Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 430
Filter
1.
Mucosal Immunol ; 11(2): 380-393, 2018 03.
Article in English | MEDLINE | ID: mdl-28745328

ABSTRACT

Lactobacillus reuteri 6475 (Lr) of the human microbiome synthesizes histamine and can suppress inflammation via type 2 histamine receptor (H2R) activation in the mammalian intestine. Gut microbes such as Lr promote H2R signaling and may suppress H1R proinflammatory signaling pathways in parallel by unknown mechanisms. In this study, we identified a soluble bacterial enzyme known as diacylglycerol kinase (Dgk) from Lr that is secreted into the extracellular milieu and presumably into the intestinal lumen. DgK diminishes diacylglycerol (DAG) quantities in mammalian cells by promoting its metabolic conversion and causing reduced protein kinase C phosphorylation (pPKC) as a net effect in mammalian cells. We demonstrated that histamine synthesized by gut microbes (Lr) activates both mammalian H1R and H2R, but Lr-derived Dgk suppresses the H1R signaling pathway. Phospho-PKC and IκBα were diminished within the intestinal epithelium of mice and humans treated by wild-type (WT) Lr, but pPKC and IκBα were not decreased in treatment with ΔdgkA Lr. Mucosal IL-6 and systemic interleukin (IL)-1α, eotaxin, and granulocyte colony-stimulating factor (G-CSF) were suppressed in WT Lr, but not in ΔdgkA Lr colonized mice. Collectively, the commensal microbe Lr may act as a "microbial antihistamine" by suppressing intestinal H1R-mediated proinflammatory responses via diminished pPKC-mediated mammalian cell signaling.


Subject(s)
Bacterial Proteins/metabolism , Diacylglycerol Kinase/metabolism , Intestinal Mucosa/immunology , Limosilactobacillus reuteri/metabolism , Protein Kinase C/metabolism , Receptors, Histamine H2/metabolism , Animals , Bacterial Proteins/genetics , Cytokines/metabolism , Diacylglycerol Kinase/genetics , Histamine/immunology , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/microbiology , Limosilactobacillus reuteri/physiology , Mice , Mice, Inbred BALB C , Microbiota , Mutation/genetics , Phosphorylation , Receptors, Histamine H1/metabolism , Signal Transduction
2.
Mucosal Immunol ; 10(6): 1504-1517, 2017 11.
Article in English | MEDLINE | ID: mdl-28198364

ABSTRACT

The risk of colon cancer is increased in patients with Crohn's disease and ulcerative colitis. Inflammation-induced DNA damage could be an important link between inflammation and cancer, although the pathways that link inflammation and DNA damage are incompletely defined. RAG2-deficient mice infected with Helicobacter hepaticus (Hh) develop colitis that progresses to lower bowel cancer. This process depends on nitric oxide (NO), a molecule with known mutagenic potential. We have previously hypothesized that production of NO by macrophages could be essential for Hh-driven carcinogenesis, however, whether Hh infection induces DNA damage in this model and whether this depends on NO has not been determined. Here we demonstrate that Hh infection of RAG2-deficient mice rapidly induces expression of iNOS and the development of DNA double-stranded breaks (DSBs) specifically in proliferating crypt epithelial cells. Generation of DSBs depended on iNOS activity, and further, induction of iNOS, the generation of DSBs, and the subsequent development of dysplasia were inhibited by depletion of the Hh-induced cytokine IL-22. These results demonstrate a strong association between Hh-induced DNA damage and the development of dysplasia, and further suggest that IL-22-dependent induction of iNOS within crypt epithelial cells rather than macrophages is a driving force in this process.


Subject(s)
Colitis, Ulcerative/immunology , Colon/pathology , Colonic Neoplasms/immunology , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Inflammation/immunology , Interleukins/metabolism , Macrophages, Peritoneal/immunology , Animals , Antibodies, Blocking/administration & dosage , Colitis, Ulcerative/complications , Colon/physiopathology , Colonic Neoplasms/complications , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Disease Models, Animal , Helicobacter Infections/complications , Humans , Interleukins/immunology , Macrophages, Peritoneal/microbiology , Mice , Mice, 129 Strain , Mice, Knockout , Neoplasms , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Interleukin-22
3.
Genome Announc ; 5(1)2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28057750

ABSTRACT

In collaboration with the CDC's Streptococcus Laboratory, we report here the whole-genome sequences of seven Streptococcus agalactiae bacteria isolated from laboratory-reared Long-Evans rats. Four of the S. agalactiae isolates were associated with morbidity accompanied by endocarditis, metritis, and fatal septicemia, providing an opportunity for comparative genomic analysis of this opportunistic pathogen.

4.
Infect Immun ; 84(8): 2307-2316, 2016 08.
Article in English | MEDLINE | ID: mdl-27245408

ABSTRACT

A urease-negative, fusiform, novel bacterium named Helicobacter saguini was isolated from the intestines and feces of cotton-top tamarins (CTTs) with chronic colitis. Helicobacter sp. was detected in 69% of feces or intestinal samples from 116 CTTs. The draft genome sequence, obtained by Illumina MiSeq sequencing, for H. saguini isolate MIT 97-6194-5, consisting of ∼2.9 Mb with a G+C content of 35% and 2,704 genes, was annotated using the NCBI Prokaryotic Genomes Automatic Annotation Pipeline. H. saguini contains homologous genes of known virulence factors found in other enterohepatic helicobacter species (EHS) and H. pylori These include flagellin, γ-glutamyl transpeptidase (ggt), collagenase, the secreted serine protease htrA, and components of a type VI secretion system, but the genome does not harbor genes for cytolethal distending toxin (cdt). H. saguini MIT 97-6194-5 induced significant levels of interleukin-8 (IL-8) in HT-29 cell culture supernatants by 4 h, which increased through 24 h. mRNAs for the proinflammatory cytokines IL-1ß, tumor necrosis factor alpha (TNF-α), IL-10, and IL-6 and the chemokine CXCL1 were upregulated in cocultured HT-29 cells at 4 h compared to levels in control cells. At 3 months postinfection, all H. saguini-monoassociated gnotobiotic C57BL/129 IL-10(-/-) mice were colonized and had seroconverted to H. saguini antigen with a significant Th1-associated increase in IgG2c (P < 0.0001). H. saguini induced a significant typhlocolitis, associated epithelial defects, mucosa-associated lymphoid tissue (MALT) hyperplasia, and dysplasia. Inflammatory cytokines IL-22, IL-17a, IL-1ß, gamma interferon (IFN-γ), and TNF-α, as well as inducible nitric oxide synthase (iNOS) were significantly upregulated in the cecal tissues of infected mice. The expression of the DNA damage response molecule γ-H2AX was significantly higher in the ceca of H. saguini-infected gnotobiotic mice than in the controls. This model using a nonhuman primate Helicobacter sp. can be used to study the pathogenic potential of EHS isolated from primates with naturally occurring inflammatory bowel disease (IBD) and colon cancer.


Subject(s)
Colitis, Ulcerative/veterinary , Colitis/microbiology , Colitis/pathology , Helicobacter/physiology , Monkey Diseases/microbiology , Animals , Antibodies, Bacterial/immunology , Cell Line , Colitis/genetics , Colitis/immunology , Cytokines/genetics , Disease Models, Animal , Feces/microbiology , Gene Expression , Genome, Bacterial , Helicobacter/classification , Helicobacter/isolation & purification , Histones/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Mice , Mice, Knockout , Phylogeny , RNA, Ribosomal, 16S/genetics
5.
Vet Pathol ; 53(2): 349-65, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26936751

ABSTRACT

Ferrets have become more popular as household pets and as animal models in biomedical research in the past 2 decades. The average life span of ferrets is about 5-11 years with onset of geriatric diseases between 3-4 years including endocrinopathies, neoplasia, gastrointestinal diseases, cardiomyopathy, splenomegaly, renal diseases, dental diseases, and cataract. Endocrinopathies are the most common noninfectious disease affecting middle-aged and older ferrets. Spontaneous neoplasms affecting the endocrine system of ferrets appear to be increasing in prevalence with a preponderance toward proliferative lesions in the adrenal cortex and pancreatic islet cells. Diet, gonadectomy, and genetics may predispose ferrets to an increased incidence of these endocrinopathies. These functional proliferative lesions cause hypersecretion of hormones that alter the physiology and metabolism of the affected ferrets resulting in a wide range of clinical manifestations. However, there is an apparent dearth of information available in the literature about the causal relationship between aging and neoplasia in ferrets. This review provides a comprehensive overview of the anatomy and physiology of endocrine organs, disease incidence, age at diagnosis, clinical signs, pathology, and molecular markers available for diagnosis of various endocrine disorders in ferrets.


Subject(s)
Aging/pathology , Endocrine System Diseases/veterinary , Ferrets , Age of Onset , Aging/genetics , Animals , Endocrine System/pathology , Endocrine System/physiology , Endocrine System Diseases/diagnosis , Endocrine System Diseases/genetics , Endocrine System Diseases/pathology , Models, Animal , Pets
6.
Mucosal Immunol ; 9(2): 515-26, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26422754

ABSTRACT

Psychological stressors are known to affect colonic diseases but the mechanisms by which this occurs, and whether probiotics can prevent stressor effects, are not understood. Because inflammatory monocytes that traffic into the colon can exacerbate colitis, we tested whether CCL2, a chemokine involved in monocyte recruitment, was necessary for stressor-induced exacerbation of infectious colitis. Mice were exposed to a social disruption stressor that entails repeated social defeat. During stressor exposure, mice were orally challenged with Citrobacter rodentium to induce a colonic inflammatory response. Exposure to the stressor during challenge resulted in significantly higher colonic pathogen levels, translocation to the spleen, increases in colonic macrophages, and increases in inflammatory cytokines and chemokines. The stressor-enhanced severity of C. rodentium-induced colitis was not evident in CCL2(-/-) mice, indicating the effects of the stressor are CCL2-dependent. In addition, we tested whether probiotic intervention could attenuate stressor-enhanced infectious colitis by reducing monocyte/macrophage accumulation. Treating mice with probiotic Lactobacillus reuteri reduced CCL2 mRNA levels in the colon and attenuated stressor-enhanced infectious colitis. These data demonstrate that probiotic L. reuteri can prevent the exacerbating effects of stressor exposure on pathogen-induced colitis, and suggest that one mechanism by which this occurs is through downregulation of the chemokine CCL2.


Subject(s)
Chemokine CCL2/immunology , Colitis/immunology , Enterobacteriaceae Infections/immunology , Limosilactobacillus reuteri/immunology , Probiotics/pharmacology , Stress, Psychological/immunology , Animals , Bacterial Translocation , Cell Movement , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colitis/microbiology , Colitis/pathology , Colitis/therapy , Colon/drug effects , Colon/immunology , Colon/microbiology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Enterobacteriaceae Infections/therapy , Gene Expression , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Limosilactobacillus reuteri/chemistry , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , RNA, Messenger/genetics , RNA, Messenger/immunology , Severity of Illness Index , Spleen/immunology , Spleen/microbiology , Stress, Psychological/microbiology , Stress, Psychological/pathology , Stress, Psychological/therapy
7.
Gut ; 64(10): 1650-68, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26342014

ABSTRACT

The stomach is traditionally regarded as a hollow muscular sac that initiates the second phase of digestion. Yet this simple view ignores the fact that it is the most sophisticated endocrine organ with unique physiology, biochemistry, immunology and microbiology. All ingested materials, including our nutrition, have to negotiate this organ first, and as such, the stomach is arguably the most important segment within the GI tract. The unique biological function of gastric acid secretion not only initiates the digestive process but also acts as a first line of defence against food-borne microbes. Normal gastric physiology and morphology may be disrupted by Helicobacter pylori infection, the most common chronic bacterial infection in the world and the aetiological agent for most peptic ulcers and gastric cancer. In this state-of-the-art review, the most relevant new aspects of the stomach in health and disease are addressed. Topics include gastric physiology and the role of gastric dysmotility in dyspepsia and gastroparesis; the stomach in appetite control and obesity; there is an update on the immunology of the stomach and the emerging field of the gastric microbiome. H. pylori-induced gastritis and its associated diseases including peptic ulcers and gastric cancer are addressed together with advances in diagnosis. The conclusions provide a future approach to gastric diseases underpinned by the concept that a healthy stomach is the gateway to a healthy and balanced host. This philosophy should reinforce any public health efforts designed to eradicate major gastric diseases, including stomach cancer.


Subject(s)
Stomach Diseases/diagnosis , Stomach Diseases/metabolism , Stomach/anatomy & histology , Stomach/physiology , Gastric Mucosa/metabolism , Humans
8.
Oncogene ; 34(22): 2856-66, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25088199

ABSTRACT

Runt domain transcription factor 3 (RUNX3) is widely regarded as a tumour-suppressor gene inactivated by DNA hypermethylation of its canonical CpG (cytidine-phosphate-guanidine) island (CGI) promoter in gastric cancer (GC). Absence of RUNX3 expression from normal gastric epithelial cells (GECs), the progenitors to GC, coupled with frequent RUNX3 overexpression in GC progression, challenge this longstanding paradigm. However, epigenetic models to better describe RUNX3 deregulation in GC have not emerged. Here, we identify lineage-specific DNA methylation at an alternate, non-CGI promoter (P1) as a new mechanism of RUNX3 epigenetic control. In normal GECs, P1 was hypermethylated and repressed, whereas in immune lineages P1 was hypomethylated and widely expressed. In human GC development, we detected aberrant P1 hypomethylation signatures associated with the early inflammatory, preneoplastic and tumour stages. Aberrant P1 hypomethylation was fully recapitulated in mouse models of gastric inflammation and tumorigenesis. Cell sorting showed that P1 hypomethylation reflects altered cell-type composition of the gastric epithelium/tumour microenvironment caused by immune cell recruitment, not methylation loss. Finally, via long-term culture of gastric tumour epithelium, we revealed that de novo methylation of the RUNX3 canonical CGI promoter is a bystander effect of oncogenic immortalization and not likely causal in GC pathogenesis as previously argued. We propose a new model of RUNX3 epigenetic control in cancer, based on immune-specific, non-CGI promoter hypomethylation. This novel epigenetic signature may have utility in early detection of GC and possibly other epithelial cancers with premalignant immune involvement.


Subject(s)
Cell Lineage/genetics , Core Binding Factor Alpha 3 Subunit/genetics , DNA Methylation , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cells, Cultured , CpG Islands , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology , Promoter Regions, Genetic , Stomach Neoplasms/pathology
9.
Oncogene ; 34(26): 3429-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25174398

ABSTRACT

Helicobacter pylori infection causes gastric cancer, the third leading cause of cancer death worldwide. More than half of the world's population is infected, making universal eradication impractical. Clinical trials suggest that antibiotic treatment only reduces gastric cancer risk in patients with non-atrophic gastritis (NAG), and is ineffective once preneoplastic lesions of multifocal atrophic gastritis (MAG) and intestinal metaplasia (IM) have occurred. Therefore, additional strategies for risk stratification and chemoprevention of gastric cancer are needed. We have implicated polyamines, generated by the rate-limiting enzyme ornithine decarboxylase (ODC), in gastric carcinogenesis. During H. pylori infection, the enzyme spermine oxidase (SMOX) is induced, which generates hydrogen peroxide from the catabolism of the polyamine spermine. Herein, we assessed the role of SMOX in the increased gastric cancer risk in Colombia associated with the Andean mountain region when compared with the low-risk region on the Pacific coast. When cocultured with gastric epithelial cells, clinical strains of H. pylori from the high-risk region induced more SMOX expression and oxidative DNA damage, and less apoptosis than low-risk strains. These findings were not attributable to differences in the cytotoxin-associated gene A oncoprotein. Gastric tissues from subjects from the high-risk region exhibited greater levels of SMOX and oxidative DNA damage by immunohistochemistry and flow cytometry, and this occurred in NAG, MAG and IM. In Mongolian gerbils, a prototype colonizing strain from the high-risk region induced more SMOX, DNA damage, dysplasia and adenocarcinoma than a colonizing strain from the low-risk region. Treatment of gerbils with either α-difluoromethylornithine, an inhibitor of ODC, or MDL 72527 (N(1),N(4)-Di(buta-2,3-dien-1-yl)butane-1,4-diamine dihydrochloride), an inhibitor of SMOX, reduced gastric dysplasia and carcinoma, as well as apoptosis-resistant cells with DNA damage. These data indicate that aberrant activation of polyamine-driven oxidative stress is a marker of gastric cancer risk and a target for chemoprevention.


Subject(s)
Adenocarcinoma , Helicobacter Infections/complications , Helicobacter pylori/physiology , Oxidoreductases Acting on CH-NH Group Donors/physiology , Stomach Neoplasms , Adenocarcinoma/epidemiology , Adenocarcinoma/genetics , Adenocarcinoma/microbiology , Adult , Animals , Cells, Cultured , Colombia/epidemiology , DNA Damage/genetics , Enzyme Induction , Gerbillinae , Helicobacter Infections/genetics , Humans , Hydrogen Peroxide/metabolism , Male , Middle Aged , Oxidative Stress/genetics , Risk Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Polyamine Oxidase
10.
Zoonoses Public Health ; 61(8): 571-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24628887

ABSTRACT

Routine necropsies of 27 asymptomatic juvenile chinchillas revealed a high prevalence of gastric ulcers with microscopic lymphoplasmacytic gastroenteritis and typhlocolitis. Polymerase chain reaction (PCR) analysis using Campylobacter genus-specific partial 16S rRNA primers revealed the presence of Campylobacter spp. DNA in the faeces of 12 of 27 animals (44.4%). Species-specific partial 16S rRNA PCR and sequencing confirmed that these animals were colonized with Campylobacter lanienae, a gram-negative, microaerophilic bacterium that was first identified on routine faecal screening of slaughterhouse employees and subsequently isolated from faeces of livestock. Campylobacter lanienae was isolated from the faeces of six PCR-positive animals and identified with species-specific PCR and full 16S rRNA sequencing. Phylogenetic analysis showed that these isolates clustered with C. lanienae strain NCTC 13004. PCR analysis of DNA extracted from gastrointestinal tissues revealed the presence of C. lanienae DNA in the caecum and colon of these chinchillas. Gastrointestinal lesions were scored and compared between C. lanienae-positive and C. lanienae-negative animals. There was no correlation between colonization status and lesion severity in the stomach, liver, duodenum, or colon. Possible routes of C. lanienae infection in chinchillas could include waterborne transmission and faecal-oral transmission from wild mice and rats or livestock. Based on these findings, the authors conclude that C. lanienae colonizes the lower bowel of chinchillas in the absence of clinical disease. This is the first report of C. lanienae in any rodent species. Campylobacter lanienae isolates from different mammalian species demonstrate heterogeneity by 16S rRNA sequence comparison. Analysis using rpoB suggests that isolates and clones currently identified as C. lanienae may represent multiple species or subspecies.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter/isolation & purification , Chinchilla/microbiology , Gastroenteritis/veterinary , Rodent Diseases/microbiology , Stomach Ulcer/veterinary , Animals , Bacterial Proteins/genetics , Base Sequence , Campylobacter/genetics , Campylobacter Infections/microbiology , DNA Primers/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Feces/microbiology , Gastroenteritis/microbiology , Male , Models, Animal , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA , Species Specificity , Stomach/microbiology , Stomach Ulcer/microbiology
11.
Vet Pathol ; 51(6): 1187-200, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24457157

ABSTRACT

Megaesophagus is defined as the abnormal enlargement or dilatation of the esophagus, characterized by a lack of normal contraction of the esophageal walls. This is called achalasia when associated with reduced or no relaxation of the lower esophageal sphincter (LES). To date, there are few naturally occurring models for this disease. A colony of transgenic (Pvrl3-Cre) rats presented with megaesophagus at 3 to 4 months of age; further breeding studies revealed a prevalence of 90% of transgene-positive animals having megaesophagus. Affected rats could be maintained on a total liquid diet long term and were shown to display the classic features of dilated esophagus, closed lower esophageal sphincter, and abnormal contractions on contrast radiography and fluoroscopy. Histologically, the findings of muscle degeneration, inflammation, and a reduced number of myenteric ganglia in the esophagus combined with ultrastructural lesions of muscle fiber disarray and mitochondrial changes in the striated muscle of these animals closely mimic that seen in the human condition. Muscle contractile studies looking at the response of the lower esophageal sphincter and fundus to electrical field stimulation, sodium nitroprusside, and L-nitro-L-arginine methyl ester also demonstrate the similarity between megaesophagus in the transgenic rats and patients with achalasia. No primary cause for megaesophagus was found, but the close parallel to the human form of the disease, as well as ease of care and manipulation of these rats, makes this a suitable model to better understand the etiology of achalasia as well as study new management and treatment options for this incurable condition.


Subject(s)
Disease Models, Animal , Esophageal Achalasia/etiology , Animals , Esophageal Achalasia/physiopathology , Esophagus/physiopathology , Esophagus/ultrastructure , Female , Humans , Male , Muscles/physiopathology , Muscles/ultrastructure , Rats , Rats, Transgenic
12.
Free Radic Res ; 47(11): 958-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23926919

ABSTRACT

The intestine comprises an exceptional venue for a dynamic and complex interplay of numerous chemical and biological processes. Here, multiple chemical and biological systems, including the intestinal tissue itself, its associated immune system, the gut microbiota, xenobiotics, and metabolites meet and interact to form a sophisticated and tightly regulated state of tissue homoeostasis. Disturbance of this homeostasis can cause inflammatory bowel disease (IBD)-a chronic disease of multifactorial etiology that is strongly associated with increased risk for cancer development. This review addresses recent developments in research into chemical and biological mechanisms underlying the etiology of inflammation-induced colon cancer. Beginning with a general overview of reactive chemical species generated during colonic inflammation, the mechanistic interplay between chemical and biological mediators of inflammation, the role of genetic toxicology, and microbial pathogenesis in disease development are discussed. When possible, we systematically compare evidence from studies utilizing human IBD patients with experimental investigations in mice. The comparison reveals that many strong pathological and mechanistic correlates exist between mouse models of colitis-associated cancer, and the clinically relevant situation in humans. We also summarize several emerging issues in the field, such as the carcinogenic potential of novel inflammation-related DNA adducts and genotoxic microbial factors, the systemic dimension of inflammation-induced genotoxicity, and the complex role of genome maintenance mechanisms during these processes. Taken together, current evidence points to the induction of genetic and epigenetic alterations by chemical and biological inflammatory stimuli ultimately leading to cancer formation.


Subject(s)
Carcinogenesis/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Inflammatory Bowel Diseases/metabolism , Animals , Carcinogenesis/pathology , Colitis/pathology , Colonic Neoplasms/pathology , Humans , Inflammatory Bowel Diseases/pathology , Mice
13.
QJM ; 105(11): 1097-103, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22908320

ABSTRACT

BACKGROUND: Central venous catheters (CVC) are a potential source of bacteraemia and have been associated with increased mortality in haemodialysis patients. We aimed to investigate the relationships between haemodialysis vascular access, taking into account changes in vascular access type during patients' lives, and cause specific mortality risk in a national cohort of dialysis patients. METHODS: Prospective cohort study including all patients receiving haemodialysis in Scotland at annual cross sectional surveys in 2009, 2010 and 2011. Data were collected through the Scottish Renal Registry and by a structured review of case records following death. Cox proportional hazards regression and multivariable logistic regression were used to model survival and risk of death from septicaemia respectively. RESULTS: Of a cohort of 2666 patients, 873 (32%) died during follow-up. After case-mix adjustment, patients using only tunnelled CVC during follow-up had a higher risk of all cause mortality across all strata of prior renal replacement therapy exposure [adjusted hazard ratio (HR): 1.83-2.08]. Case-mix adjusted risks of cardiovascular death (adjusted HR: 2.20-2.95) and infection-related death (adjusted HR: 3.10-3.63) were also higher in this group. Patients using tunnelled CVCs during follow-up and prior to death had 6.9-fold higher odds of death from septicaemia compared with those using only arteriovenous fistulae or grafts. CONCLUSION: Compared with an arteriovenous fistula or graft, sustained use of tunnelled CVCs for vascular access is associated with higher risks of all-cause, cardiovascular and infection-related mortality.


Subject(s)
Bacteremia/mortality , Catheterization, Central Venous/adverse effects , Registries , Renal Dialysis/mortality , Renal Insufficiency/mortality , Adult , Aged , Catheterization, Central Venous/statistics & numerical data , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Risk Assessment , Risk Factors , United Kingdom
14.
Phys Med Biol ; 56(21): 6779-91, 2011 Nov 07.
Article in English | MEDLINE | ID: mdl-21971037

ABSTRACT

Structural changes in breast tissue at the nanometre scale have been shown to differentiate between tissue types using synchrotron SAXS techniques. Classification of breast tissues using information acquired from a laboratory SAXS camera source could possibly provide a means of adopting SAXS as a viable diagnostic procedure. Tissue samples were obtained from surgical waste from 66 patients and structural components of the tissues were examined between q = 0.25 and 2.3 nm(-1). Principal component analysis showed that the amplitude of the fifth-order axial Bragg peak, the magnitude of the integrated intensity and the full-width at half-maximum of the fat peak were significantly different between tissue types. A discriminant analysis showed that excellent classification can be achieved; however, only 30% of the tissue samples provided the 16 variables required for classification. This suggests that the presence of disease is represented by a combination of factors, rather than one specific trait. A closer examination of the amorphous scattering intensity showed not only a trend of increased scattering intensity with disease severity, but also a corresponding decrease in the size of the scatterers contributing to this intensity.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Scattering, Small Angle , X-Ray Diffraction/methods , Adipose Tissue/diagnostic imaging , Adipose Tissue/pathology , Breast Neoplasms/classification , Breast Neoplasms/diagnostic imaging , Case-Control Studies , Discriminant Analysis , Female , Humans , Microfibrils/diagnostic imaging , Microfibrils/pathology , Principal Component Analysis , Radiography , Severity of Illness Index , Synchrotrons
15.
Mucosal Immunol ; 4(1): 22-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20944559

ABSTRACT

Pioneering work in the 1990s first linked a novel microaerobic bacterium, Helicobacter hepaticus, with chronic active hepatitis and inflammatory bowel disease in several murine models. Targeted H. hepaticus infection experiments subsequently demonstrated its ability to induce colitis, colorectal cancer, and extraintestinal diseases in a number of mouse strains with defects in immune function and/or regulation. H. hepaticus is now widely utilized as a model system to dissect how intestinal microbiota interact with the host to produce both inflammatory and tolerogenic responses. This model has been used to make important advances in understanding factors that regulate both acquired and innate immune response within the intestine. Further, it has been an effective tool to help define the function of regulatory T cells, including their ability to directly inhibit the innate inflammatory response to gut microbiota. The complete genomic sequence of H. hepaticus has advanced the identification of several virulence factors and aided in the elucidation of H. hepaticus pathogenesis. Delineating targets of H. hepaticus virulence factors could facilitate novel approaches to treating microbially induced lower bowel inflammatory diseases.


Subject(s)
Colorectal Neoplasms , Disease Models, Animal , Helicobacter Infections , Helicobacter hepaticus , Host-Pathogen Interactions , Inflammatory Bowel Diseases , Lower Gastrointestinal Tract , Animals , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter hepaticus/genetics , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Lower Gastrointestinal Tract/immunology , Lower Gastrointestinal Tract/microbiology , Lower Gastrointestinal Tract/pathology , Mice , Peptic Ulcer/microbiology , T-Lymphocytes/immunology , Virulence Factors
16.
Vet Pathol ; 48(4): 856-67, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21118799

ABSTRACT

A research colony of Xenopus (Silurana) tropicalis frogs presented with nodular and ulcerative skin lesions. Additional consistent gross findings included splenomegaly with multiple tan-yellow nodular foci in the spleen and liver of diseased frogs. Copious acid-fast positive bacteria were present in touch impression smears of spleen, skin, and livers of diseased frogs. Histologically, necrotizing and granulomatous dermatitis, splenitis, and hepatitis with numerous acid-fast bacilli were consistently present, indicative of systemic mycobacteriosis. Infrequently, granulomatous inflammation was noted in the lungs, pancreas, coelomic membranes, and rarely reproductive organs. Ultrastructurally, both extracellular bacilli and intracellular bacilli within macrophages were identified. Frogs in the affected room were systematically depopulated, and control measures were initiated. Cultured mycobacteria from affected organs were identified and genetically characterized as Mycobacterium liflandii by polymerase chain reaction amplification of the enoyl reductase domain and specific variable numbers of tandem repeats. In recent years, M. liflandii has had a devastating impact on research frog colonies throughout the United States. This detailed report with ultrastructural description of M. liflandii aids in further understanding of this serious disease in frogs.


Subject(s)
Mycobacterium Infections, Nontuberculous/veterinary , Mycobacterium/classification , Xenopus , Animals , Disease Outbreaks , Female , Hemorrhage , Liver/pathology , Lung/pathology , Mycobacterium Infections, Nontuberculous/epidemiology , Mycobacterium Infections, Nontuberculous/pathology , Ovary/pathology , Pancreas/pathology , Spleen/pathology
17.
J Clin Microbiol ; 48(5): 1908-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20220161

ABSTRACT

Helicobacter pullorum is a bacterial pathogen in humans. By using microaerobic culture techniques, H. pullorum was isolated from the feces of barrier-maintained mice and identified, on the basis of biochemical, restriction fragment length polymorphism, and 16S rRNA gene sequence analyses. This finding presents an opportunity to study H. pullorum pathogenesis in mice.


Subject(s)
Disease Outbreaks , Helicobacter Infections/veterinary , Helicobacter/isolation & purification , Mice, Inbred C3H/microbiology , Mice, Inbred C57BL/microbiology , Rodent Diseases/microbiology , Animals , Bacterial Typing Techniques , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Feces/microbiology , Helicobacter/classification , Helicobacter/genetics , Mice , Molecular Sequence Data , Phylogeny , Polymorphism, Restriction Fragment Length , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
18.
Gut ; 59(1): 88-97, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19850960

ABSTRACT

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) frequently results from synergism between chemical and infectious liver carcinogens. Worldwide, the highest incidence of HCC is in regions endemic for the foodborne contaminant aflatoxin B1 (AFB1) and hepatitis B virus (HBV) infection. Recently, gut microbes have been implicated in multisystemic diseases including obesity and diabetes. Here, the hypothesis that specific intestinal bacteria promote liver cancer was tested in chemical and viral transgenic mouse models. METHODS: Helicobacter-free C3H/HeN mice were inoculated with AFB1 and/or Helicobacter hepaticus. The incidence, multiplicity and surface area of liver tumours were quantitated at 40 weeks. Molecular pathways involved in tumourigenesis were analysed by microarray, quantitative real-time PCR, liquid chromatography/mass spectrometry, ELISA, western blot and immunohistochemistry. In a separate experiment, C57BL/6 FL-N/35 mice harbouring a full-length hepatitis C virus (HCV) transgene were crossed with C3H/HeN mice and cancer rates compared between offspring with and without H hepaticus. RESULTS: Intestinal colonisation by H hepaticus was sufficient to promote aflatoxin- and HCV transgene-induced HCC. Neither bacterial translocation to the liver nor induction of hepatitis was necessary. From its preferred niche in the intestinal mucus layer, H hepaticus activated nuclear factor-kappaB (NF-kappaB)-regulated networks associated with innate and T helper 1 (Th1)-type adaptive immunity both in the lower bowel and liver. Biomarkers indicative of tumour progression included hepatocyte turnover, Wnt/beta-catenin activation and oxidative injury with decreased phagocytic clearance of damaged cells. CONCLUSIONS: Enteric microbiota define HCC risk in mice exposed to carcinogenic chemicals or hepatitis virus transgenes. These results have implications for human liver cancer risk assessment and prevention.


Subject(s)
Aflatoxin B1/toxicity , Hepatitis B/complications , Intestines/microbiology , Liver Neoplasms, Experimental/etiology , Adaptive Immunity , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Chemokines/blood , Cocarcinogenesis , Female , Helicobacter Infections/complications , Helicobacter hepaticus , Hepatitis B/immunology , Immunity, Innate , Interleukin-12 Subunit p40/blood , Liver Neoplasms, Experimental/microbiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/physiology , Sex Factors , Signal Transduction/physiology , Th1 Cells/immunology
19.
J Clin Microbiol ; 47(11): 3673-81, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19759229

ABSTRACT

We recently described helicobacter-associated progressive, proliferative, and dysplastic typhlocolitis in aging (18- to 24-month-old) Syrian hamsters. Other pathogens associated with typhlocolitis in hamsters, Clostridium difficile, Lawsonia intracellularis, and Giardia spp., were not indentified. The presence of Helicobacter genus-specific DNA was noted by PCR in cecal and paraffin-embedded liver samples from aged hamsters by the use of Helicobacter-specific PCR primers. By 16S rRNA analysis, the Helicobacter sp. isolated from the liver tissue was identical to the cecal isolates from hamsters. The six hamster 16S rRNA sequences form a genotypic cluster most closely related to Helicobacter sp. Flexispira taxon 8, part of the Helicobacter bilis/H. cinaedi group. Livers from aged helicobacter-infected hamsters showed various stages of predominantly portocentric and, to a lesser extent, perivenular fibrosis. Within nodules, there was cellular atypia consistent with nodular dysplasia. The livers also exhibited a range of chronic active portal/interface and lobular inflammation, with significant portal hepatitis being present. The inflammation was composed of a mixture of lymphocytes, neutrophils, and macrophages, indicative of its chronic-active nature in these aged hamsters infected with Helicobacter spp. The isolation of novel Helicobacter spp., their identification by PCR from the diseased livers of aged hamsters, and their taxonomic classification as belonging to the Helicobacter bilis cluster strengthen the argument that H. bilis and closely related Helicobacter spp. play an etiological role in hepatobiliary disease in both animals and humans.


Subject(s)
Biliary Tract Diseases/veterinary , Fibrosis/veterinary , Helicobacter Infections/veterinary , Helicobacter/pathogenicity , Hepatitis, Chronic/veterinary , Hyperplasia/veterinary , Liver Cirrhosis/veterinary , Animals , Biliary Tract Diseases/microbiology , Cecum/microbiology , Cluster Analysis , Cricetinae , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Fibrosis/microbiology , Helicobacter/classification , Helicobacter/genetics , Helicobacter/isolation & purification , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Hepatitis, Chronic/microbiology , Hepatitis, Chronic/pathology , Hyperplasia/microbiology , Inflammation/pathology , Liver/microbiology , Liver/pathology , Liver Cirrhosis/microbiology , Liver Cirrhosis/pathology , Lymphocytes/immunology , Macrophages/immunology , Mesocricetus/microbiology , Molecular Sequence Data , Neutrophils/immunology , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid
20.
J Med Primatol ; 38(5): 377-81, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19737242

ABSTRACT

BACKGROUND: Seizures were observed in a 16-year old male Guyanese squirrel monkey with a history of inappetence and weakness. METHODS AND RESULTS: Complete blood count, biochemical profile, and urinalysis indicated systemic disease. Nematode larvae were detected in the feces. Cerebrospinal fluid (CSF) analysis revealed leukocytes and gram-positive cocci. Staphylococcus aureus was isolated from the CSF. Histopathological evaluation revealed systemic lesions with inflammation and nematodes in the small and large intestine. CONCLUSION: This is the first report describing spontaneous staphylococcal CNS infection in a squirrel monkey.


Subject(s)
Meningoencephalitis/veterinary , Saimiri , Secernentea Infections/veterinary , Staphylococcal Infections/veterinary , Animals , Brain/pathology , Colitis/complications , Colitis/parasitology , Colitis/veterinary , Enterobius/isolation & purification , Male , Meningoencephalitis/complications , Meningoencephalitis/microbiology , Meningoencephalitis/pathology , Secernentea Infections/complications , Secernentea Infections/parasitology , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/isolation & purification , Strongyloides/isolation & purification , Typhlitis/complications , Typhlitis/parasitology , Typhlitis/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...