Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Microbiol Spectr ; 12(6): e0001524, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38682907

ABSTRACT

Gastric cancer is the fifth most diagnosed cancer in the world. Infection by the bacteria Helicobacter pylori (HP) is associated with approximately 75% of gastric cancer cases. HP infection induces chronic gastric inflammation, damaging the stomach and fostering carcinogenesis. Most mechanistic studies on gastric cancer initiation are performed in mice and utilize either mouse-adapted strains of HP or the natural mouse pathogen Helicobacter felis (HF). Here, we identified the differences in gastric inflammation, atrophy, and metaplasia associated with HP and HF infection in mice. PMSS1 HP strain or the CS1 HF strain were co-cultured with mouse peritoneal macrophages to assess their immunostimulatory effects. HP and HF induced similar cytokine production from cultured mouse peritoneal macrophages revealing that both bacteria exhibit similar immunostimulatory effects in vitro. Next, C57BL/6J mice were infected with HP or HF and were assessed 2 months post-infection. HP-infected mice caused modest inflammation within both the gastric corpus and antrum, and did not induce significant atrophy within the gastric corpus. In contrast, HF induced significant inflammation throughout the gastric corpus and antrum. Moreover, HF infection was associated with significant atrophy of the chief and parietal cell compartments and induced the expression of pyloric metaplasia (PM) markers. HP is poorly immunogenic compared to HF. HF induces dramatic CD4+ T cell activation, which is associated with increased gastric cancer risk in humans. Thus, HP studies in mice are better suited for studies on colonization, while HF is more strongly suited for studies on the effects of gastric inflammation on tumorigenesis. . IMPORTANCE: Mouse infection models with Helicobacter species are widely used to study Helicobacter pathogenesis and gastric cancer initiation. However, Helicobacter pylori is not a natural mouse pathogen, and mouse-adapted H. pylori strains are poorly immunogenic. In contrast, Helicobacter felis is a natural mouse pathogen that induces robust gastric inflammation and is often used in mice to investigate gastric cancer initiation. Although both bacterial strains are widely used, their disease pathogenesis in mice differs dramatically. However, few studies have directly compared the pathogenesis of these bacterial species in mice, and the contrasting features of these two models are not clearly defined. This study directly compares the gastric inflammation, atrophy, and metaplasia development triggered by the widely used PMSS1 H. pylori and CS1 H. felis strains in mice. It serves as a useful resource for researchers to select the experimental model best suited for their studies.


Subject(s)
Gastric Mucosa , Helicobacter Infections , Helicobacter felis , Helicobacter pylori , Metaplasia , Mice, Inbred C57BL , Animals , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter Infections/immunology , Mice , Helicobacter felis/pathogenicity , Metaplasia/microbiology , Metaplasia/pathology , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/immunology , Gastritis/microbiology , Gastritis/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Inflammation/microbiology , Inflammation/pathology , Female , Cytokines/metabolism , Disease Models, Animal , Stomach/pathology , Stomach/microbiology
2.
Front Immunol ; 14: 1147695, 2023.
Article in English | MEDLINE | ID: mdl-37744359

ABSTRACT

Introduction: Gastric myeloid-derived suppressor cells (MDSCs) are a prominent population that expands during gastric pre-neoplastic and neoplastic development in humans and mice. However, the heterogeneity of this population has circumvented the ability to study these cells or understand their functions. Aside from Schlafen-4+ (Slfn-4+) MDSCs in mouse studies, which constitute a subset of this population, limitations exist in characterizing the heterogeneity of the gastric CD11b+Ly6G+ population and targeting its different subsets. Here we identify S100a8 as a pan-specific marker for this population and utilize it to study the role of the S100a8+Cxcr2+ subset. Methods: We profiled gastric CD11b+Ly6G+ versus CD11b+Ly6G- myeloid cells by transcriptomic and single-cell RNA sequencing. We identified S100a8 as a pan-specific marker of the gastric granulocytic MDSC (G-MDSC) population, and generated S100a8CreCxcr2flox/flox to study the effects of Cxcr2 knockdown. Results: Following 6-months of Helicobacter felis infection, gastric CD11b+Ly6G+ G-MDSCs were highly enriched for the expression of S100a8, S100a9, Slfn4, Cxcr2, Irg1, Il1f9, Hcar2, Retnlg, Wfdc21, Trem1, Csf3R, Nlrp3, and Il1b. The expression of these distinct genes following 6mo H. felis infection marked heterogeneous subpopulations, but they all represented a subset of S100a8+ cells. S100a8 was identified as a pan-marker for CD11b+Ly6G+ cells arising in chronic inflammation, but not neutrophils recruited during acute gut infection. 6mo Helicobacter felis-infected S100a8CreCxcr2flox/flox mice exhibited worsened gastric metaplastic pathology than Cxcr2flox/flox mice, which was associated with dysregulated lipid metabolism and peroxidation. Conclusion: S100a8 is a pan-specific marker that can be used to target gastric G-MDSC subpopulations, of which the Cxcr2+ subset regulates gastric immunopathology and associates with the regulation of lipid peroxidation.


Subject(s)
Myeloid-Derived Suppressor Cells , Stomach Neoplasms , Animals , Mice , Calgranulin A/genetics , Helicobacter felis , Myeloid Cells
3.
Microb Pathog ; 159: 105151, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34450200

ABSTRACT

CD4+ CD8αα+ double-positive intraepithelial T lymphocytes (DP T cells), a newly characterized subset of intraepithelial T cells, are reported to contribute to local immunosuppression. However, the presence of DP T cells in Helicobacter. pylori -induced gastritis and their relationship with disease prognosis has yet to be elucidated. In this study, a chronic gastritis model was established by infecting mice with Helicobacter felis. Gastric-infiltrating lymphocytes were isolated from these mice and analyzed by flow cytometry. The frequency of DP T cells in H. felis-induced gastritis mice was higher than that in uninfected mice. The gastric DP T cells were derived from lamina propria cells but were predominantly distributed in the gastric epithelial layer. These gastric DP T cells also exhibited anti-inflammatory functions, and they inhibited the maturation of dendritic cells and proliferation of CD4+ T lymphocytes in vitro. Elimination of DP T cells simultaneously resulted in severe gastritis and a reduction of H. felis load in vivo. Finally, vaccine mixed with different adjuvants was used to explore the relationship between vaccine efficacy and DP cells. Silk fibroin as the vaccine delivery system enhanced vaccine efficacy by reducing the number of DP T cells. This study demonstrated that DP T cells perform an immunosuppressive role in Helicobacter felis-induced gastritis, and consequently, DP T cells may affect disease prognosis and vaccine efficacy.


Subject(s)
Helicobacter felis , T-Lymphocytes , Animals , CD4-Positive T-Lymphocytes , Epithelium , Gastric Mucosa , Inflammation , Mice
4.
Turk J Biol ; 45(2): 214-224, 2021.
Article in English | MEDLINE | ID: mdl-33907502

ABSTRACT

Regulatory B cells (Bregs) produce antiinflammatory cytokines and inhibits proinflammatory response. Recently, immunosuppressive roles of Bregs in the effector functions of dendritic cells (DCs) were demonstrated. However, cross talk between Bregs and DCs in Helicobacter infection remains unknown. Here, we showed that direct stimulation of bone marrow-derived DCs (BM-DCs) with Helicobacter felis (H. felis) antigen upregulates their CD86 surface expression and causes the production of interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), interleukin-12 (IL-12), and interleukin-10 (IL-10). Furthermore, prestimulation of DCs with supernatants derived from both Helicobacter-stimulated IL-10- B (Hf stim -IL-10- B) or IL-10+ B (Hf stim -IL-10+) cells suppresses the secretion of TNF-α and IL-6, but does not affect the expression of CD86 and secretion of IL-12 by lipopolysaccharide (LPS) or H. felis-activated BM-DCs. Remarkably, soluble factors secreted by Hf stim -IL-10- B cells, but not by Hf stim -IL-10+ B cells, suppress the secretion of IL-10 by BM-DCs upon subsequent LPS stimulation. In contrast, prestimulation with BM-DCs with supernatants of Hf stim -IL-10+ B cells before H. felis antigen stimulation induces significantly their IL-10 production. Collectively, our data indicated that prestimulation with soluble factors secreted by Hf stim -IL-10+ B cells, DCs exhibit a tolerogenic phenotype in response to LPS or Helicobacter antigen by secreting high levels of IL-10, but decreased levels of IL-6 and TNF-α.

5.
Gut ; 70(4): 654-665, 2021 04.
Article in English | MEDLINE | ID: mdl-32709613

ABSTRACT

BACKGROUND AND AIMS: The gastric epithelium undergoes continuous turnover. Corpus epithelial stem cells located in the gastric isthmus serve as a source of tissue self-renewal. We recently identified the transcription factor Mist1 as a marker for this corpus stem cell population that can give rise to cancer. The aim here was to investigate the regulation of the Mist1+ stem cells in the response to gastric injury and inflammation. METHODS: We used Mist1CreERT;R26-Tdtomato mice in two models of injury and inflammation: the acetic acid-induced ulcer and infection with Helicobacter felis. We analysed lineage tracing at both early (7 to 30 days) and late (30 to 90 days) time points. Mist1CreERT;R26-Tdtomato;Lgr5DTR-eGFP mice were used to ablate the corpus basal Lgr5+ cell population. Constitutional and conditional Wnt5a knockout mice were used to investigate the role of Wnt5a in wound repair and lineage tracing from the Mist1+ stem cells. RESULTS: In both models of gastric injury, Mist1+ isthmus stem cells more rapidly proliferate and trace entire gastric glands compared with the normal state. In regenerating tissue, the number of traced gastric chief cells was significantly reduced, and ablation of Lgr5+ chief cells did not affect Mist1-derived lineage tracing and tissue regeneration. Genetic deletion of Wnt5a impaired proliferation in the gastric isthmus and lineage tracing from Mist1+ stem cells. Similarly, depletion of innate lymphoid cells, the main source of Wnt5a, also resulted in reduced proliferation and Mist1+ isthmus cell tracing. CONCLUSION: Gastric Mist1+ isthmus cells are the main supplier of regenerated glands and are activated in part through Wnt5a pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chief Cells, Gastric/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Cell Proliferation , Inflammation/metabolism , Mice , Mice, Knockout , Stomach Ulcer/metabolism , Wound Healing
6.
Emerg Microbes Infect ; 9(1): 2289-2302, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33000989

ABSTRACT

Tissue-resident memory T (TRM) cells, located in the epithelium of most peripheral tissues, constitute the first-line defense against pathogen infections. Our previous study reported that gastric subserous layer (GSL) vaccination induced a "pool" of protective tissue-resident memory CD4+T (CD4+TRM) cells in the gastric epithelium. However, the mechanistic details how CD4+TRM cells form in the gastric epithelium are unknown. Here, our results suggested that the vaccine containing CCF in combination with Silk fibroin hydrogel (SF) broadened the distribution of gastric intraepithelial CD4+TRM cells. It was revealed that the gastric intraepithelial TRM cells were even more important than circulating memory T cells against infection by Helicobacter felis. It was also shown that gastric-infiltrating neutrophils were involved as indispensable mediators which secreted CXCL10 to chemoattract CXCR3+CD4+T cells into the gastric epithelium. Blocking of CXCR3 or neutrophils significantly decreased the number of gastric intraepithelial CD4+TRM cells due to reduced recruitment of CD4+T cells. This study demonstrated the protective efficacy of gastric CD4+TRM cells against H. felis infection, and highlighted the influence of neutrophils on gastric intraepithelial CD4+TRM cells formation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Fibroins/administration & dosage , Helicobacter Infections/prevention & control , Helicobacter felis/immunology , Animals , Bacterial Vaccines , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL10/genetics , Fibroins/chemistry , Fibroins/immunology , Gastric Mucosa/immunology , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Hydrogels , Immunologic Memory , Mice , Neutrophils/metabolism , Receptors, CXCR3/genetics
7.
Helicobacter ; 25(6): e12763, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33025641

ABSTRACT

BACKGROUND: Helicobacter pylori infection leads to regulatory T-cell (Treg) induction in infected mice, which contributes to H. pylori immune escape. However, the mechanisms responsible for H. pylori induction of Treg and immune tolerance remain unclear. We hypothesized DC-produced TGF-ß may be responsible for Treg induction and immune tolerance. MATERIALS AND METHODS: To test this hypothesis, we generated TGF-ß∆DC mice (CD11c+ DC-specific TGF-ß deletion) and assessed the impact of DC-specific TGF-ß deletion on DC function during Helicobacter infection in vitro and in vivo. To examine the T cell-independent DC function, we crossed TGF-ß∆DC mice onto Rag1KO background to generate TGF-ß∆DC xRag1KO mice. RESULTS: When stimulated with H. pylori, TGF-ß∆DC BMDC/splenocyte cocultures showed increased levels of proinflammatory cytokines and decreased levels of anti-inflammatory cytokines compared to control, indicating a proinflammatory DC phenotype. Following 6 months of H. felis infection, TGF-ß∆DC mice developed more severe gastritis and a trend toward more metaplasia compared to TGF-ßfl/fl with increased levels of inflammatory Th1 cytokine mRNA and lower gastric H. felis colonization compared to infected TGF-ßfl/fl mice. In a T cell-deficient background using TGF-ß∆DC xRag1KO mice, H. felis colonization was significantly lower when DC-derived TGF-ß was absent, revealing a direct, innate function of DC in controlling H. felis infection independent of Treg induction. CONCLUSIONS: Our findings indicate that DC-derived TGF-ß mediates Helicobacter-induced Treg response and attenuates the inflammatory Th1 response. We also demonstrated a previously unrecognized innate role of DC controlling Helicobacter colonization via a Treg-independent mechanism. DC TGF-ß signaling may represent an important target in the management of H. pylori.


Subject(s)
Dendritic Cells/immunology , Helicobacter Infections/immunology , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Gastric Mucosa , Helicobacter pylori , Mice , Mice, Inbred C57BL
8.
Gut ; 69(10): 1750-1761, 2020 10.
Article in English | MEDLINE | ID: mdl-31980446

ABSTRACT

The myeloid differentiation factor Schlafen4 (Slfn4) marks a subset of myeloid-derived suppressor cells (MDSCs) in the stomach during Helicobacter-induced spasmolytic polypeptide-expressing metaplasia (SPEM). OBJECTIVE: To identify the gene products expressed by Slfn4+-MDSCs and to determine how they promote SPEM. DESIGN: We performed transcriptome analyses for both coding genes (mRNA by RNA-Seq) and non-coding genes (microRNAs using NanoString nCounter) using flow-sorted SLFN4+ and SLFN4- cells from Helicobacter-infected mice exhibiting metaplasia at 6 months postinfection. Thioglycollate-elicited myeloid cells from the peritoneum were cultured and treated with IFNα to induce the T cell suppressor phenotype, expression of MIR130b and SLFN4. MIR130b expression in human gastric tissue including gastric cancer and patient sera was determined by qPCR and in situ hybridisation. Knockdown of MiR130b in vivo in Helicobacter-infected mice was performed using Invivofectamine. Organoids from primary gastric cancers were used to generate xenografts. ChIP assay and Western blots were performed to demonstrate NFκb p65 activation by MIR130b. RESULTS: MicroRNA analysis identified an increase in MiR130b in gastric SLFN4+ cells. Moreover, MIR130b colocalised with SLFN12L, a human homologue of SLFN4, in gastric cancers. MiR130b was required for the T-cell suppressor phenotype exhibited by the SLFN4+ cells and promoted Helicobacter-induced metaplasia. Treating gastric organoids with the MIR130b mimic induced epithelial cell proliferation and promoted xenograft tumour growth. CONCLUSION: Taken together, MiR130b plays an essential role in MDSC function and supports metaplastic transformation.


Subject(s)
Carrier Proteins/metabolism , Helicobacter Infections , MicroRNAs/metabolism , Stomach Neoplasms , Animals , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori/physiology , Interferon-alpha/metabolism , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/metabolism , Precancerous Conditions , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
9.
Lipids ; 53(4): 393-401, 2018 04.
Article in English | MEDLINE | ID: mdl-29766504

ABSTRACT

Helicobacter pylori infects the human stomach and is closely linked with the development of gastric cancer. When detected, this pathogen can be eradicated from the human stomach using wide-spectrum antibiotics. However, year by year, H. pylori strains resistant to the antibacterial action of antibiotics have been increasing. The development of new antibacterial substances effective against drug-resistant H. pylori is urgently required. Our group has recently identified extremely selective bactericidal effects against H. pylori in (1R,3aR,7aR)-1-[(1R)-1,5-dimethylhexyl]octahydro-7a-methyl-4H-inden-4-one (VDP1) (otherwise known as Grundmann's ketone), an indene compound derived from the decomposition of vitamin D3 and proposed the antibacterial mechanism whereby VDP1 induces the bacteriolysis by interacting at least with PtdEtn (dimyristoyl-phosphatidylethanolamine [di-14:0 PtdEtn]) retaining two 14:0 fatty acids of the membrane lipid constituents. In this study, we synthesized new indene compounds ((1R,3aR,7aR)-1-((2R,E)-5,6-dimethylhept-3-en-2-yl)-7a-methyloctahydro-4H-inden-4-one [VD2-1], (1R,3aR,7aR)-1-((S)-1-hydroxypropan-2-yl)-7a-methyloctahydro-1H-inden-4-ol [VD2-2], and (1R,3aR,7aR)-7a-methyl-1-((R)-6-methylheptan-2-yl)octahydro-1H-inden-4-ol [VD3-1]) using either vitamin D2 or vitamin D3 as materials. VD2-1 and VD3-1 selectively disrupted the di-14:0 PtdEtn vesicles without destructing the vesicles of PtdEtn (dipalmitoyl-phosphatidylethanolamine) retaining two 16:0 fatty acids. In contrast, VD2-2, an indene compound lacking an alkyl group, had no influence on the structural stability of both PtdEtn vesicles. In addition, VD2-1 and VD3-1 exerted extremely selective bactericidal action against H. pylori without affecting the viability of commonplace bacteria. Meanwhile, VD2-2 almost forfeited the bactericidal effects on H. pylori. These results suggest that the alkyl group of the indene compounds has a crucial conformation to interact with di-14:0 PtdEtn of H. pylori membrane lipid constituents whereby the bacteriolysis is ultimately induced.


Subject(s)
Anti-Bacterial Agents/pharmacology , Helicobacter pylori/drug effects , Indenes/pharmacology , Vitamin D/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Indenes/chemical synthesis , Indenes/chemistry , Microbial Sensitivity Tests , Molecular Conformation , Structure-Activity Relationship , Vitamin D/chemistry
10.
Mol Immunol ; 96: 48-60, 2018 04.
Article in English | MEDLINE | ID: mdl-29494848

ABSTRACT

Regulatory B cells (Bregs) play a crucial role in immunological tolerance primarily through the production of IL-10 in many diseases including autoimmune disorders, allergy, infectious diseases, and cancer. To date, various Breg subsets with overlapping phenotypes have been identified. However, the roles of Bregs in Helicobacter infection are largely unknown. In the present study, we investigate the phenotype and function of Helicobacter -stimulated B cells. Our results demonstrate that Helicobacter felis -stimulated IL-10- producing B cells (Hfstim- IL-10+ B) are composed of B10 and Transitional 2 Marginal Zone Precursor (T2-MZP) cells with expression of CD9, Tim-1, and programmed death 1 (PD-1). On the other hand, Helicobacter felis -stimulated IL-10- nonproducing B (Hfstim- IL-10- B) cells are mainly marginal zone (MZ) B cells that express PD-L1 and secrete TGF-ß, IL-6, and TNF-α, and IgM and IgG2b. Furthermore, we show that both Hfstim- IL-10+ B cells and Hfstim- IL-10- B cells induce CD49b+LAG-3+ Tr1 cells. Here, we describe a novel mechanism for PD-1/PD-L1- driven B cell-dependent Tr1 cell differentiation. Finally, we explore the capability of Hfstim- IL-10- B cells to induce Th17 cell differentiation, which we find to be dependent on TGF-ß. Taken together, the current study demonstrates that Hfstim- B cells induce Tr1 cells through the PD-1/PD-L1 axis and Th17 cells by secreting TGF-ß.


Subject(s)
B-Lymphocyte Subsets/immunology , Helicobacter Infections/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , B7-H1 Antigen/immunology , Cell Differentiation/immunology , Helicobacter felis , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology
11.
J Cancer Prev ; 21(3): 182-186, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27722144

ABSTRACT

BACKGROUND: The aim of this study was to investigate the effect of N-methyl-N-nitrosourea (MNU) treatment followed by chronic Helicobacter pylori SS1 and H. felis colonization on the stomachs of C57BL/6 mice. The role of MNU and Helicobacter species in gastric carcinogenesis was also elucidated. METHODS: A total of 69 C57BL/6 mice at 4 weeks of age were divided into 6 groups according to MNU treatment and H. pylori SS1 or H. felis infection. The mice were sacrificed at 21 and 50 weeks. The degree of inflammation was determined by histopathology. The levels of gastric mucosal myeloperoxidase, TNF-α, and interleukin-1ß (IL-1ß) were measured by ELISA. RESULTS: In the H. felis groups with or without MNU, the incidence of gastric tumors was 21.1% and 35.0% at 21 and 50 weeks, respectively. No gastric tumors were observed in all control mice. At 50 weeks, 37.5% of gastric adenoma cases were observed in the H. felis alone and MNU + H. felis groups. Furthermore, 12.5% of gastric adenocarcinoma cases were observed in the MNU alone and MNU + H. felis groups. The gastric mucosal IL-1ß level was significantly higher in the MNU + H. felis group at 21 weeks and H. felis group at 50 weeks, respectively, than that for control mice (P < 0.05). However, the effect of MNU on H. pylori SS1-induced gastric carcinogenesis was low compared to that on H. felis. CONCLUSIONS: Administration of MNU before H. felis infection provokes severe inflammation through IL-1ß, and eventually induces gastric cancer. However, the role of MNU in H. pylori SS1-induced gastric carcinogenesis model is minor.

12.
Lab Anim Res ; 32(2): 122-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27382381

ABSTRACT

A 3-month-old male cat in the animal facility was presented for investigation of anorexia and occasional vomiting. We collected the specimens from gastroscopic biopsy and stool collection. The gastroscopic biopsy specimens were tested using a rapid urease test, CLO Helicobacter-detection kits. Stool specimens were gathered and evaluated using the commercially available SD Bioline H. pylori Ag kit according to the manufacturer's instructions. Genomic DNAs from gastroscopic biopsy and stool specimens of the cat were extracted and submitted to the consensus PCR to amplify Helicobacter rpoB gene. Then the DNAs from gastroscopic biopsy and stool specimens were conducted a multiplex species-specific PCR to amplify urease B gene for H. heilmannii, H. pylori and H. felis. As the results, the rapid urease test with gastroscopic biopsy was revealed positive reaction. The result of H. pylori Stool Ag assay was one red line, negative for H. pylori. The gastroscopic biopsy and stool specimen were positive reactions by the consensus PCR reaction using the RNA polymerase beta-subunit-coding gene (rpoB) to detect Helicobacter species. By multiplex species-specific PCR with gastroscopic biopsy and stool specimens, no amplification products corresponding to either H. heilmannii or H. pylori were detected, but the specimens tested were positive for H. felis. This case was confirmed as gastroenteric disease induced by H. felis infection. On our knowledge, this is a very rare report about H. felis-induced gastroenteric disease in cat and may provide a valuable data on the study of feline Helicobacter infection.

13.
J Cancer Prev ; 21(1): 48-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27051649

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the anti-inflammatory and anti-tumorigenic effect of açai berry after chronic Helicobacter felis colonization in the stomachs of C57BL/6 mice. METHODS: A total of 57 four-week-old female C57BL/6 mice (18 control mice and 39 experimental mice) were used. The mice were administered orogastrically with vehicle only or vehicle containing H. felis, 5 times every other day. After inoculation of H. felis, mice were fed either a standard or an açai-containing diet and then sacrificed at 4, 24, and 52 weeks. The infection status and degree of inflammation were determined by culture and histopathology. The level of gastric mucosal myeloperoxidase (MPO), TNF-α, and interleukin-1ß (IL-1ß) were measured by ELISA. RESULTS: At 24 weeks after inoculation, mucosal atrophy and mucous metaplasia appeared in all infected mice. At 52 weeks after inoculation, dysplastic change was noted in 10%, 25%, and 50% of mice in the H. felis-control, H. felis-açai 5%, and H. felis-açai 10% groups, respectively. The neutrophil, monocyte, atrophy, and metaplasia grades of infected mice showed no significant difference among the H. felis-infected groups. H. felis-infected mice fed with açai berry showed no significant difference compared with H. felis-infected control mice in gastric mucosal MPO, TNF-α, and IL-1ß levels. CONCLUSIONS: H. felis that colonized the stomachs of C57BL/6 mice provoked inflammation, and induced mucosal atrophy, metaplasia, and dysplasia. However, açai berry did not effectively prohibit the gastric carcinogenesis which was induced by chronic H. felis infection.

14.
Geriatr Psychol Neuropsychiatr Vieil ; 14(1): 86-94, 2016 Mar.
Article in French | MEDLINE | ID: mdl-27005340

ABSTRACT

Alzheimer's disease (AD) is associated with Aß peptide and Tau protein deposits, but the initial process inducing the disease and ultimately neurodegeneration has not yet been elucidated. An infectious hypothesis is suggested by the alteration of the blood-brain barrier and the activation of neuroinflammation in the brain, which could play a role, especially in the decrease of Aß peptide clearance. Several viral or bacterial agents have been incriminated, including Helicobacter pylori. Infection by H. pylori is acquired during childhood and often lifetime persisting, inducing a chronic gastric inflammation, which remains asymptomatic in approximately 80% of cases. However H. pylori infection can induce systemic inflammation and increase homocysteine levels, contributing to worsen AD lesions. Association between H. pylori and AD is suggested by 1) epidemiologic studies, which show higher AD prevalence and more pronounced cognitive impairment in infected than in non-infected subjects; 2) experimental studies in murine models: a) in a first study we evaluated the impact of H. pylori infection on the brain of non-AD predisposed C57BL/6J mice. After an 18-month infection, H. pylori induced a significant gastric inflammation but no brain Aß deposit nor increased neuroinflammation was observed in their brain; b) we currently study the impact of Helicobacter species infection on behavior and cerebral lesions of AD transgenic (APPswe/PS1dE9) mice and their wild type littermate. The results of these studies do not allow to conclude a significant association between AD and H. pylory infection but may contribute to a better understanding of the role of brain neuroinflammation in AD.


Subject(s)
Alzheimer Disease/etiology , Helicobacter Infections/complications , Helicobacter pylori , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Animals , Biomarkers , Brain/pathology , Helicobacter Infections/cerebrospinal fluid , Humans , Mice , Mice, Inbred C57BL
15.
Laboratory Animal Research ; : 122-127, 2016.
Article in English | WPRIM (Western Pacific) | ID: wpr-169021

ABSTRACT

A 3-month-old male cat in the animal facility was presented for investigation of anorexia and occasional vomiting. We collected the specimens from gastroscopic biopsy and stool collection. The gastroscopic biopsy specimens were tested using a rapid urease test, CLO Helicobacter-detection kits. Stool specimens were gathered and evaluated using the commercially available SD Bioline H. pylori Ag kit according to the manufacturer's instructions. Genomic DNAs from gastroscopic biopsy and stool specimens of the cat were extracted and submitted to the consensus PCR to amplify Helicobacter rpoB gene. Then the DNAs from gastroscopic biopsy and stool specimens were conducted a multiplex species-specific PCR to amplify urease B gene for H. heilmannii, H. pylori and H. felis. As the results, the rapid urease test with gastroscopic biopsy was revealed positive reaction. The result of H. pylori Stool Ag assay was one red line, negative for H. pylori. The gastroscopic biopsy and stool specimen were positive reactions by the consensus PCR reaction using the RNA polymerase beta-subunit-coding gene (rpoB) to detect Helicobacter species. By multiplex species-specific PCR with gastroscopic biopsy and stool specimens, no amplification products corresponding to either H. heilmannii or H. pylori were detected, but the specimens tested were positive for H. felis. This case was confirmed as gastroenteric disease induced by H. felis infection. On our knowledge, this is a very rare report about H. felis-induced gastroenteric disease in cat and may provide a valuable data on the study of feline Helicobacter infection.


Subject(s)
Animals , Cats , Humans , Infant , Male , Animals, Laboratory , Anorexia , Biopsy , Consensus , DNA , DNA-Directed RNA Polymerases , Felis , Helicobacter felis , Helicobacter Infections , Helicobacter , Polymerase Chain Reaction , Stomach Diseases , Urease , Vomiting
16.
Article in English | WPRIM (Western Pacific) | ID: wpr-159296

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the anti-inflammatory and anti-tumorigenic effect of açai berry after chronic Helicobacter felis colonization in the stomachs of C57BL/6 mice. METHODS: A total of 57 four-week-old female C57BL/6 mice (18 control mice and 39 experimental mice) were used. The mice were administered orogastrically with vehicle only or vehicle containing H. felis, 5 times every other day. After inoculation of H. felis, mice were fed either a standard or an açai-containing diet and then sacrificed at 4, 24, and 52 weeks. The infection status and degree of inflammation were determined by culture and histopathology. The level of gastric mucosal myeloperoxidase (MPO), TNF-α, and interleukin-1β (IL-1β) were measured by ELISA. RESULTS: At 24 weeks after inoculation, mucosal atrophy and mucous metaplasia appeared in all infected mice. At 52 weeks after inoculation, dysplastic change was noted in 10%, 25%, and 50% of mice in the H. felis-control, H. felis-açai 5%, and H. felis-açai 10% groups, respectively. The neutrophil, monocyte, atrophy, and metaplasia grades of infected mice showed no significant difference among the H. felis-infected groups. H. felis-infected mice fed with açai berry showed no significant difference compared with H. felis-infected control mice in gastric mucosal MPO, TNF-α, and IL-1β levels. CONCLUSIONS: H. felis that colonized the stomachs of C57BL/6 mice provoked inflammation, and induced mucosal atrophy, metaplasia, and dysplasia. However, açai berry did not effectively prohibit the gastric carcinogenesis which was induced by chronic H. felis infection.


Subject(s)
Animals , Cats , Female , Humans , Mice , Atrophy , Carcinogenesis , Colon , Diet , Enzyme-Linked Immunosorbent Assay , Felis , Fruit , Helicobacter felis , Helicobacter , Inflammation , Metaplasia , Monocytes , Neutrophils , Peroxidase , Stomach
17.
Article in English | WPRIM (Western Pacific) | ID: wpr-201286

ABSTRACT

BACKGROUND: The aim of this study was to investigate the effect of N-methyl-N-nitrosourea (MNU) treatment followed by chronic Helicobacter pylori SS1 and H. felis colonization on the stomachs of C57BL/6 mice. The role of MNU and Helicobacter species in gastric carcinogenesis was also elucidated. METHODS: A total of 69 C57BL/6 mice at 4 weeks of age were divided into 6 groups according to MNU treatment and H. pylori SS1 or H. felis infection. The mice were sacrificed at 21 and 50 weeks. The degree of inflammation was determined by histopathology. The levels of gastric mucosal myeloperoxidase, TNF-α, and interleukin-1β (IL-1β) were measured by ELISA. RESULTS: In the H. felis groups with or without MNU, the incidence of gastric tumors was 21.1% and 35.0% at 21 and 50 weeks, respectively. No gastric tumors were observed in all control mice. At 50 weeks, 37.5% of gastric adenoma cases were observed in the H. felis alone and MNU + H. felis groups. Furthermore, 12.5% of gastric adenocarcinoma cases were observed in the MNU alone and MNU + H. felis groups. The gastric mucosal IL-1β level was significantly higher in the MNU + H. felis group at 21 weeks and H. felis group at 50 weeks, respectively, than that for control mice (P < 0.05). However, the effect of MNU on H. pylori SS1-induced gastric carcinogenesis was low compared to that on H. felis. CONCLUSIONS: Administration of MNU before H. felis infection provokes severe inflammation through IL-1β, and eventually induces gastric cancer. However, the role of MNU in H. pylori SS1-induced gastric carcinogenesis model is minor.


Subject(s)
Animals , Cats , Mice , Adenocarcinoma , Adenoma , Carcinogenesis , Colon , Enzyme-Linked Immunosorbent Assay , Felis , Helicobacter , Helicobacter felis , Helicobacter pylori , Incidence , Inflammation , Methylnitrosourea , Peroxidase , Stomach , Stomach Neoplasms
18.
Am J Physiol Gastrointest Liver Physiol ; 309(6): G420-30, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26159699

ABSTRACT

Helicobacter infection causes a chronic superficial gastritis that in some cases progresses via atrophic gastritis to adenocarcinoma. Proapoptotic bak has been shown to regulate radiation-induced apoptosis in the stomach and colon and also susceptibility to colorectal carcinogenesis in vivo. Therefore we investigated the gastric mucosal pathology following H. felis infection in bak-null mice at 6 or 48 wk postinfection. Primary gastric gland culture from bak-null mice was also used to assess the effects of bak deletion on IFN-γ-, TNF-α-, or IL-1ß-induced apoptosis. bak-null gastric corpus glands were longer, had increased epithelial Ki-67 expression, and contained fewer parietal and enteroendocrine cells compared with the wild type (wt). In wt mice, bak was expressed at the luminal surface of gastric corpus glands, and this increased 2 wk post-H. felis infection. Apoptotic cell numbers were decreased in bak-null corpus 6 and 48 wk following infection and in primary gland cultures following cytokine administration. Increased gastric epithelial Ki-67 labeling index was observed in C57BL/6 mice after H. felis infection, whereas no such increase was detected in bak-null mice. More severe gastric atrophy was observed in bak-null compared with C57BL/6 mice 6 and 48 wk postinfection, and 76% of bak-null compared with 25% of C57BL/6 mice showed evidence of gastric dysplasia following long-term infection. Collectively, bak therefore regulates gastric epithelial cell apoptosis, proliferation, differentiation, mucosal thickness, and susceptibility to gastric atrophy and dysplasia following H. felis infection.


Subject(s)
Cell Proliferation/genetics , Epithelium/growth & development , Helicobacter Infections/pathology , Helicobacter felis , Stomach/cytology , Stomach/pathology , bcl-2 Homologous Antagonist-Killer Protein/genetics , Animals , Atrophy , Cell Differentiation/genetics , Cytokines/pharmacology , Female , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture
19.
Nanoscale Res Lett ; 10: 35, 2015.
Article in English | MEDLINE | ID: mdl-25852332

ABSTRACT

Silver nanoparticles (AgNPs) are prominent group of nanomaterials and are recognized for their diverse applications in various health sectors. This study aimed to synthesize the AgNPs using the leaf extract of Artemisia princeps as a bio-reductant. Furthermore, we evaluated the multidimensional effect of the biologically synthesized AgNPs in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma (A549) cells. UV-visible (UV-vis) spectroscopy confirmed the synthesis of AgNPs. X-ray diffraction (XRD) indicated that the AgNPs are specifically indexed to a crystal structure. The results from Fourier transform infrared spectroscopy (FTIR) indicate that biomolecules are involved in the synthesis and stabilization of AgNPs. Dynamic light scattering (DLS) studies showed the average size distribution of the particle between 10 and 40 nm, and transmission electron microscopy (TEM) confirmed that the AgNPs were significantly well separated and spherical with an average size of 20 nm. AgNPs caused dose-dependent decrease in cell viability and biofilm formation and increase in reactive oxygen species (ROS) generation and DNA fragmentation in H. pylori and H. felis. Furthermore, AgNPs induced mitochondrial-mediated apoptosis in A549 cells; conversely, AgNPs had no significant effects on L132 cells. The results from this study suggest that AgNPs could cause cell-specific apoptosis in mammalian cells. Our findings demonstrate that this environmentally friendly method for the synthesis of AgNPs and that the prepared AgNPs have multidimensional effects such as anti-bacterial and anti-biofilm activity against H. pylori and H. felis and also cytotoxic effects against human cancer cells. This report describes comprehensively the effects of AgNPs on bacteria and mammalian cells. We believe that biologically synthesized AgNPs will open a new avenue towards various biotechnological and biomedical applications in the near future.

20.
Article in English | WPRIM (Western Pacific) | ID: wpr-77768

ABSTRACT

Recently, several companies have released H. pylori stool antigen (HpSA) test kits. However, there is little information about the usefulness of HpSA testing for Helicobacter felis, which is the major Helicobacter species in cats. The aim of the present study was to compare diagnostic methods for diagnosis of H. felis with HpSA tests and PCR assay using cat stools or gastric mucosa. Male cats (n=6) were infected with H. felis ATCC 49179 (1.0 x 10(9) CFU /cat) by intragastric inoculation two times at 3-day intervals, and stool specimens of cats were collected 1, 3, 5, 7, 14, and 21 days after infection for HpSA testing and H. felis-specific PCR. For the results, sensitivities of the HpSA test and PCR analysis were 50.0% and 83.3% respectively. Cats were sacrificed 21 days after H. felis inoculation, and gastric tissues were homogenized. All gastric biopsy specimens were positive based on a rapid urease test (RUT) (6/6, 100%) and PCR (6/6, 100%). Based on these results, the HpSA kit is useful and effective for monitoring H. felis infection using stool specimens. If an HpSA test could be made with H. felis antibodies in the future, its sensitivity could be increased further. Further, PCR assay could be successfully used to detect H. felis in stools. Application of this HpSA kit and PCR assay can be utilized as a non-invasive strategy to identify H. felis in cats.


Subject(s)
Animals , Cats , Humans , Male , Antibodies , Biopsy , Diagnosis , Felis , Gastric Mucosa , Helicobacter felis , Helicobacter , Natural Resources , Polymerase Chain Reaction , Urease
SELECTION OF CITATIONS
SEARCH DETAIL