Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.966
Filter
1.
Parasit Vectors ; 15(1): 25, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35033166

ABSTRACT

BACKGROUND: Due to increased anthelmintic resistance, alternative methods to drugs are necessary to control gastrointestinal nematodes (GINs). Some of the most promising alternatives are based on the immune response of the host, such as the selection of genetically resistant breeds or the use of vaccines against these parasites. Given the limited information available on the immune response against GINs in goats, this study investigated the local immune response of goat kids of an indigenous Canary Islands breed (Majorera breed) experimentally infected with Teladorsagia circumcincta, one of the most pathogenic and prevalent GIN species. METHODS: For this purpose, the relationship between different parasitological (number of mature and immature worms, worm length, and number of intrauterine eggs) and immunological parameters at the local level (related to both the humoral and cellular immune response) was analyzed at early (1 week post-infection [wpi]) and late (8 wpi) stages of infection. RESULTS: Primary infection of goat kids with T. circumcincta infective larvae (L3) generated a complex immune response that could be defined as Th2 type, characterized by increased infiltration in abomasal tissues of several effector cells as well as a progressive presence of specific antibodies against parasitic antigens in the gastric mucus. Cellular responses were evidenced from 1 wpi onward, showing an increase in antigen-presenting cells and various lymphocyte subsets in the gastric mucosa. CONCLUSIONS: The complexity of the host response was evidenced by statistically significant changes in the number of all these subpopulations (MHCII+, CD4+, CD8+, γδ+, CD45R+, IgA+, and IgG+), as well as in the evolution of the relative cytokine gene expression. From a functional point of view, negative associations were observed between the number of most of the immune cells (CD4, IgA, IgG, and CD45R cells) and parameters that could be related to the fecundity of worms, a phenomenon that was especially evident when the number of IgG and CD45R cells or the specific IgA levels of the gastric mucus were compared with parasitological parameters such as the female worm length or fecal egg counts at 8 wpi.


Subject(s)
Goat Diseases/immunology , Goat Diseases/parasitology , Trichostrongyloidea/immunology , Trichostrongyloidiasis/veterinary , Abomasum/immunology , Abomasum/parasitology , Animals , Feces/parasitology , Female , Gastric Mucosa/immunology , Gastric Mucosa/parasitology , Goats , Parasite Egg Count/veterinary , Spain , Trichostrongyloidiasis/immunology , Trichostrongyloidiasis/parasitology
2.
J Sci Food Agric ; 102(3): 1255-1262, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34358346

ABSTRACT

BACKGROUND: Artemisia capillaris is among the most abundantly used traditional medicines, utilized in East Asia to treat diverse illnesses, including gastrointestinal tract diseases. We previously reported that an aqueous extract of A. capillaris (AEAC) inhibited gastric inflammation induced by HCl/ethanol via reactive oxygen species scavenging and NF-κB downregulation. To date, the pharmacological potential of AEAC for promoting mucosal integrity has not been studied. RESULTS: Here, we report that a single treatment with AEAC increased mucus production, and repeated administration of AEAC abolished HCl/ethanol-induced mucosal injury in vivo. Single- and multiple-dose AEAC treatments measurably increased the expression of mucosal stabilizing factors in vivo, including mucin (MUC) 5 AC, MUC6, and trefoil factor (TFF) 1 and TFF2 (but not TFF3). AEAC also induced mucosal stabilizing factors in both SNU-601 cells and RGM cells through phosphorylation of extracellular signal-regulated kinases. CONCLUSION: Taken together, our results suggest that AEAC protects against HCl/ethanol-induced gastritis by upregulating MUCs and TFFs and stabilizing the mucosal epithelium. © 2021 Society of Chemical Industry.


Subject(s)
Artemisia/chemistry , Drugs, Chinese Herbal/pharmacology , Gastric Mucosa/drug effects , Stomach Diseases/drug therapy , Animals , Gastric Mucosa/immunology , Gastric Mucosa/injuries , Humans , Male , Mucins/genetics , Mucins/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Plant Leaves/chemistry , Rats , Rats, Sprague-Dawley , Stomach Diseases/genetics , Stomach Diseases/immunology , Trefoil Factor-1/genetics , Trefoil Factor-1/immunology
3.
Cell Rep Med ; 2(10): 100419, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34755133

ABSTRACT

Immune checkpoint blockade using PD-1 inhibition is an effective approach for treating a wide variety of cancer subtypes. While lower gastrointestinal (GI) side effects are more common, upper gastrointestinal adverse events are rarely reported. Here, we present a case of nivolumab-associated autoimmune gastritis. To elucidate the immunology underlying this condition, we leverage multiplexed ion beam imaging by time-of-flight (MIBI-TOF) to identify the presence and proportion of infiltrating immune cells from a single section of biopsy specimen. Using MIBI-TOF, we analyze formalin-fixed, paraffin-embedded human gastric tissue with 28 labels simultaneously. Our analyses reveal a gastritis characterized by severe mucosal injury, interferon gamma (IFN-γ)-producing gastric epithelial cells, and mixed inflammation that includes CD8 and CD4 T cell infiltrates with reduced expression of granzyme B and FOXP3, respectively. Here, we provide a comprehensive multiplexed histopathological mapping of gastric tissue, which identifies IFN-γ-producing epithelial cells as possible contributors to the nivolumab-associated gastritis.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Gastritis/chemically induced , Immune Checkpoint Inhibitors/adverse effects , Interferon-gamma/immunology , Nivolumab/adverse effects , Antineoplastic Agents, Immunological/administration & dosage , Biopsy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/genetics , Gastritis/immunology , Gastritis/pathology , Gene Expression , Granzymes/genetics , Granzymes/immunology , Humans , Immune Checkpoint Inhibitors/administration & dosage , Interferon-gamma/genetics , Middle Aged , Nivolumab/administration & dosage , Stomach/drug effects , Stomach/immunology , Stomach/pathology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology
4.
Clin Sci (Lond) ; 135(22): 2541-2558, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34730176

ABSTRACT

OBJECTIVE: Regulated in development and DNA damage responses-1 (REDD1) is a conserved and ubiquitous protein, which is induced in response to multiple stimuli. However, the regulation, function and clinical relevance of REDD1 in Helicobacter pylori-associated gastritis are presently unknown. APPROACH: Immunohistochemistry, real-time PCR and Western blot analyses were performed to examine the levels of REDD1 in gastric samples from H. pylori-infected patients and mice. Gastric tissues from Redd1-/- and wildtype (WT, control) mice were examined for inflammation. Gastric epithelial cells (GECs), monocytes and T cells were isolated, stimulated and/or cultured for REDD1 regulation and functional assays. RESULTS: REDD1 was increased in gastric mucosa of H. pylori-infected patients and mice. H. pylori induced GECs to express REDD1 via the phosphorylated cytotoxin associated gene A (cagA) that activated MAPKp38 pathway to mediate NF-κB directly binding to REDD1 promoter. Human gastric REDD1 increased with the severity of gastritis, and mouse REDD1 from non-marrow chimera-derived cells promoted gastric inflammation that was characterized by the influx of MHCII+ monocytes. Importantly, gastric inflammation, MHCII+ monocyte infiltration, IL-23 and IL-17A were attenuated in Redd1-/- mice. Mechanistically, REDD1 in GECs regulated CXCL1 production, which attracted MHCII+ monocytes migration by CXCL1-CXCR2 axis. Then H. pylori induced MHCII+ monocytes to secrete IL-23, which favored IL-17A-producing CD4+ cell (Th17 cell) polarization, thereby contributing to the development of H. pylori-associated gastritis. CONCLUSIONS: The present study identifies a novel regulatory network involving REDD1, which collectively exert a pro-inflammatory effect within gastric microenvironment. Efforts to inhibit this REDD1-dependent pathway may prove valuable strategies in treating of H. pylori-associated gastritis.


Subject(s)
Cytokines/metabolism , Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Th17 Cells/microbiology , Transcription Factors/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Case-Control Studies , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastritis/immunology , Gastritis/metabolism , Helicobacter Infections/complications , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Host-Pathogen Interactions , Humans , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phenotype , Phosphorylation , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Front Immunol ; 12: 736269, 2021.
Article in English | MEDLINE | ID: mdl-34589088

ABSTRACT

Gastric CD4+T cells contribute to Helicobacter pylori (H. pylori)-induced gastritis by amplifying mucosal inflammation and exacerbating mucosal injuries. However, the pathogenic CD4+ T cell subset involved in gastritis and the potential regulators are still unclear. Here we identified an IL-21-producing gastric CD4+T cell subset, which exhibited tissue-resident CXCR5-BTLA-PD-1hi TFH-like phenotype in H. pylori-positive gastritis patients. Meanwhile, we identified glucocorticoid-induced tumor necrosis factor receptor (GITR) as an important regulator to facilitate IL-21 production by CD4+T cells and accelerate mucosal inflammation in gastritis patients with H. pylori infection. Moreover, GITR expression was increased in gastric CD4+T cells of gastritis patients compared to healthy controls, along with the upregulated expression of its ligand GITRL in mucosal macrophages (Mϕ) of gastritis patients. Further observations showed that the activation of GITR/GITRL signal promoted the IL-21 production of CD4+T cells via the STAT3 pathway. Besides this, IL-21 from CD4+T cells induced the proliferation of B cell and promoted the production of inflammatory cytokines IL-1ß and IL-6 and chemokines MIP-3α and CCL-25 as well as matrix metalloproteinase (MMP)-3 and MMP-9 by human gastric epithelial cells, suggesting the facilitating effect of IL-21-producing CD4+T cells on mucosal inflammation and injuries. Taking these data together, we revealed that GITR/GITRL signal promoted the polarization of mucosal IL-21-producing CD4+T cells in H. pylori-positive gastritis, which may provide therapeutic strategies for the clinical treatment of H. pylori-induced gastritis.


Subject(s)
Gastric Mucosa/metabolism , Gastritis/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Immunity, Mucosal , Interleukins/metabolism , T Follicular Helper Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/diagnosis , Gastritis/immunology , Gastritis/microbiology , Helicobacter Infections/diagnosis , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Ligands , Phenotype , Signal Transduction , T Follicular Helper Cells/immunology , Tumor Necrosis Factors/metabolism
6.
Immunol Lett ; 239: 1-11, 2021 11.
Article in English | MEDLINE | ID: mdl-34363898

ABSTRACT

BACKGROUND: The progression to gastric cancer has been linked to chronic infection with Helicobacter pylori (H. pylori). Immune checkpoint inhibitors (programmed cell death -1, PD-1; programmed cell death -ligand 1, PD-L1) have a role in cancer immune escape. The relationship between H. pylori virulence factors with PD-1, PD-L1 T helper 1 (Th1), T helper 17 (Th17), and regulatory T cell (Treg) response genes, has not been thoroughly investigated in the development of gastric cancer. Therefore, we evaluated how H. pylori virulence factors influence the expression levels of immune-related genes in the development of gastric immunopathology. METHODS: A total of 92 gastric tissues of normal controls and patients with gastritis, gastric ulcer, and gastric cancer were examined for the expression of immune-checkpoint inhibitor genes (PD-1 PD-L1), Th1 (interferon- γ, IFN-γ), Th17 (interleukin- 17, IL-17, Retinoic-acid-receptor- related orphan nuclear receptor gamma t, RORγ-t), and Treg (Forkhead box P3, FOXP3) response genes with quantitative real-time PCR (qRT-PCR). Furthermore, correlation of H. pylori virulence factors' (cytotoxin-associated gene A, cagA; vacuolating cytotoxin gene A, vacA (s1,s2,m1,m2); blood group antigen-binding adhesin gene A, babA, duodenal ulcer promoting gene A, dupA; the putative neuraminyllactose-binding hemagglutinin homolog, hpaA; neutrophil-activating protein A napA; outer inflammatory protein A, oipA; urease A, ureA; and urease B, ureB) genotypes with a degree of inflammation and density of H. pylori were investigated. Next, the relationship between H. pylori virulence factors and immune-checkpoint inhibitor genes, and T-cell response genes was evaluated. Eventually, a decision tree model was developed to determine the clinical outcome of patients using expression data. RESULTS: The intensity of PD-1 and PD-L1 mRNA expression was increased significantly in gastric tissue of patients with gastric ulcer (PD-1: 2.3 fold, p=0.01; PD-L1: 2.1 fold, p=0.004), and gastric cancer (PD-1: 2 fold, p= 0.04; PD-L1: 1.8 fold, p=0.05) compared with control subjects. Also, PD-1: PD-L1 expression was significantly higher in patients with gastritis, who were infected with a marked density of H. pylori compared with its mildly infected counterparts. Furthermore, a novel negative correlation was found between PD-1 (r= -0.43) and PD-L1 (r= -0.42) with FOXP3 in patients with gastritis. CagA-positive H. pylori strain's negative association with PD-L1 expression (r=-0.34) was detected in patients with gastritis. Interestingly, PD-1 mRNA expression correlated positively with vacA s2/m2, in gastritis (r=0.43) and ulcer (r=0.43) patients. Furthermore, PD-1: PDL1 expression negatively correlated with vacA m1/m2 (r=-0.43 for PD-1; r=-0.38 for PD-L1) in gastritis patients. Moreover, an inverse correlation of PDL1 was present with vacA m1 (r=0.52) and vacA s1/m1 (r=0.46) versus vacA m2 (r=-0.44) and vacA m1 (r=0.52) and vacA s1/m2 (r=-0.14) in ulcer patients, respectively. Also, a correlation of vacA m2 (r=-0.47) and vacA s1/s2 (r= 0.45) with PD-1 was detected in ulcer patients. In addition, a novel negative correlation between FOXP3 mRNA levels and napA was shown in patients with gastritis and ulcer (r=-0.59). Finally, a computer-based model that was developed showed that knowing the expression levels of PD-L1, RORγ-t, and vacA s1/m2 would be useful to detect the clinical outcome of a patient. CONCLUSION: Our results suggested that PD-1:PD-L1 immune checkpoint inhibitors were increased in gastric pre-cancerous lesions that progress to gastric cancer. Herein, we report the relationship between H. pylori virulence factors and expression of host immune checkpoint inhibitors for diagnostic prediction of gastric malignancies using computer-based models.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Helicobacter Infections/pathology , Programmed Cell Death 1 Receptor/metabolism , Stomach Neoplasms/diagnosis , Virulence Factors/immunology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Biomarkers, Tumor/analysis , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Progression , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Programmed Cell Death 1 Receptor/analysis , Signal Transduction/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Ulcer/immunology , Stomach Ulcer/microbiology , Stomach Ulcer/pathology , Virulence Factors/metabolism , Young Adult
7.
Eur J Immunol ; 51(11): 2641-2650, 2021 11.
Article in English | MEDLINE | ID: mdl-34398472

ABSTRACT

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


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Colostrum/immunology , Helicobacter pylori/immunology , Immunoglobulin A, Secretory/immunology , Cytoskeleton , Epithelial Cells , Female , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Humans , Pregnancy
8.
Food Funct ; 12(17): 7664-7675, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34236362

ABSTRACT

The aim of this study was to investigate the immunomodulatory effect and mechanism of the glycopeptides from Paecilomyces sinensis (CPS-II) on ethanol induced ulcers in mice. In this study, histopathological evaluation (H&E staining) and the gastric ulcer score, ulcer index, total acid secretion and gastric pH value were used to determine the anti-ulcer activity. The expression levels of interleukin (IL)-6, interleukin (IL)-10 and tumor necrosis factor-α (TNF-α) were detected by ELISA. The contents of superoxide dismutase (SOD), malondialdehyde (MDA) and epidermal growth factor (PEG2) in serum were measured according to the instructions for the reagents. Western blotting was used to detect the effect of CPS-II on the MEK/ERK pathway. The results showed that CPS-II could inhibit the ulcer score and ulcer index compared with the disease control group. CPS-II could significantly increase gastric pH and decrease gastric acid secretion in mice. The ELISA analysis showed that the expression levels of IL-6 and TNF-α in the CPS-II treatment group were significantly decreased, while the expression levels of IL-10 were significantly increased in the CPS-II treatment group. In the resveratrol treatment group, the content of MDA in serum was decreased, and the level of PEG2 and the activity of SOD in serum were significantly increased, which indicated that CPS-II has immunoregulation and anti-ulcer properties. The CPS-II treatment group could reduce the expression level of miR-9-5p in gastric tissue. pEGFR had been identified as a potential target of miR-9-5p. Western blot analysis showed that CPS-II could up-regulate the relative protein expression of pEGFR/EGFR, pRaf/Raf, pMEK/MEK, pERK/ERK, and ZO-1. The results showed that CPS-II could reduce oxidative stress and inflammatory response by regulating the miR-9-5p-MEK/ERK signaling pathway, thus protecting the gastric mucosa and improving stress gastric ulcers.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Glycopeptides/administration & dosage , Hypocreales/chemistry , MicroRNAs/immunology , Mitogen-Activated Protein Kinase Kinases/immunology , Stomach Ulcer/drug therapy , Animals , Ethanol/adverse effects , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Oxidative Stress/drug effects , Stomach Ulcer/chemically induced , Stomach Ulcer/genetics , Stomach Ulcer/immunology
9.
J Immunol Res ; 2021: 6539702, 2021.
Article in English | MEDLINE | ID: mdl-34258299

ABSTRACT

Serum CD4, CD8, and CD19 are markers of systemic inflammation. However, there is little evidence on the influence of inflammation on the tumor microenvironment and the prognostic indicators of gastric cancer (GC). In this study, two hundred and eight patients who underwent radical gastrectomy for GC were included. Preoperative peripheral blood samples were used to analyze Serum CD4, CD8, and CD19. The optimal cutoff levels for CD4, CD8, and CD19 were defined by receiver operating characteristic curve analysis (CD4 = 38.85%, CD8 = 14.35%, and CD19 = 7.40%). The areas with specific CD4+T cells, CD8+T cells, and CD19+B cells within the tumor microenvironment were measured in paraffin sections by immunohistochemistry and analyzed by Image-Pro Plus. 94 patients had low CD4, and 124 patients had high CD4 levels. 31 patients had low CD8, and 187 patients had high CD8 levels. 64 patients had low CD19, and 154 patients had high CD19 levels. Infiltration of CD4+T cells was associated with serum CD4 (P < 0.001). Serum CD4 and CD19 and the infiltration of CD4+T cells, CD8+T cells, and CD19+B cells were significant in predicting the prognosis of GC. Low CD4 level, infiltration of CD8+T cells, and high infiltration of CD4+T cells and CD19+B cells were correlated with worse overall survival in multivariate analysis. Collectively, our results provide evidence that serum CD4 is associated with the infiltration of CD4+T cells in the tumor microenvironment, which indicates the prognostic value of systemic inflammation in GC.


Subject(s)
Biomarkers, Tumor/blood , CD4 Antigens/blood , CD4-Positive T-Lymphocytes/immunology , Stomach Neoplasms/immunology , Aged , Antigens, CD19/blood , CD8 Antigens/blood , Female , Follow-Up Studies , Gastrectomy , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastric Mucosa/surgery , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating , Male , Prognosis , Stomach Neoplasms/blood , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery , Survival Rate , Tumor Microenvironment/immunology
10.
Future Oncol ; 17(25): 3383-3396, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34291647

ABSTRACT

Gastric cancer is the fourth most common type of cancer worldwide and the second most lethal. Gastric cancer biomarkers can be used for diagnosis, prediction of sensitivity to treatment, and prognosis. The following search terms were applied to PubMed as of December 2020: 'gastric cancer classification', 'gastric cancer epidemiology', 'cancer metastasis' and 'gastric cancer biomarker'. Only experimental studies were reported in the 'biomarkers' section. Some biomarkers can serve as therapeutic targets for antitumoral drugs. The genes analyzed include E-cadherin, RPRM, XAF1, MINT25, TFF1, p16 and p53. The miRNAs analyzed include miR-18a, miR185-5p, miR-125b and miR-21. Some molecules were associated with metastasis of gastric cancer, specifically those involved with EMT process and tissue degradation.


Lay abstract Gastric cancer is the fourth most common type of cancer worldwide and the second most lethal. Gastric cancer biomarkers are molecules that have different expressions in tumor cells than in normal body cells, and can be used for diagnosis, prediction of sensitivity to treatment, and prognosis. Biomarkers in gastric cancer can include genes that suppress tumor progression, genes that increase tumor progression by binding to growth molecules, molecules related to the body's immune response to the tumor, and non-coding RNA molecules (RNA molecules that do not produce proteins but regulate the cell's genetic material). Some biomarkers can serve as therapeutic targets for anti-tumoral drugs.


Subject(s)
Biomarkers, Tumor/analysis , Stomach Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Promoter Regions, Genetic , Risk Assessment/methods , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality
11.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946994

ABSTRACT

The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.


Subject(s)
Bacteriophages/physiology , Diet , Enteric Nervous System/physiology , Gastric Mucosa/virology , Gastrointestinal Microbiome , Host Microbial Interactions/physiology , Intestinal Mucosa/virology , Viruses , Defensins/physiology , Digestion , Disease Susceptibility , Enteric Nervous System/virology , Food/virology , Gastric Mucosa/immunology , Gastric Mucosa/innervation , Gastric Mucosa/metabolism , Gastroenteritis/virology , Host Microbial Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Malnutrition/virology , Mucus/metabolism , Mucus/virology , Neurons/virology , Opportunistic Infections/virology , Plant Viruses , Virus Diseases/microbiology , Virus Diseases/physiopathology
12.
Infect Immun ; 89(7): e0073820, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33941576

ABSTRACT

Trypanosoma cruzi is the intracellular parasite of Chagas disease, a chronic condition characterized by cardiac and gastrointestinal morbidity. Protective immunity requires CD4+ T cells, and Th1 cells and gamma interferon (IFN-γ) are important players in host defense. More recently, Th17 cells and interleukin 17A (IL-17A) have been shown to exert protective functions in systemic T. cruzi infection. However, it remains unclear whether Th17 cells and IL-17A protect in the mucosa, the initial site of parasite invasion in many human cases. We found that IL-17RA knockout (KO) mice are highly susceptible to orogastric infection, indicating an important function for this cytokine in mucosal immunity to T. cruzi. To investigate the specific role of Th17 cells for mucosal immunity, we reconstituted RAG1 KO mice with T. cruzi-specific T cell receptor transgenic Th17 cells prior to orogastric T. cruzi challenges. We found that Th17 cells provided protection against gastric mucosal T. cruzi infection, indicated by significantly lower stomach parasite burdens. In vitro macrophage infection assays revealed that protection by Th17 cells is reduced with IL-17A neutralization or reversed by loss of macrophage NADPH oxidase activity. Consistently with this, mice lacking functional NADPH oxidase were not protected by Th17 cell transfer. These data are the first report that Th17 cells protect against mucosal T. cruzi infection and identify a novel protective mechanism involving the induction of NADPH oxidase activity by IL-17A. These studies provide important insights for Chagas vaccine development and, more broadly, increase our understanding of the diverse roles of Th17 cells in host defense.


Subject(s)
Chagas Disease/immunology , Gastric Mucosa/immunology , Gastric Mucosa/parasitology , Host-Parasite Interactions/immunology , Immunity, Mucosal , Th17 Cells/immunology , Trypanosoma cruzi/immunology , Animals , Chagas Disease/metabolism , Chagas Disease/parasitology , Disease Models, Animal , Interleukin-17/genetics , Interleukin-17/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/parasitology , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Th17 Cells/metabolism
13.
Clin Transl Gastroenterol ; 12(5): e00349, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33979305

ABSTRACT

INTRODUCTION: It is unclear how immune perturbations may influence the pathogenesis of idiopathic gastroparesis, a prevalent functional disorder of the stomach which lacks animal models. Several studies have noted altered immune characteristics in the deep gastric muscle layer associated with gastroparesis, but data are lacking for the mucosal layer, which is endoscopically accessible. We hypothesized that immune dysregulation is present in the gastroduodenal mucosa in idiopathic gastroparesis and that specific immune profiles are associated with gastroparesis clinical parameters. METHODS: In this cross-sectional prospective case-control study, routine endoscopic biopsies were used for comprehensive immune profiling by flow cytometry, multicytokine array, and gene expression in 3 segments of the stomach and the duodenal bulb. Associations of immune endpoints with clinical parameters of gastroparesis were also explored. RESULTS: The gastric mucosa displayed large regional variation of distinct immune profiles. Furthermore, several-fold increases in innate and adaptive immune cells were found in gastroparesis. Various immune cell types showed positive correlations with duration of disease, proton pump inhibitor dosing, and delayed gastric emptying. DISCUSSION: This initial observational study showed immune compartmentalization of the human stomach mucosa and significant immune dysregulation at the level of leukocyte infiltration in idiopathic gastroparesis patients that extends to the duodenum. Select immune cells, such as macrophages, may correlate with clinicopathological traits of gastroparesis. This work supports further mucosal studies to advance our understanding of gastroparesis pathophysiology.


Subject(s)
Gastric Mucosa/immunology , Gastroparesis/immunology , Adaptive Immunity , Adolescent , Adult , Aged , CD8 Antigens , Case-Control Studies , Cross-Sectional Studies , Cytokines/blood , Duodenum/immunology , Female , Gastric Emptying , Gastroparesis/physiopathology , Gene Expression , Humans , Immunity, Innate , Intestinal Mucosa/immunology , Macrophages/immunology , Male , Middle Aged , Prospective Studies , T-Lymphocytes/immunology , Young Adult
14.
Gastroenterology ; 161(2): 637-652.e4, 2021 08.
Article in English | MEDLINE | ID: mdl-33971182

ABSTRACT

BACKGROUND & AIMS: The immune compartment is critical for maintaining tissue homeostasis. A weak immune response increases susceptibility to infection, but immune hyperactivation causes tissue damage, and chronic inflammation may lead to cancer development. In the stomach, inflammation damages the gastric glands and drives the development of potentially preneoplastic metaplasia. Glucocorticoids are potent anti-inflammatory steroid hormones that are required to suppress gastric inflammation and metaplasia. However, these hormones function differently in males and females. Here, we investigate the impact of sex on the regulation of gastric inflammation. METHODS: Endogenous glucocorticoids and male sex hormones were removed from mice using adrenalectomy and castration, respectively. Mice were treated with 5α-dihydrotestosterone (DHT) to test the effects of androgens on regulating gastric inflammation. Single-cell RNA sequencing of gastric leukocytes was used to identify the leukocyte populations that were the direct targets of androgen signaling. Type 2 innate lymphoid cells (ILC2s) were depleted by treatment with CD90.2 antibodies. RESULTS: We show that adrenalectomized female mice develop spontaneous gastric inflammation and spasmolytic polypeptide-expressing metaplasia (SPEM) but that the stomachs of adrenalectomized male mice remain quantitatively normal. Simultaneous depletion of glucocorticoids and sex hormones abolished the male-protective effects and triggered spontaneous pathogenic gastric inflammation and SPEM. Treatment of female mice with DHT prevented gastric inflammation and SPEM development when administered concurrent with adrenalectomy and also reversed the pathology when administered after disease onset. Single-cell RNAseq of gastric leukocytes revealed that ILC2s expressed abundant levels of both the glucocorticoid receptor (Gr) and androgen receptor (Ar). We demonstrated that DHT treatment potently suppressed the expression of the proinflammatory cytokines Il13 and Csf2 by ILC2s. Moreover, ILC2 depletion protected the stomach from SPEM development. CONCLUSIONS: Here, we report a novel mechanism by which glucocorticoids and androgens exert overlapping effects to regulate gastric inflammation. Androgen signaling within ILC2s prevents their pathogenic activation by suppressing the transcription of proinflammatory cytokines. This work revealed a critical role for sex hormones in regulating gastric inflammation and metaplasia.


Subject(s)
Androgens/pharmacology , Anti-Inflammatory Agents/pharmacology , Dihydrotestosterone/pharmacology , Gastric Mucosa/drug effects , Gastritis, Atrophic/metabolism , Glucocorticoids/metabolism , Gonadal Steroid Hormones/metabolism , Lymphocytes/drug effects , Adrenalectomy , Animals , Cellular Microenvironment , Disease Models, Animal , Disease Susceptibility , Female , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis, Atrophic/immunology , Gastritis, Atrophic/pathology , Gastritis, Atrophic/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Metaplasia , Mice, Inbred C57BL , Orchiectomy , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Sex Factors , Signal Transduction , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism
15.
J Immunol Res ; 2021: 5580672, 2021.
Article in English | MEDLINE | ID: mdl-33855088

ABSTRACT

METHODS: Monoamine neurotransmitters were detected in gastric cancer tissue and paired normal tissue, and The Cancer Genome Atlas was used to identify differentially expressed norepinephrine-degrading and synthetic enzymes. Quantitative real-time PCR and the Seahorse assay were used to determine the effect of norepinephrine on gastric cancer cell glycolysis. MAOA expression in cancer tissues was analyzed by immunohistochemistry and was compared with the patient SUVmax value of PET-CT and other clinicopathological characteristics. RESULTS: The norepinephrine levels were markedly high in gastric cancer tissue, while the norepinephrine-degrading enzymes MAOA and MAOB showed low expression. High norepinephrine levels were associated with activated glycolysis. The MAOA or MAOB expression levels in tumor tissue were closely correlated with the patient SUV max values of PET-CT and immunotherapy evaluation indices, such as PD-L1 and the microsatellite status. CONCLUSIONS: Norepinephrine shows relatively higher expression in gastric cancer tissue than in normal tissue, and its expression level is associated with the glycolysis levels in patients. The norepinephrine-degrading enzymes MAOA and MAOB have significant expression differences in cancer and normal tissue, and their missing or low expression may predict immune therapy outcomes for gastric cancer patients. High norepinephrine levels with metabolic abnormalities may be more suitable for metabolic targeted therapy or immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Norepinephrine/metabolism , Stomach Neoplasms/immunology , Warburg Effect, Oncologic , Antimetabolites, Antineoplastic , Antineoplastic Agents, Immunological , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Gastric Mucosa/enzymology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Humans , Immunohistochemistry , Monoamine Oxidase/analysis , Monoamine Oxidase/metabolism , Norepinephrine/analysis , Positron Emission Tomography Computed Tomography , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
16.
J Tradit Chin Med ; 41(2): 270-275, 2021 04.
Article in English | MEDLINE | ID: mdl-33825407

ABSTRACT

OBJECTIVE: To investigate the efficacy of the extract from Ononis spinosa L. (O. spinosa) on ethanol-induced gastric ulcer in rats. METHODS: Phytochemical constituents of the extract from O. spinosa were analyzed using liquid chromatography-mass spectrometry. Rats were classified into 4 equal groups; ulcer control received oral vehicle; positive control was administered with 40 mg/kg esomeprazole (standard drug) and 2 groups received 0.5 and 1 g/kg of O. spinosa extract, respectively. Gastric ulcer was induced by absolute ethanol (5 mL/kg) orally to all groups. Measurement of ulcer index, cyclooxygenase-2 (COX-2) expression and determination of total glutathione level in gastric mucosa were performed. RESULTS: Oral administration of the extract from O. spinosa at doses 0.5 and 1 g/kg lowered the ulcer indices by 80.39% and 98.71% , respectively, compared to 67.89% by esomeprazole (40 mg/kg). Histologically, treatment with the extract decreased necrosis and hemorrhage in mucosa and edema and infiltration by inflammatory cells in submucosa. Using immunohistochemical technique, it was demonstrated that COX-2 expression increased in mucosa of animals treated with the extract as well as by esomeprazole. O. spinosa and esomeprazole increased total glutathione level in the stomach compared to control. Ononin was the major compound of the extract followed by trifolirhizin, myricitrin, gentisic acid, cycloartenol and quercetin. CONCLUSION: The present study demonstrated that the extract from O. spinosa was able to protect gastric mucosa from ethanol injury by at least 2 mechanisms, namely the induction of COX-2 and decreasing oxidative stress in the stomach.


Subject(s)
Ononis/chemistry , Plant Extracts/administration & dosage , Stomach Ulcer/drug therapy , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Ethanol/adverse effects , Female , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Humans , Phytotherapy , Plant Extracts/isolation & purification , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Stomach Ulcer/genetics , Stomach Ulcer/immunology
17.
Front Immunol ; 12: 625586, 2021.
Article in English | MEDLINE | ID: mdl-33841407

ABSTRACT

Background: Helicobacter pylori infection is the main cause of chronic gastritis in children. Little is known about the effect of Helicobacter pylori on microbiota and immunity. This study was aimed at characterizing stomach microbiota and immune-regulatory properties of children with Helicobacter pylori colonization. Methods: We studied 122 children who had undergone gastric endoscopy due to gastrointestinal symptoms, 57 were diagnosed with Helicobacter pylori infection. Endoscopic mucosal biopsy samples were obtained for DNA and RNA extraction. Microbiomes were analyzed by 16S rRNA profiling, with the differentially expressed genes analyzed using RNA sequencing. The RNA-sequencing results of selected genes were validated by qRT-PCR. Results: Bacterial diversity of Helicobacter pylori-positive gastric specimens were lower than those of negative, and both groups were clearly separated according to beta diversity. Helicobacter pylori-positive group significantly reduced proportions of six phyla and eight genera; only Helicobacter taxa were more abundant in Helicobacter pylori-negative group. Gastric tissues RNA sequencing showed increased expression of multiple immune response genes in Helicobacter pylori -infection. Helicobacter pylori -infected children with restructured gastric microbiota had higher levels of FOXP3, IL-10, TGF-ß1 and IL-17A expressions, which were consistent with increased CD4+T cell and macrophagocyte, compared with non-infected children. Conclusions: Presence of Helicobacter pylori significantly influences gastric microbiota and results in lower abundance of multiple taxonomic levels in children. Meanwhile, it affects gastric immune environment and promotes the occurrence of gastritis. Clinical Trial Registration: [http://www.chictr.org.cn], identifier [ChiCTR1800015190].


Subject(s)
Duodenum/microbiology , Gastric Mucosa/microbiology , Gastritis/microbiology , Gastrointestinal Microbiome , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Immunity, Mucosal , Intestinal Mucosa/microbiology , Adolescent , Age Factors , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Case-Control Studies , Child , Duodenum/immunology , Dysbiosis , Endoscopy, Gastrointestinal , Female , Forkhead Transcription Factors/analysis , Gastric Mucosa/immunology , Gastritis/diagnosis , Gastritis/immunology , Helicobacter Infections/diagnosis , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Host-Pathogen Interactions , Humans , Interleukin-10/analysis , Interleukin-17/analysis , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/microbiology , Male , Ribotyping , Transforming Growth Factor beta1/analysis
18.
Front Immunol ; 12: 626017, 2021.
Article in English | MEDLINE | ID: mdl-33777009

ABSTRACT

Mucosal associated invariant T (MAIT) cells play a critical role in Helicobacter pylori (H. pylori)-induced gastritis by promoting mucosal inflammation and aggravating mucosal injuries (1, 2). However, the underlying mechanism and key molecules involved are still uncertain. Here we identified OX40, a co-stimulatory molecule mainly expressed on T cells, as a critical regulator to promote proliferation and IL-9 production by MAIT cells and facilitate mucosal inflammation in H. pylori-positive gastritis patients. Serum examination revealed an increased level of IL-9 in gastritis patients. Meanwhile, OX40 expression was increased in mucosal MAIT cells, and its ligand OX40L was also up-regulated in mucosal dendritic cells (DCs) of gastritis patients, compared with healthy controls. Further results demonstrated that activation of the OX40/OX40L pathway promoted IL-9 production by MAIT cells, and MAIT cells displayed a highly-activated phenotype after the cross-linking of OX40 and OX40L. Moreover, the level of IL-9 produced by MAIT cells was positively correlated with inflammatory indexes in the gastric mucosa, suggesting the potential role of IL-9-producing MAIT cells in mucosal inflammation. Taken together, we elucidated that OX40/OX40L axis promoted mucosal MAIT cell proliferation and IL-9 production in H. pylori-induced gastritis, which may provide potential targeting strategies for gastritis treatment.


Subject(s)
Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Interleukin-9/metabolism , Mucosal-Associated Invariant T Cells/microbiology , OX40 Ligand/metabolism , Receptors, OX40/metabolism , Adult , Case-Control Studies , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastritis/immunology , Gastritis/metabolism , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Host-Pathogen Interactions , Humans , Immunity, Mucosal , Lymphocyte Activation , Male , Middle Aged , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Phenotype , Signal Transduction , Young Adult
19.
Front Immunol ; 12: 619246, 2021.
Article in English | MEDLINE | ID: mdl-33717114

ABSTRACT

Primary immunodeficiency (PID) with immune dysregulation may present with early onset gastrointestinal autoimmune disorders. When gastrointestinal autoimmunity is associated with multiple extraintestinal immune system dysfunction the diagnosis of PID is straightforward. However, with the advent of next generation sequencing technologies, genetic defects in PID genes have been increasingly recognized even when a single or no extraintestinal signs of immune dysregulation are present. A genetic diagnosis is especially important considering the expanding armamentarium of therapies designed to inhibit specific molecular pathways. We describe a boy with early-onset severe, refractory autoimmune gastritis and biallelic mutations in the LRBA gene causing a premature STOP-codon who was successfully treated with CTLA4-Ig, abatacept, with long term clinical and endoscopic remission. The case underscores the importance to consider a monogenetic defect in early onset autoimmune disorders, since the availability of targeted treatments may significantly improve patient prognosis.


Subject(s)
Abatacept/therapeutic use , Adaptor Proteins, Signal Transducing/deficiency , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/etiology , Gastritis/drug therapy , Gastritis/etiology , Immunosuppressive Agents/therapeutic use , Biomarkers , Biopsy , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Disease Management , Disease Susceptibility , Drug Resistance , Endoscopes, Gastrointestinal , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/diagnosis , Humans , Male , Retreatment , Treatment Outcome
20.
Methods Mol Biol ; 2283: 215-223, 2021.
Article in English | MEDLINE | ID: mdl-33765320

ABSTRACT

Analysis of human and mouse T-helper (Th) cell subset responses is key to understanding how the inflammatory response in the stomach is regulated, and links with pathology and disease outcomes. Here, we present methods for extracting cells from blood and tissue, and analyzing the balance of Th1, Th2, Th17, and regulatory T cell (Treg) subsets using flow cytometry and cytokine ELISA .


Subject(s)
Helicobacter Infections/immunology , Helicobacter pylori/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cytokines/metabolism , Flow Cytometry , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter pylori/pathogenicity , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...