Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Microbiol Spectr ; 12(1): e0345023, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38014984

ABSTRACT

IMPORTANCE: H. pylori infects half of the world population and is the leading cause of gastric cancer. We previously demonstrated that gastric cancer risk is associated with gastric microbiota. Specifically, gastric urease-positive Staphylococcus epidermidis and Streptococcus salivarius had contrasting effects on H. pylori-associated gastric pathology and immune responses in germ-free INS-GAS mice. As gastritis progresses to gastric cancer, the oncogenic transcription factor Foxm1 becomes increasingly expressed. In this study, we evaluated the gastric commensal C. acnes, certain strains of which produce thiopeptides that directly inhibit FOXM1. Thiopeptide-positive C. acnes was isolated from Nicaraguan patient gastric biopsies and inoculated into germ-free INS-GAS mice with H. pylori. We, therefore, asked whether coinfection with C. acnes expressing thiopeptide and H. pylori would decrease gastric Foxm1 expression and pro-inflammatory cytokine mRNA and protein levels. Our study supports the growing literature that specific non-H. pylori gastric bacteria affect inflammatory and cancer biomarkers in H. pylori pathogenesis.


Subject(s)
Coinfection , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Mice , Animals , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Disease Models, Animal , Biomarkers, Tumor , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Forkhead Box Protein M1/genetics
2.
Microbes Infect ; 25(3): 105045, 2023.
Article in English | MEDLINE | ID: mdl-36162750

ABSTRACT

Iron deficiency, the most common micronutrient deficiency in humans, is associated with long-term deficits in cognition and memory if left untreated. Infection with the gastric pathogen Helicobacter pylori has been linked to iron deficiency anemia (IDA). The H. pylori virulence factor cytotoxin-associated gene A (cagA) is proposed to be especially pertinent in iron deficiency. Male INS-GAS/FVB mice were infected with the CagA+ strain pre-murine Sydney strain 1 (PMSS1) for 12-13 or 27-29 weeks to investigate the role of chronic H. pylori infection in iron deficiency and neurological sequelae. Mice at both timepoints demonstrated significantly elevated gastric histopathology scores and inflammatory cytokines compared to sham-dosed controls. However, only mice at 27-29 weeks post infection had changes in hematological parameters, with significantly decreased erythrocyte count, hematocrit, serum hemoglobin, and increased serum total iron binding capacity. Gastric transcription of iron-regulatory genes Hamp and Bmp4 were significantly downregulated at both timepoints. In the brain, iron-dependent myelingergic and synaptic markers were significantly downregulated at 27-29 weeks. These results indicated that long-term infection of the CagA + PMSS1 strain of H. pylori in this study caused anemia, altered gastric iron homeostasis, and neurological changes similar to those reported in other rodent H. pylori CagA- strain infection models.


Subject(s)
Anemia, Iron-Deficiency , Helicobacter Infections , Helicobacter pylori , Iron Deficiencies , Humans , Male , Mice , Animals , Helicobacter pylori/genetics , Iron/metabolism , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/pathology , Brain/pathology , Helicobacter Infections/pathology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
3.
mSphere ; 7(1): e0077221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138124

ABSTRACT

In populations with similar prevalence of Helicobacter pylori infection, cancer risk can vary dramatically. Changes in composition or structure of bacterial communities in the stomach, either at the time of exposure or over the course of H. pylori infection, may contribute to gastric pathology. In this study, a population of 37 patients from the low-gastric-cancer-risk (LGCR) region of Tumaco, Colombia, and the high-gastric-cancer-risk (HGCR) region of Túquerres, Colombia, were recruited for gastric endoscopy. Antral biopsy specimens were processed for histology and bacterial isolation. Fifty-nine distinct species among 26 genera were isolated by aerobic, anaerobic, and microaerobic culture and confirmed by 16S rRNA analysis. Urease-positive Staphylococcus epidermidis and Streptococcus salivarius were frequently isolated from gastric biopsy specimens. We asked whether coinfection of H. pylori with urease-positive S. salivarius and/or S. epidermidis had a demonstrable effect on H. pylori-induced gastritis in the germfree (GF) INS-GAS mouse model. Coinfections with S. salivarius and/or S. epidermidis did not affect gastric H. pylori colonization. At 5 months postinfection, GF INS-GAS mice coinfected with H. pylori and S. salivarius had statistically higher pathological scores in the stomachs than mice infected with H. pylori only or H. pylori with S. epidermidis (P < 0.05). S. epidermidis coinfection with H. pylori did not significantly change stomach pathology, but levels of the proinflammatory cytokine genes Il-1ß, Il-17A , and Il-22 were significantly lower than in H. pylori-monoinfected mice. This study demonstrates that non-H. pylori urease-positive bacteria may play a role in the severity of H. pylori-induced gastric cancer in humans. IMPORTANCE Chronic infection with H. pylori is the main cause of gastric cancer, which is a global health problem. In two Colombian populations with high levels of H. pylori prevalence, the regional gastric cancer rates are considerably different. Host genetic background, H. pylori biotype, environmental toxins, and dietary choices are among the known risk factors for stomach cancer. The potential role of non-H. pylori gastric microbiota in gastric carcinogenesis is being increasingly recognized. In this study, we isolated 59 bacterial species from 37 stomach biopsy samples of Colombian patients from both low-gastric-cancer-risk and high-gastric-cancer-risk regions. Urease-positive S. epidermidis and S. salivarius commonly cultured from the stomachs, along with H. pylori, were inoculated into germfree INS-GAS mice. S. salivarius coinfection with H. pylori induced significantly higher gastric pathology than in H. pylori-monoinfected mice, whereas S. epidermidis coinfection caused significantly lower H. pylori-induced proinflammatory cytokine responses than in H. pylori-monoinfected mice. This study reinforces the argument that the non-H. pylori stomach microflora play a role in the severity of H. pylori-induced gastric cancer.


Subject(s)
Coinfection , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Streptococcus salivarius , Animals , Coinfection/complications , Cytokines , Disease Models, Animal , Helicobacter Infections/complications , Humans , Immunity , Mice , RNA, Ribosomal, 16S/genetics , Staphylococcus epidermidis/genetics , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Streptococcus salivarius/genetics , Urease
4.
J Med Microbiol ; 70(3)2021 Mar.
Article in English | MEDLINE | ID: mdl-33475481

ABSTRACT

Introduction. Helicobacter suis (Helicobacter heilmannii type 1) commonly infects nonhuman primates but its clinical importance is in question.Aim. To characterize H. suis infection in a colony of rhesus macaques (Macaca mulatta) used in cognitive neuroscience research.Hypothesis/Gap Statement. Inquiries into the nature of Helicobacter suis in nonhuman primates are required to further define the organism's virulence and the experimental animal's gastric microbiome.Methodology. Animals with and without clinical signs of vomiting and abdominal pain (n=5 and n=16, respectively) were evaluated by histology, culture, PCR amplification and sequencing, fluorescent in situ hybridization (FISH) and serology. Three of the five animals with clinical signs, an index case and two others, were evaluated before and after antimicrobial therapy.Results. The index animal had endoscopically visible ulcers and multifocal, moderate, chronic lymphoplasmacytic gastritis with intraglandular and luminal spiral bacteria. Antimicrobial therapy in the index animal achieved histologic improvement, elimination of endoscopically visible ulcers, and evident eradication but clinical signs persisted. In the other treated animals, gastritis scores were not consistently altered, gastric bacteria persisted, but vomiting and abdominal discomfort abated.Nineteen of 21 animals were PCR positive for H. suis and five animals were also PCR positive for H. pylori. Organisms were detected by FISH in 17 of 21 animals: 16S rRNA sequences of two of these were shown to be H. suis. Mild to moderate lymphoplasmacytic gastritis was seen in antrum, body and cardia, with antral gastritis more likely to be moderate than that of the body.Conclusion. No clear association between the bacterial numbers of Helicobacter spp. and the degree of inflammation was observed. H. suis is prevalent in this colony of Macaca mulatta but its clinical importance remains unclear. This study corroborates many of the findings in earlier studies of H. suis infection in macaques but also identifies at least one animal in which gastritis and endoscopically visible gastric ulcers were strongly associated with H. suis infection. In this study, serology was an inadequate biomarker for endoscopic evaluation in diagnosis of H. suis infection.


Subject(s)
Gastritis/veterinary , Helicobacter Infections/veterinary , Helicobacter heilmannii/isolation & purification , Helicobacter pylori/isolation & purification , Monkey Diseases/microbiology , Stomach Ulcer/veterinary , Animals , Female , Gastritis/microbiology , Helicobacter Infections/microbiology , Macaca mulatta/microbiology , Male , Stomach Ulcer/microbiology
5.
J Exp Med ; 198(1): 111-22, 2003 Jul 07.
Article in English | MEDLINE | ID: mdl-12835477

ABSTRACT

During maturation, dendritic cells (DCs) regulate their capacity to process and present major histocompatibility complex (MHC) II-restricted antigens. Here we show that presentation of exogenous antigens by MHC I is also subject to developmental control, but in a fashion strikingly distinct from MHC II. Immature mouse bone marrow-derived DCs internalize soluble ovalbumin and sequester the antigen intracellularly until they receive an appropriate signal that induces cross presentation. At that time, peptides are generated in a proteasome-dependent fashion and used to form peptide-MHC I complexes that appear at the plasma membrane. Unlike MHC II, these events do not involve a marked redistribution of preexisting MHC I molecules from intracellular compartments to the DC surface. Moreover, out of nine stimuli well known to induce the phenotypic maturation of DCs and to promote MHC II presentation, only two (CD40 ligation, disruption of cell-cell contacts) activated cross presentation on MHC I. In contrast, formation of peptide-MHC I complexes from endogenous cytosolic antigens occurs even in unstimulated, immature DCs. Thus, the MHC I and MHC II pathways of antigen presentation are differentially regulated during DC maturation.


Subject(s)
Antigen Presentation , Dendritic Cells/physiology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/metabolism , Animals , CD11c Antigen/analysis , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , beta 2-Microglobulin/metabolism
6.
Microbiol Resour Announc ; 9(11)2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32165379

ABSTRACT

A fast-growing Mycobacterium species was cultured from draining, purulent lesions on the caudal abdomen of a 12-year-old male domestic long-haired cat. Whole-genome sequencing identified the organism as Mycobacterium porcinum.

7.
Gut Microbes ; 11(4): 820-841, 2020 07 03.
Article in English | MEDLINE | ID: mdl-31955643

ABSTRACT

Infection with Helicobacter pylori causes chronic inflammation and is a risk factor for gastric cancer. Antibiotic treatment or increased dietary folate prevents gastric carcinogenesis in male INS-GAS mice. To determine potential synergistic effects, H. pylori-infected male INS-GAS mice were fed an amino acid defined (AAD) diet with increased folate and were treated with antibiotics after 18 weeks of H. pylori infection. Antibiotic therapy decreased gastric pathology, but dietary folate had no effect. However, the combination of antibiotics and the AAD diet induced anemia, gastric hemorrhage, and mortality. Clinical presentation suggested hypovitaminosis K potentially caused by dietary deficiency and dysbiosis. Based on current dietary guidelines, the AAD diet was deficient in vitamin K. Phylloquinone administered subcutaneously and via a reformulated diet led to clinical improvement with no subsequent mortalities and increased hepatic vitamin K levels. We characterized the microbiome and menaquinone profiles of antibiotic-treated and antibiotic-free mice. Antibiotic treatment decreased the abundance of menaquinone producers within orders Bacteroidales and Verrucomicrobiales. PICRUSt predicted decreases in canonical menaquinone biosynthesis genes, menA and menD. Reduction of menA from Akkermansia muciniphila, Bacteroides uniformis, and Muribaculum intestinale were confirmed in antibiotic-treated mice. The fecal menaquinone profile of antibiotic-treated mice had reduced MK5 and MK6 and increased MK7 and MK11 compared to antibiotic-free mice. Loss of menaquinone-producing microbes due to antibiotics altered the enteric production of vitamin K. This study highlights the role of diet and the microbiome in maintaining vitamin K homeostasis.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Dysbiosis/etiology , Food, Formulated/adverse effects , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Microbiome , Helicobacter Infections/drug therapy , Vitamin K Deficiency/etiology , Amino Acids/administration & dosage , Anemia/diet therapy , Anemia/etiology , Animals , Anti-Bacterial Agents/adverse effects , Bacteria/drug effects , Bacteria/genetics , Bacteria/metabolism , Diet , Dietary Supplements , Folic Acid/administration & dosage , Folic Acid/biosynthesis , Folic Acid/genetics , Gastrointestinal Microbiome/drug effects , Helicobacter Infections/microbiology , Helicobacter pylori/drug effects , Liver/metabolism , Male , Mice , Vitamin K 1/administration & dosage , Vitamin K 1/metabolism , Vitamin K 2/metabolism
8.
J Am Assoc Lab Anim Sci ; 58(6): 796-801, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31662161

ABSTRACT

Current methods for detecting mites in mouse colonies have limitations in terms of cost, accuracy, and throughput. To address these limitations, we developed PCR assays to detect Myocoptes musculinus in fecal samples. Using a newly generated ribosomal RNA sequence of M. musculinus (MC28S), we developed PCR and qPCR assays capable of detecting M. musculinus mites or eggs ingested during grooming. To determine our ability to detect mites, we tested fur swabs and feces from mouse colonies experimentally infested with M. musculinus and Demodex musculi, 2) Myobia musculi and Radfordia affinis, 3) M. musculinus and M. musculi, and 4) no mites (negative control). The MC28S PCR and qPCR assays positively identified M. musculinus in groups 1 and 3. The MC28S PCR assay detected M. musculinus in 9 of 10 fecal samples from known-positive animals, whereas the qPCR assay correctly identified M. musculinus in all 10 fecal samples. To our knowledge, this report is the first description of PCR-based detection of murine mites in feces. By eliminating the need for pelt examinations, mite detection from fecal samples can facilitate mite detection in sentinel or quarantine programs.


Subject(s)
Animal Fur/parasitology , Feces/parasitology , Mite Infestations/veterinary , Mites/classification , Rodent Diseases/diagnosis , Animals , Mice , Mite Infestations/diagnosis , Mites/genetics , RNA, Ribosomal/chemistry , Real-Time Polymerase Chain Reaction , Rodent Diseases/parasitology
9.
Comp Med ; 68(1): 25-30, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29460718

ABSTRACT

A laboratory-housed, wild-caught, subadult, male meadow jumping mouse (Zapus hudsonius) presented with extensive scaling of the face, limbs, and tail and severe edema of the paws. Postmortem examination revealed marked distal limb edema with focal digital hematomas and white scales, scabs, and crusts affecting the majority of nonhaired skin. Histopathologic analysis revealed severe, multifocal, chronic-active exudative and proliferative dermatitis characterized by multilaminated crusts covering the epidermis. The epidermis was expanded by hyperkeratosis, acanthosis, and hyperplasia. The superficial dermis contained moderate edema, hemorrhage, and pigmentary incontinence, and was infiltrated by granulocytes and mononuclear cells. The laminated crusts contained numerous branching filaments of gram-positive coccoid bodies arranged in parallel rows, consistent with cutaneous Dermatophilus congolensis infection. This diagnosis was confirmed through bacterial culture and 16S rRNA PCR analysis. In the presented case, factors that might have contributed to disease progression include climatic conditions at the capture site and stress associated with trapping and laboratory housing.


Subject(s)
Dermatitis/veterinary , Rodent Diseases/microbiology , Rodentia , Skin Diseases, Bacterial/veterinary , Actinobacteria/isolation & purification , Animals , Dermatitis/microbiology , Dermatitis/pathology , Male , Skin Diseases, Bacterial/microbiology
10.
J Am Assoc Lab Anim Sci ; 45(4): 14-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16884173

ABSTRACT

Microbial infections are common sequelae in humans and animals implanted with long-term intravascular catheters. Understanding the pathophysiology of infectious morbidity is critical to improving quality of care in catheterized subjects. Here, we describe findings in 6 clinically healthy, male sheep implanted with indwelling aortic or cardiac catheters for 6 to 10 mo. We isolated multiple bacterial species including Serratia spp., Enterobacter agglomerans, Eschericia coli, Klebsiella oxytoca, and K. pneumoniae in aerobic cultures from catheter tips. Although sheep were clinically asymptomatic, 1 or both kidneys from all animals contained wedge-shaped infarcts of varying size and number. Microscopic examination revealed (a) marked fibrosis with mild inflammatory cell infiltrate consistent with chronic foreign body reaction around catheters; (b) moderate to severe, diffuse, subacute to chronic membranoproliferative glomerulonephritis and mild, multifocal chronic interstitial nephritis; and (c) mesangial immune-complex deposition as demonstrated by direct immunofluorescence technique. The finding of bacterial colonization of catheters together with chronic glomerulonephritis and immune-complex deposits in kidneys in clinically asymptomatic sheep underscores the need for close microbiologic monitoring of catheter implants and assessment of kidney function in animals instrumented for long-term vascular access.


Subject(s)
Bacterial Infections/veterinary , Catheters, Indwelling/veterinary , Glomerulonephritis/veterinary , Infarction/veterinary , Kidney/blood supply , Sheep Diseases/etiology , Animals , Bacterial Infections/etiology , Bacterial Infections/pathology , Catheterization , Catheters, Indwelling/microbiology , Enterobacteriaceae/cytology , Enterobacteriaceae/isolation & purification , Glomerulonephritis/etiology , Glomerulonephritis/immunology , Infarction/etiology , Kidney/pathology , Male , Sheep , Sheep Diseases/immunology , Sheep Diseases/microbiology
11.
J Immunol ; 169(9): 4982-9, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391212

ABSTRACT

Immunosuppressive agents are commonly used in the prevention of graft rejection following transplantation and in the treatment of autoimmunity. In this study, we examined the immunosuppressive mechanism of the drug 15-deoxyspergualin (DSG), which has shown efficacy in the enhancement of graft survival and in the treatment of autoimmunity. Using a murine model of chronic relapsing and remitting experimental autoimmune encephalomyelitis, we were able to demonstrate that DSG both delayed and reduced the severity of experimental autoimmune encephalomyelitis. Subsequent in vitro studies to examine the mechanism of immune suppression showed that DSG was not able to inhibit early activation of naive CD4 T cells, but DSG did effectively inhibit the growth of naive CD4 T cells after activation. An analysis of cell proliferation and cell cycle showed that DSG treatment led to a block in cell cycle progression 2-3 days following Ag stimulation. In addition, DSG treatment inhibited the production of IFN-gamma by Th1 effector T cells. These studies suggest that CD4 T cells are a predominant target for DSG and the immunosuppressive effects of the drug may result from reduced CD4 T cell expansion and decreased polarization into IFN-gamma-secreting Th1 effector T cells in the induction of certain autoimmune disorders.


Subject(s)
Cell Cycle/drug effects , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Growth Inhibitors/pharmacology , Guanidines/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , T-Lymphocyte Subsets/cytology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/immunology , Growth Inhibitors/therapeutic use , Guanidines/therapeutic use , Immunosuppressive Agents/therapeutic use , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interphase/drug effects , Interphase/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/immunology , Peptide Fragments/immunology , Severity of Illness Index , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
J Immunol ; 171(3): 1484-92, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12874241

ABSTRACT

We have previously presented evidence demonstrating that mice deficient in NF-kappaB subunits are susceptible to colitis induced by the pathogenic enterohepatic Helicobacter species, H. hepaticus. However, it has not been determined whether NF-kappaB is required within inhibitory lymphocyte populations, within cells of the innate immune system, or both, to suppress inflammation. To examine these issues, we have performed a series of adoptive transfer experiments using recombination-activating gene (Rag)-2(-/-) or p50(-/-)p65(+/-)Rag-2(-/-) mice as hosts for wild-type (WT) and p50(-/-)p65(+/-) lymphocyte populations. We have shown that although the ability of H. hepaticus to induce colitis in Rag-2(-/-) mice is inhibited by the presence of either WT or p50(-/-)p65(+/-) splenocytes, these splenocyte populations are unable to suppress H. hepaticus-induced colitis in p50(-/-)p65(+/-)Rag-2(-/-) mice. Colitis in these animals is characterized by increased expression of inflammatory cytokines including IL-12 p40, and depletion of IL-12 p40 from p50(-/-)p65(+/-) mice ameliorates H. hepaticus-induced disease. Consistent with a primary defect in the regulation of IL-12 expression, H. hepaticus induced markedly higher levels of IL-12 p40 in p50(-/-)p65(+/-) macrophages than in WT macrophages. These results suggest that inhibition of H. hepaticus-induced IL-12 p40 expression by NF-kappaB subunits is critical to preventing colonic inflammation in response to inflammatory microflora.


Subject(s)
Colitis/microbiology , Colitis/prevention & control , Helicobacter/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , NF-kappa B/physiology , Protein Subunits/antagonists & inhibitors , Protein Subunits/biosynthesis , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Colitis/genetics , Colitis/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Immune Sera/administration & dosage , Immunity, Innate/genetics , Injections, Intraperitoneal , Interleukin-12/immunology , Interleukin-12 Subunit p40 , Intestinal Mucosa/pathology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/transplantation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Protein Subunits/immunology , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Spleen/transplantation , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL