various immune cells into the infected gastric mucosa, the host immune response is unable to clear the infection and may actually contribute to the associated pathogenesis. Here, we review the host responses involved during infection with H. pylori and how they are influenced by this bacterium.


Subject(s)
Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori/pathogenicity , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Humans , Immunity, Mucosal/immunology , T-Lymphocytes/pathology
16.
World J Gastroenterol ; 12(33): 5306-10, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16981259

ABSTRACT

AIM: To investigate the role of MHC class II in the modulation of gastric epithelial cell apoptosis induced by H pylori infection. METHODS: After stimulating a human gastric epithelial cell line with bacteria or agonist antibodies specific for MHC class II and CD95, the quantitation of apoptotic and anti-apoptotic events, including caspase activation, BCL-2 activation, and FADD recruitment, was performed with a fluorometric assay, a cytometric bead array, and confocal microscopy, respectively. RESULTS: Pretreatment of N87 cells with the anti-MHC class II IgM antibody RFD1 resulted in a reduction in global caspase activation at 24 h of H pylori infection. When caspase 3 activation was specifically measured, crosslinking of MHC class II resulted in a marked reduced caspase activation, while simple ligation of MHC class II did not. Crosslinking of MHC class II also resulted in an increased activation of the anti-apoptosis molecule BCL-2 compared to simple ligation. Confocal microscope analysis demonstrated that the pretreatment of gastric epithelial cells with a crosslinking anti-MHC class II IgM blocked the recruitment of FADD to the cell surface. CONCLUSION: The results presented here demonstrate that the ability of MHC class II to modulate gastric epithelial apoptosis is at least partially dependent on its crosslinking. Furthermore, while previous research has demonstrated that MHC class II signaling can be pro-apoptotic during extended ligation, we have shown that the crosslinking of this molecule has anti-apoptotic effects during the earlier time points of H pylori infection. This effect is possibly mediated by the ability of MHC class II to modulate the activation of the pro-apoptotic receptor Fas by blocking the recruitment of the accessory molecule FADD, and this delay in apoptosis induction could allow for prolonged cytokine secretion by H pylori-infected gastric epithelial cells.


Subject(s)
Apoptosis , Epithelium/metabolism , Epithelium/microbiology , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Genes, MHC Class II , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Histocompatibility Antigens Class II/metabolism , Caspase 3/metabolism , Caspases/metabolism , Cell Line , Cell Line, Tumor , DNA Fragmentation , Enzyme Activation , Humans , Microscopy, Confocal , Proto-Oncogene Proteins c-bcl-2/metabolism , fas Receptor/biosynthesis
17.
World J Gastroenterol ; 12(29): 4689-93, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16937440

ABSTRACT

AIM: To investigate the role of MHC class II in the modulation of gastric epithelial cell apoptosis induced by H pylori infection. METHODS: After stimulating a human gastric epithelial cell line with bacteria or agonist antibodies specific for MHC class II and CD95, the quantitation of apoptotic and anti-apoptotic events, including caspase activation, BCL-2 activation, and FADD recruitment, was performed with a fluorometric assay, a cytometric bead array, and confocal microscopy, respectively. RESULTS: Pretreatment of N87 cells with the anti-MHC class II IgM antibody RFD1 resulted in a reduction in global caspase activation at 24 h of H pylori infection. When caspase 3 activation was specifically measured, crosslinking of MHC class II resulted in markedly reduced caspase activation, while simple ligation of MHC class II did not. Crosslinking of MHC class II also resulted in an increased activation of the anti-apoptosis molecule BCL-2 compared to simple ligation. Confocal microscope analysis demonstrated that the pretreatment of gastric epithelial cells with a crosslinking anti-MHC class II IgM blocked the recruitment of FADD to the cell surface. CONCLUSION: The ability of MHC class II to modulate gastric epithelial apoptosis is at least partially dependent on its crosslinking. The crosslinking of this molecule has anti-apoptotic effects during the earlier time points of H pylori infection. This effect is possibly mediated by the ability of MHC class II to modulate the activation of the pro-apoptotic receptor Fas by blocking the recruitment of the accessory molecule FADD, and this delay in apoptosis induction could allow for prolonged cytokine secretion by H pylori-infected gastric epithelial cells.


Subject(s)
Apoptosis/physiology , Helicobacter Infections/physiopathology , Helicobacter pylori/immunology , Histocompatibility Antigens Class II/physiology , Stomach/microbiology , Stomach/pathology , Adaptor Proteins, Signal Transducing/physiology , Antibodies/immunology , Antibodies/pharmacology , Caspase 3 , Caspases/physiology , Cell Line , Enzyme Activation/physiology , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/pathology , Fas-Associated Death Domain Protein , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Histocompatibility Antigens Class II/immunology , Humans , Proto-Oncogene Proteins c-bcl-2/physiology , Stomach/drug effects , Stomach/physiopathology , fas Receptor/immunology , fas Receptor/physiology
18.
J Histochem Cytochem ; 53(12): 1481-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15923369

ABSTRACT

CD74 is known as the major histocompatibility complex (MHC) class II-associated invariant chain (Ii) that regulates the cell biology and functions of MHC class II molecules. Class II MHC and Ii expression was believed to be restricted to classical antigen-presenting cells (APC); however, during inflammation, other cell types, including mucosal epithelial cells, have also been reported to express class II MHC molecules. Given the importance of Ii in the biology of class II MHC, we sought to examine the expression of Ii by gastric epithelial cells (GEC) to determine whether class II MHC molecules in these nonconventional APC cells were under the control of Ii and to further support the role that these cells may play in local immune and inflammatory responses during Helicobacter pylori infection. Thus we examined the expression of Ii on GEC from human biopsy samples and then confirmed this observation using independent methods on several GEC lines. The mRNA for Ii was detected by RT-PCR, and the various protein isoforms were also detected. Interestingly, these cells have a high level expression of surface Ii, which is polarized to the apical surface. These studies are the first to demonstrate the constitutive expression of Ii by human GEC.


Subject(s)
Antigen-Presenting Cells/metabolism , Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Histocompatibility Antigens Class II/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Cell Line , Gastric Mucosa/cytology , Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/metabolism , Helicobacter pylori , Histocompatibility Antigens Class II/genetics , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
19.
PLoS One ; 10(3): e0121841, 2015.
Article in English | MEDLINE | ID: mdl-25807464

ABSTRACT

During Helicobacter pylori (H. pylori) infection CD4+ T cells in the gastric lamina propria are hyporesponsive and polarized by Th1/Th17 cell responses controlled by Treg cells. We have previously shown that H. pylori upregulates B7-H1 expression on GEC, which, in turn, suppress T cell proliferation, effector function, and induce Treg cells in vitro. In this study, we investigated the underlying mechanisms and the functional relevance of B7-H1 induction by H. pylori infection to chronic infection. Using H. pylori wild type (WT), cag pathogenicity island (cag PAI-) and cagA- isogenic mutant strains we demonstrated that H. pylori requires its type 4 secretion system (T4SS) as well as its effector protein CagA and peptidoglycan (PG) fragments for B7-H1 upregulation on GEC. Our study also showed that H. pylori uses the p38 MAPK pathway to upregulate B7-H1 expression in GEC. In vivo confirmation was obtained when infection of C57BL/6 mice with H. pylori PMSS1 strain, which has a functional T4SS delivery system, but not with H. pylori SS1 strain lacking a functional T4SS, led to a strong upregulation of B7-H1 expression in the gastric mucosa, increased bacterial load, induction of Treg cells in the stomach, increased IL-10 in the serum. Interestingly, B7-H1-/- mice showed less Treg cells and reduced bacterial loads after infection. These studies demonstrate how H. pylori T4SS components activate the p38 MAPK pathway, upregulate B7-H1 expression by GEC, and cause Treg cell induction; thus, contribute to establishing a persistent infection characteristic of H. pylori.


Subject(s)
B7-H1 Antigen/metabolism , Genomic Islands , Helicobacter pylori/genetics , Immune Evasion/genetics , Animals , B7-H1 Antigen/immunology , Cell Proliferation , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Interleukin-10/metabolism , Mice , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Up-Regulation
20.
J Neuroimmunol ; 134(1-2): 104-10, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12507777

ABSTRACT

Class II MHC mutant bm12 mice have an increased resistance to experimental autoimmune myasthenia gravis (EAMG) compared to C57BL/6 mice. In vitro, this relative resistance was mainly associated with a reduced cytokine response to acetylcholine receptor (AChR) and its dominant pathogenic peptide alpha 146-162, whereas the response to the epitope alpha 111-126 remained intact. Calcium mobilization after stimulation of AChR-immune T cells with AChR or alpha 146-162 peptide, but not alpha 111-126 peptide, was decreased in bm12 compared to C57BL/6. Thus, the reduced incidence of clinical EAMG in bm12 is linked to lower IFN-gamma and IL-10 release, and intracellular calcium mobilization by alpha 146-162-specific T cells.


Subject(s)
Calcium Signaling/genetics , Histocompatibility Antigens Class II/immunology , Immune Tolerance/genetics , Immunity, Innate/genetics , Myasthenia Gravis, Autoimmune, Experimental/genetics , Receptors, Cholinergic/immunology , T-Lymphocytes/immunology , Animals , Calcium/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , Epitopes/immunology , Histocompatibility Antigens Class II/genetics , Immunity, Innate/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Receptors, Cholinergic/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL