Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Article in English | MEDLINE | ID: mdl-33189701

ABSTRACT

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Subject(s)
Cell Transformation, Neoplastic/immunology , Colitis-Associated Neoplasms/immunology , Lipids/immunology , Precancerous Conditions/immunology , Stomach Neoplasms/immunology , Animals , Benzylamines/pharmacology , Benzylamines/therapeutic use , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/drug effects , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Disease Models, Animal , Epithelial Cells , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Gerbillinae , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Helicobacter pylori/isolation & purification , Humans , Lipids/antagonists & inhibitors , Metaplasia/immunology , Metaplasia/microbiology , Metaplasia/pathology , Mice , Mice, Transgenic , Organoids , Precancerous Conditions/drug therapy , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control
2.
Cell Rep ; 33(11): 108510, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326776

ABSTRACT

Innate responses of myeloid cells defend against pathogenic bacteria via inducible effectors. Deoxyhypusine synthase (DHPS) catalyzes the transfer of the N-moiety of spermidine to the lysine-50 residue of eukaryotic translation initiation factor 5A (EIF5A) to form the amino acid hypusine. Hypusinated EIF5A (EIF5AHyp) transports specific mRNAs to ribosomes for translation. We show that DHPS is induced in macrophages by two gastrointestinal pathogens, Helicobacter pylori and Citrobacter rodentium, resulting in enhanced hypusination of EIF5A. EIF5AHyp was also increased in gastric macrophages from patients with H. pylori gastritis. Furthermore, we identify the bacteria-induced immune effectors regulated by hypusination. This set of proteins includes essential constituents of antimicrobial response and autophagy. Mice with myeloid cell-specific deletion of Dhps exhibit reduced EIF5AHyp in macrophages and increased bacterial burden and inflammation. Thus, regulation of translation through hypusination is a critical hallmark of the defense of eukaryotic hosts against pathogenic bacteria.


Subject(s)
Anti-Infective Agents/therapeutic use , Lysine/analogs & derivatives , Macrophages/immunology , Animals , Anti-Infective Agents/pharmacology , Disease Models, Animal , Humans , Lysine/therapeutic use , Mice
3.
mBio ; 10(5)2019 10 29.
Article in English | MEDLINE | ID: mdl-31662455

ABSTRACT

The reverse transsulfuration pathway is the major route for the metabolism of sulfur-containing amino acids. The role of this metabolic pathway in macrophage response and function is unknown. We show that the enzyme cystathionine γ-lyase (CTH) is induced in macrophages infected with pathogenic bacteria through signaling involving phosphatidylinositol 3-kinase (PI3K)/MTOR and the transcription factor SP1. This results in the synthesis of cystathionine, which facilitates the survival of pathogens within myeloid cells. Our data demonstrate that the expression of CTH leads to defective macrophage activation by (i) dysregulation of polyamine metabolism by depletion of S-adenosylmethionine, resulting in immunosuppressive putrescine accumulation and inhibition of spermidine and spermine synthesis, and (ii) increased histone H3K9, H3K27, and H3K36 di/trimethylation, which is associated with gene expression silencing. Thus, CTH is a pivotal enzyme of the innate immune response that disrupts host defense. The induction of the reverse transsulfuration pathway by bacterial pathogens can be considered an unrecognized mechanism for immune escape.IMPORTANCE Macrophages are professional immune cells that ingest and kill microbes. In this study, we show that different pathogenic bacteria induce the expression of cystathionine γ-lyase (CTH) in macrophages. This enzyme is involved in a metabolic pathway called the reverse transsulfuration pathway, which leads to the production of numerous metabolites, including cystathionine. Phagocytized bacteria use cystathionine to better survive in macrophages. In addition, the induction of CTH results in dysregulation of the metabolism of polyamines, which in turn dampens the proinflammatory response of macrophages. In conclusion, pathogenic bacteria can evade the host immune response by inducing CTH in macrophages.


Subject(s)
Bacteria/immunology , Bacteria/metabolism , Immunity, Innate , Macrophages/metabolism , Metabolic Networks and Pathways/physiology , Sulfur/metabolism , Animals , Bacteria/pathogenicity , Gene Silencing , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Histones/metabolism , Humans , Immune Evasion , Immunoglobulins , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polyamines/metabolism , RAW 264.7 Cells , Spermidine/metabolism , Spermine/metabolism , Transcription Factors
4.
Mucosal Immunol ; 11(5): 1363-1374, 2018 09.
Article in English | MEDLINE | ID: mdl-29907869

ABSTRACT

Blood vessel epicardial substance (BVES), or POPDC1, is a tight junction-associated transmembrane protein that modulates epithelial-to-mesenchymal transition (EMT) via junctional signaling pathways. There have been no in vivo studies investigating the role of BVES in colitis. We hypothesized that BVES is critical for maintaining colonic epithelial integrity. At baseline, Bves-/- mouse colons demonstrate increased crypt height, elevated proliferation, decreased apoptosis, altered intestinal lineage allocation, and dysregulation of tight junctions with functional deficits in permeability and altered intestinal immunity. Bves-/- mice inoculated with Citrobacter rodentium had greater colonic injury, increased colonic and mesenteric lymph node bacterial colonization, and altered immune responses after infection. We propose that increased bacterial colonization and translocation result in amplified immune responses and worsened injury. Similarly, dextran sodium sulfate (DSS) treatment resulted in greater histologic injury in Bves-/- mice. Two different human cell lines (Caco2 and HEK293Ts) co-cultured with enteropathogenic E. coli showed increased attaching/effacing lesions in the absence of BVES. Finally, BVES mRNA levels were reduced in human ulcerative colitis (UC) biopsy specimens. Collectively, these studies suggest that BVES plays a protective role both in ulcerative and infectious colitis and identify BVES as a critical protector of colonic mucosal integrity.


Subject(s)
Colitis, Ulcerative/metabolism , Colon/metabolism , Epithelial Cells/metabolism , Intestinal Absorption/physiology , Membrane Proteins/metabolism , Adult , Animals , Caco-2 Cells , Cell Adhesion Molecules , Cell Line , Cell Line, Tumor , Citrobacter rodentium/pathogenicity , Coculture Techniques , Colon/drug effects , Dextran Sulfate/pharmacology , Epithelial Cells/drug effects , Escherichia coli/metabolism , Female , HEK293 Cells , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Proteins , Permeability/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tight Junctions/drug effects , Tight Junctions/metabolism
5.
Front Immunol ; 9: 1242, 2018.
Article in English | MEDLINE | ID: mdl-29922289

ABSTRACT

Polyamines have been implicated in numerous biological processes, including inflammation and carcinogenesis. Homeostatic regulation leads to interconversion of the polyamines putrescine and the downstream metabolites spermidine and spermine. The enzyme spermine oxidase (SMOX), which back-converts spermine to spermidine, contributes to regulation of polyamine levels, but can also have other effects. We have implicated SMOX in gastric inflammation and carcinogenesis due to infection by the pathogen Helicobacter pylori. In addition, we reported that SMOX can be upregulated in humans with inflammatory bowel disease. Herein, we utilized Smox-deficient mice to examine the role of SMOX in two murine colitis models, Citrobacter rodentium infection and dextran sulfate sodium (DSS)-induced epithelial injury. In C. rodentium-infected wild-type (WT) mice, there were marked increases in colon weight/length and histologic injury, with mucosal hyperplasia and inflammatory cell infiltration; these changes were ameliorated in Smox-/- mice. In contrast, with DSS, Smox-/- mice exhibited substantial mortality, and increased body weight loss, colon weight/length, and histologic damage. In C. rodentium-infected WT mice, there were increased colonic levels of the chemokines CCL2, CCL3, CCL4, CXCL1, CXCL2, and CXCL10, and the cytokines IL-6, TNF-α, CSF3, IFN-γ, and IL-17; each were downregulated in Smox-/- mice. In DSS colitis, increased levels of IL-6, CSF3, and IL-17 were further increased in Smox-/- mice. In both models, putrescine and spermidine were increased in WT mice; in Smox-/- mice, the main effect was decreased spermidine and spermidine/spermine ratio. With C. rodentium, polyamine levels correlated with histologic injury, while with DSS, spermidine was inversely correlated with injury. Our studies indicate that SMOX has immunomodulatory effects in experimental colitis via polyamine flux. Thus, SMOX contributes to the immunopathogenesis of C. rodentium infection, but is protective in DSS colitis, indicating the divergent effects of spermidine.


Subject(s)
Colitis/etiology , Colitis/metabolism , Immunomodulation , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Animals , Citrobacter rodentium/physiology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Gene Deletion , Immunity, Mucosal/genetics , Immunomodulation/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Oxidoreductases Acting on CH-NH Group Donors/genetics , Spermidine/metabolism , Spermidine/pharmacology , Spermine/metabolism , Spermine/pharmacology , Polyamine Oxidase
6.
Gut ; 67(7): 1247-1260, 2018 07.
Article in English | MEDLINE | ID: mdl-28473630

ABSTRACT

OBJECTIVE: Gastric cancer is the third leading cause of cancer death worldwide and infection by Helicobacter pylori is the strongest risk factor. We have reported increased epidermal growth factor receptor (EGFR) phosphorylation in the H. pylori-induced human carcinogenesis cascade, and association with DNA damage. Our goal was to determine the role of EGFR activation in gastric carcinogenesis. DESIGN: We evaluated gefitinib, a specific EGFR inhibitor, in chemoprevention of H. pylori-induced gastric inflammation and cancer development. Mice with genetically targeted epithelial cell-specific deletion of Egfr (EfgrΔepi mice) were also used. RESULTS: In C57BL/6 mice, gefitinib decreased Cxcl1 and Cxcl2 expression by gastric epithelial cells, myeloperoxidase-positive inflammatory cells in the mucosa and epithelial DNA damage induced by H. pylori infection. Similar reductions in chemokines, inflammatory cells and DNA damage occurred in infected EgfrΔepi versus Egfrfl/fl control mice. In H. pylori-infected transgenic insulin-gastrin (INS-GAS) mice and gerbils, gefitinib treatment markedly reduced dysplasia and carcinoma. Gefitinib blocked H. pylori-induced activation of mitogen-activated protein kinase 1/3 (MAPK1/3) and activator protein 1 in gastric epithelial cells, resulting in inhibition of chemokine synthesis. MAPK1/3 phosphorylation and JUN activation was reduced in gastric tissues from infected wild-type and INS-GAS mice treated with gefitinib and in primary epithelial cells from EfgrΔepi versus Egfrfl/fl mice. Epithelial EGFR activation persisted in humans and mice after H. pylori eradication, and gefitinib reduced gastric carcinoma in INS-GAS mice treated with antibiotics. CONCLUSIONS: These findings suggest that epithelial EGFR inhibition represents a potential strategy to prevent development of gastric carcinoma in H. pylori-infected individuals.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Gastritis/pathology , Helicobacter Infections/pathology , Quinazolines/therapeutic use , Stomach Neoplasms/prevention & control , Animals , Cell Culture Techniques , Epithelial Cells , Gastritis/microbiology , Gefitinib , Gerbillinae , Helicobacter pylori , Mice , Mice, Inbred C57BL , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
7.
Sci Rep ; 6: 39399, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982124

ABSTRACT

The intestinal microbiota of patients with constipated-predominant irritable bowel syndrome (C-IBS) displays chronic dysbiosis. Our aim was to determine whether this microbial imbalance instigates perturbation of the host intestinal mucosal immune response, using a model of human microbiota-associated rats (HMAR) and dextran sulfate sodium (DSS)-induced experimental colitis. The analysis of the microbiota composition revealed a decrease of the relative abundance of Bacteroides, Roseburia-Eubacterium rectale and Bifidobacterium and an increase of Enterobacteriaceae, Desulfovibrio sp., and mainly Akkermansia muciniphila in C-IBS patients compared to healthy individuals. The bacterial diversity of the gut microbiota of healthy individuals or C-IBS patients was maintained in corresponding HMAR. Animals harboring a C-IBS microbiota had reduced DSS colitis with a decreased expression of pro-inflammatory cytokines from innate, Th1, and Th17 responses. The pre-treatment of conventional C57BL/6 mice or HMAR with A. muciniphila, but not with Escherichia coli, prior exposure to DSS also resulted in a reduction of colitis severity, highlighting that the anti-inflammatory effect of the gut microbiota of C-IBS patients is mediated, in part, by A. muciniphila. This work highlights a novel aspect of the crosstalk between the gut microbiota of C-IBS patients and host intestinal homeostasis.


Subject(s)
Anti-Inflammatory Agents/immunology , Gastrointestinal Microbiome/immunology , Intestines/microbiology , Irritable Bowel Syndrome/microbiology , Animals , Colitis/immunology , Colitis/microbiology , Cytokines/immunology , Dextran Sulfate/immunology , Dysbiosis/immunology , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Microbiota/immunology , Rats
8.
PLoS Pathog ; 12(10): e1005984, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783672

ABSTRACT

Solute carrier family 7 member 2 (SLC7A2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in immune responses to pathogens. We assessed the role of SLC7A2 in murine infection with Citrobacter rodentium, an attaching and effacing enteric pathogen that causes colitis. Induction of SLC7A2 was upregulated in colitis tissues, and localized predominantly to colonic epithelial cells. Compared to wild-type mice, Slc7a2-/-mice infected with C. rodentium had improved survival and decreased weight loss, colon weight, and histologic injury; this was associated with decreased colonic macrophages, dendritic cells, granulocytes, and Th1 and Th17 cells. In infected Slc7a2-/-mice, there were decreased levels of the proinflammatory cytokines G-CSF, TNF-α, IL-1α, IL-1ß, and the chemokines CXCL1, CCL2, CCL3, CCL4, CXCL2, and CCL5. In bone marrow chimeras, the recipient genotype drove the colitis phenotype, indicative of the importance of epithelial, rather than myeloid SLC7A2. Mice lacking Slc7a2 exhibited reduced adherence of C. rodentium to the colonic epithelium and decreased expression of Talin-1, a focal adhesion protein involved in the attachment of the bacterium. The importance of SLC7A2 and Talin-1 in the intimate attachment of C. rodentium and induction of inflammatory response was confirmed in vitro, using conditionally-immortalized young adult mouse colon (YAMC) cells with shRNA knockdown of Slc7a2 or Tln1. Inhibition of L-Arg uptake with the competitive inhibitor, L-lysine (L-Lys), also prevented attachment of C. rodentium and chemokine expression. L-Lys and siRNA knockdown confirmed the role of L-Arg and SLC7A2 in human Caco-2 cells co-cultured with enteropathogenic Escherichia coli. Overexpression of SLC7A2 in human embryonic kidney cells increased bacterial adherence and chemokine expression. Taken together, our data indicate that C. rodentium enhances its own pathogenicity by inducing the expression of SLC7A2 to favor its attachment to the epithelium and thus create its ecological niche.


Subject(s)
Cationic Amino Acid Transporter 2/metabolism , Enterobacteriaceae Infections/metabolism , Host-Parasite Interactions/physiology , Animals , Blotting, Western , Cationic Amino Acid Transporter 2/immunology , Cell Line , Citrobacter rodentium , Disease Models, Animal , Enterobacteriaceae Infections/immunology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Transfection
9.
J Clin Invest ; 126(9): 3296-312, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27482886

ABSTRACT

EGFR signaling regulates macrophage function, but its role in bacterial infection has not been investigated. Here, we assessed the role of macrophage EGFR signaling during infection with Helicobacter pylori, a bacterial pathogen that causes persistent inflammation and gastric cancer. EGFR was phosphorylated in murine and human macrophages during H. pylori infection. In human gastric tissues, elevated levels of phosphorylated EGFR were observed throughout the histologic cascade from gastritis to carcinoma. Deleting Egfr in myeloid cells attenuated gastritis and increased H. pylori burden in infected mice. EGFR deficiency also led to a global defect in macrophage activation that was associated with decreased cytokine, chemokine, and NO production. We observed similar alterations in macrophage activation and disease phenotype in the Citrobacter rodentium model of murine infectious colitis. Mechanistically, EGFR signaling activated NF-κB and MAPK1/3 pathways to induce cytokine production and macrophage activation. Although deletion of Egfr had no effect on DC function, EGFR-deficient macrophages displayed impaired Th1 and Th17 adaptive immune responses to H. pylori, which contributed to decreased chronic inflammation in infected mice. Together, these results indicate that EGFR signaling is central to macrophage function in response to enteric bacterial pathogens and is a potential therapeutic target for infection-induced inflammation and associated carcinogenesis.


Subject(s)
Bacterial Infections/metabolism , ErbB Receptors/metabolism , Macrophage Activation , Macrophages/metabolism , Animals , Chemokines/metabolism , Citrobacter rodentium , Cytokines/metabolism , Disease Progression , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori , Humans , Inflammation , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Phosphorylation , Signal Transduction
10.
Amino Acids ; 48(10): 2375-88, 2016 10.
Article in English | MEDLINE | ID: mdl-27074721

ABSTRACT

We reported that arginase 2 (ARG2) deletion results in increased gastritis and decreased bacterial burden during Helicobacter pylori infection in mice. Our studies implicated a potential role for inducible nitric oxide (NO) synthase (NOS2), as Arg2 (-/-) mice exhibited increased NOS2 levels in gastric macrophages, and NO can kill H. pylori. We now bred Arg2 (-/-) to Nos2 (-/-) mice, and infected them with H. pylori. Compared to wild-type mice, both Arg2 (-/-) and Arg2 (-/-) ;Nos2 (-/-) mice exhibited increased gastritis and decreased colonization, the latter indicating that the effect of ARG2 deletion on bacterial burden was not mediated by NO. While Arg2 (-/-) mice demonstrated enhanced M1 macrophage activation, Nos2 (-/-) and Arg2 (-/-) ;Nos2 (-/-) mice did not demonstrate these changes, but exhibited increased CXCL1 and CXCL2 responses. There was an increased expression of the Th1/Th17 cytokines, interferon gamma and interleukin 17, in gastric tissues and splenic T-cells from Arg2 (-/-), but not Nos2 (-/-) or Arg2 (-/-) ;Nos2 (-/-) mice. Gastric tissues from infected Arg2 (-/-) mice demonstrated increased expression of arginase 1, ornithine decarboxylase, adenosylmethionine decarboxylase 1, spermidine/spermine N (1)-acetyltransferase 1, and spermine oxidase, along with increased spermine levels. These data indicate that ARG2 deletion results in compensatory upregulation of gastric polyamine synthesis and catabolism during H. pylori infection, which may contribute to increased gastric inflammation and associated decreased bacterial load. Overall, the finding of this study is that ARG2 contributes to the immune evasion of H. pylori by restricting M1 macrophage activation and polyamine metabolism.


Subject(s)
Arginase/immunology , Biogenic Polyamines/immunology , Helicobacter Infections/immunology , Helicobacter pylori/metabolism , Immune Evasion , Macrophage Activation , Macrophages , Stomach , Animals , Arginase/genetics , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
11.
Am J Physiol Gastrointest Liver Physiol ; 309(4): G270-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26113297

ABSTRACT

Nonalcoholic fatty liver disease occurs frequently in the setting of metabolic syndrome, but the factors leading to nonalcoholic steatohepatitis (NASH) are not fully understood. This study investigated Toll-like receptor 4 (TLR4) signaling in human liver with the goal of delineating whether activation of this pathway segregates those with nonalcoholic fatty liver from those with NASH. Experiments were performed using liver biopsy tissue obtained from class III obese subjects undergoing bariatric surgery, and extended to an immortalized human hepatocyte HepaRG cell line and primary human hepatocytes. The bacterial endotoxin lipopolysaccharide (LPS) and total free fatty acid levels were significantly increased in plasma of NASH patients. TLR4 mRNA levels were significantly increased in subjects with NASH compared with NAFL as was interferon regulatory factor (IRF) 3 in the myeloid differentiation factor 88-independent signaling pathway. In HepaRG cells, nuclear factor-κB (NF-κB) nuclear translocation and functional activity increased following treatment with the fatty acid, palmitate, and following exposure to LPS compared with hepatocytes stimulated with a lipogenic treatment that induced de novo lipogenesis. Palmitate and LPS induction of NF-κB activity was partially attenuated by chemical- or small-interfering RNA-mediated inhibition of TLR4. Expression of TLR4 and its downstream mediators was upregulated with palmitate and LPS. Similar results were observed using primary human hepatocytes from a lean donor. Interestingly, NF-κB activity assays showed obese donor hepatocytes were resistant to chemical TLR4 inhibition. In conclusion, TLR4 expression is upregulated in a large cohort of NASH patients, compared with those with NAFL, and this occurs within the setting of increased LPS and fatty acids.


Subject(s)
Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Adult , Cell Line , Cells, Cultured , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/blood , Lipopolysaccharides/pharmacology , Male , Middle Aged , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/complications , Obesity/complications , Palmitic Acid/blood , Palmitic Acid/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/genetics
12.
J Immunol ; 193(6): 3013-22, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25108023

ABSTRACT

Helicobacter pylori incites a futile inflammatory response, which is the key feature of its immunopathogenesis. This leads to the ability of this bacterial pathogen to survive in the stomach and cause peptic ulcers and gastric cancer. Myeloid cells recruited to the gastric mucosa during H. pylori infection have been directly implicated in the modulation of host defense against the bacterium and gastric inflammation. Heme oxygenase-1 (HO-1) is an inducible enzyme that exhibits anti-inflammatory functions. Our aim was to analyze the induction and role of HO-1 in macrophages during H. pylori infection. We now show that phosphorylation of the H. pylori virulence factor cytotoxin-associated gene A (CagA) in macrophages results in expression of hmox-1, the gene encoding HO-1, through p38/NF (erythroid-derived 2)-like 2 signaling. Blocking phagocytosis prevented CagA phosphorylation and HO-1 induction. The expression of HO-1 was also increased in gastric mononuclear cells of human patients and macrophages of mice infected with cagA(+) H. pylori strains. Genetic ablation of hmox-1 in H. pylori-infected mice increased histologic gastritis, which was associated with enhanced M1/Th1/Th17 responses, decreased regulatory macrophage (Mreg) response, and reduced H. pylori colonization. Gastric macrophages of H. pylori-infected mice and macrophages infected in vitro with this bacterium showed an M1/Mreg mixed polarization type; deletion of hmox-1 or inhibition of HO-1 in macrophages caused an increased M1 and a decrease of Mreg phenotype. These data highlight a mechanism by which H. pylori impairs the immune response and favors its own survival via activation of macrophage HO-1.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Heme Oxygenase-1/immunology , Macrophages/immunology , Membrane Proteins/immunology , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line , Enzyme Inhibitors/pharmacology , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastritis/immunology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Imidazoles/pharmacology , Inflammation/immunology , Interleukin-10/biosynthesis , MAP Kinase Signaling System/immunology , Macrophages/enzymology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/immunology , Nitric Oxide Synthase Type II/biosynthesis , Phagocytosis/immunology , Phosphorylation/immunology , Pyridines/pharmacology , Signal Transduction/immunology , Stomach/microbiology , Stomach/pathology , Th1 Cells/immunology , Th17 Cells/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/immunology
13.
Cell Microbiol ; 15(1): 145-56, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23051580

ABSTRACT

The cytotoxin-associated gene A protein (CagA) plays a pivotal role in the aetiology of Helicobacter pylori-associated gastric diseases. CagA is injected into the cytoplasm of host cells by a type IV secretion system, and is phosphorylated on tyrosine residues by the host enzyme c-Src. We previously reported that the enzyme haem oxygenase-1 (HO-1) inhibits IL-8 secretion by H. pylori-infected cells. However, the cellular mechanism by which HO-1 regulates the innate immune function of infected cells remains unknown. We now show that nitric oxide and haemin, two inducers of HO-1, decrease the level of phosphorylated CagA (p-CagA) in H. pylori-infected gastric epithelial cells and this is blocked by either pharmacological inhibition of HO-1 or siRNA knockdown of hmox-1. Moreover, forced expression of HO-1 by transfection of a plasmid expressing hmox-1 also results in a strong attenuation of CagA phosphorylation. This occurs through the inhibition of H. pylori-induced c-Src phosphorylation/activation by HO-1. Consequently, H. pylori-induced cytoskeletal rearrangements and activation of the pro-inflammatory response mediated by p-CagA are inhibited in HO-1-expressing cells. These data highlight a mechanism by which the innate immune response of the host can restrict the pathogenicity of H. pylori by attenuating CagA phosphorylation in gastric epithelial cells.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Heme Oxygenase-1/metabolism , Protein Processing, Post-Translational , Cell Line , Host-Pathogen Interactions , Humans , Oncogene Proteins/immunology , Oncogene Proteins/metabolism , Phosphorylation
14.
J Immunol ; 187(10): 5370-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21987660

ABSTRACT

A strong cellular cross-talk exists between the pathogen Helicobacter pylori and high-output NO production. However, how NO and H. pylori interact to signal in gastric epithelial cells and modulate the innate immune response is unknown. We show that chemical or cellular sources of NO induce the anti-inflammatory effector heme oxygenase-1 (HO-1) in gastric epithelial cells through a pathway that requires NF-κB. However, H. pylori decreases NO-induced NF-κB activation, thereby inhibiting HO-1 expression. This inhibitory effect of H. pylori results from activation of the transcription factor heat shock factor-1 by the H. pylori virulence factor CagA and by the host signaling molecules ERK1/2 and JNK. Consistent with these findings, HO-1 is downregulated in gastric epithelial cells of patients infected with cagA(+) H. pylori but not in gastric epithelial cells of patients infected with cagA(-) H. pylori. Enhancement of HO-1 activity in infected cells or in H. pylori-infected mice inhibits chemokine generation and reduces inflammation. These data define a mechanism by which H. pylori favors its own pathogenesis by inhibiting HO-1 induction through the action of CagA.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Helicobacter pylori/immunology , Heme Oxygenase-1/antagonists & inhibitors , Inflammation Mediators/physiology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Signal Transduction/immunology , Up-Regulation/immunology , Animals , Cell Line , Cell Line, Transformed , Gastric Mucosa/enzymology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter pylori/pathogenicity , Heme Oxygenase-1/biosynthesis , Humans , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Virulence Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...