Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Animals (Basel) ; 13(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37685068

ABSTRACT

Chronic inflammatory enteropathies (CIEs) in dogs involve the infiltration of gastrointestinal tissue with inflammatory cells. This study aimed to assess the sensitivity of serum and fecal 3-bromotyrosine (3-BrY) concentrations in dogs with CIE. The difference in 3-BrY concentrations in dogs with different gastrointestinal (GI) pathological changes was also assessed. In total, 68 dogs with CIE were enrolled in the study. The median serum 3-BrY concentration was 3.3 µmol/L, while the median 3-day mean and maximum fecal 3-BrY concentrations were 38.9 and 63.2 mmol/g of feces, respectively. The median serum C-reactive protein concentration was 45.0 mg/L. The median 3-day mean and maximum fecal α1-proteinase inhibitor concentrations were 6.1 and 9 µg/g of feces, respectively. Increased 3-BrY concentrations were observed in 90.9% of CIE dogs based on serum concentrations, 75.8% based on mean fecal concentrations, and 69.4% based on maximum fecal concentrations. A weak correlation (ρ = 0.31, p < 0.0118) was found between serum CRP and serum 3-BrY concentrations. There was no correlation between the canine chronic enteropathy clinical activity index and serum or fecal 3-BrY concentrations (p > 0.05). Additionally, no significant difference in serum or fecal 3-BrY concentrations was found among CIE dogs with different GI pathological changes (p > 0.05). In conclusion, dogs with CIE have increased 3-BrY concentrations in serum and fecal samples. However, 3-BrY concentrations may not accurately indicate the severity of gastrointestinal inflammation.

2.
Nutrients ; 13(4)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920187

ABSTRACT

Various proteins or protein fractions reportedly positively affect gastrointestinal integrity and inflammation in diets providing >45% energy as fat. This study tested whether benefits were seen in diets providing 30% of energy as fat. Purified diets (PD) with isolated soy protein (ISP), dried whole milk powder (DWMP), milk fat globule membrane (MFGM), or milk protein concentrate (MPC) as protein sources were fed to C57BL/6J mice (n = 15/diet group) for 13 weeks. MFGM-fed mice were heaviest (p < 0.005) but remained within breeder norms. Growth rates and gut motility were similar for all PD-fed mice. FITC-dextran assessed gut permeability was lowest in DWMP and MFGM (p = 0.054); overall, plasma endotoxin and unprovoked circulating cytokines indicated a non-inflammatory state for all PD-fed mice. Despite differences in cecal butyrate and intestinal gene expression, all PDs supported gastrointestinal health. Whole milk provided more positive effects compared to its fractions. However, ISP-fed mice showed a >370%, (p < 0.006) increase in colonic myeloperoxidase activity indicative of tissue neutrophil infiltration. Surprisingly, FITC-dextran and endotoxin outcomes were many folds better in PD-fed mice than mice (strain, vendor, age and sex matched) fed a "chow-type" nutritionally adequate non-PD. Additional variables within a diet's matrix appear to affect routine indicators or gastrointestinal health.


Subject(s)
Feeding Behavior/physiology , Gastrointestinal Tract/physiology , Glycolipids/administration & dosage , Glycoproteins/administration & dosage , Milk Proteins/administration & dosage , Soybean Proteins/administration & dosage , Animal Feed , Animals , Biomarkers , Gastrointestinal Motility , Lipid Droplets , Male , Mice , Mice, Inbred C57BL , Models, Animal
3.
Front Vet Sci ; 7: 192, 2020.
Article in English | MEDLINE | ID: mdl-32363202

ABSTRACT

The aim was to characterize differences in fecal consistency, and fecal microbiota and metabolome profiles in dogs with acute diarrhea (AD) treated with either fecal microbiota transplantation as enema (FMT; n = 11) or oral metronidazole (MET; n = 7) for 7 days. On days 0, 7, and 28 fecal samples were obtained. Fecal samples from healthy dogs (HC; n = 14) were used for comparison. Samples were analyzed by the previously validated qPCR based canine Dysbiosis Index (DI; increased values indicate microbiota dysbiosis) and 16S rRNA gene sequencing. The fecal metabolome was analyzed using a previously validated targeted canine assay for fecal unconjugated bile acids, and untargeted metabolomics. Fecal consistency improved significantly in dogs treated with FMT and MET by day 7 and day 28 (p < 0.01) compared to day 0. However, on day 28 fecal consistency was significantly better in FMT compared to MET (p = 0.040). At day 0, dogs with AD had an altered microbiota indicated by significantly increased DI, decreased alpha-diversity, and altered beta-diversity. In the FMT group, the DI decreased over time, while MET led to a significant increase in the dysbiosis index at day 7 and 28 compared to FMT. Sequencing data revealed that in FMT microbial diversity and beta-diversity was similar to HC at day 28, while in MET these parameters were still significantly different from HC. In dogs treated with FMT, a decrease in cholic acid and the percentage of primary bile acids was observed, whereas treatment with metronidazole led to an increase in cholic acid at day 7 and an increase in percentage of primary bile acids over time. Based on untargeted metabolomics, dogs with AD had an altered fecal metabolome compared to HC. Dogs treated with FMT clustered closer to HC at day 28, while dogs treated with MET did not. In this pilot study, dogs with AD had significant differences in fecal microbiota and metabolome profiles. Dogs treated with MET still had altered microbial and metabolic profiles at day 28 compared to dogs treated with FMT or healthy dogs.

4.
Vet Sci ; 7(2)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326394

ABSTRACT

Akkermansia muciniphila is a mucin-degrading bacterium that has shown the potential to provide anti-inflammatory and anti-obesity effects in mouse and man. We here focus on companion animals, specifically cats and dogs, and evaluate the microbial degradation of mucus and its health impact in the context of the worldwide epidemic of pet obesity. A literature survey revealed that the two presently known Akkermansia spp., A. muciniphila and A. glycaniphila, as well as other members of the phylum of Verrucomicrobia seem to be neither very prevalent nor abundant in the digestive tract of cats and dog. While this may be due to methodological aspects, it suggests that bacteria related to Akkermansia are not the major mucus degraders in these pets and hence other mucus-utilizing taxa may deserve attention. Hence, we will discuss the potential of these endogenous mucus utilizers and dietary interventions to boost these as well as the use of Akkermansia spp. related bacteria or their components as strategies to target feline and canine obesity.

5.
J Vet Intern Med ; 33(4): 1608-1618, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31099928

ABSTRACT

BACKGROUND: Accumulating evidence shows an important relationship between the gastrointestinal (GI) microbiota and host health. Microbial metabolites are believed to play a critical role in host-microbial interactions. Short-chain fatty acids (SCFAs) are major end products of bacterial carbohydrate fermentation in the intestinal tract. Decreased concentrations of SCFAs have been observed in humans with GI disease. However, large-scale clinical data in dogs are lacking. HYPOTHESIS/OBJECTIVE: To evaluate fecal concentrations of SCFAs and the fecal microbiota in healthy control (HC) dogs and dogs with chronic enteropathy (CE). ANIMALS: Forty-nine privately owned HC dogs and 73 dogs with CE. METHODS: Prospective cohort study. Fecal concentrations of SCFAs were measured using gas chromatography/mass spectrometry. Illumina sequencing and quantitative real-time polymerase chain reaction were utilized to evaluate the fecal microbiota. RESULTS: Fecal concentrations (median [range] µmol/g of dry matter) of acetate were lower (P = .03) in dogs with CE (185.8 [20.1-1042.1]) than in HC dogs (224.0 [87.7-672.8]). Propionate were also lower (P < .001) in dogs with CE (46.4 [0.4-227.9]) than in HC dogs (105.9 [1.6-266.9]). Moreover, total SCFAs were lower (P = .005) in dogs with CE (268.1 [21.8-1378.2]) than in HC dogs (377.2 [126.6-927.0]). Dysbiosis in dogs with CE was characterized by decreased bacterial diversity and richness, distinct microbial community clustering compared with that in HC dogs, and a higher dysbiosis index. CONCLUSIONS AND CLINICAL IMPORTANCE: Dogs with CE had an altered fecal SCFA concentration accompanied by significant changes of the fecal microbiota.


Subject(s)
Dog Diseases/microbiology , Dysbiosis/veterinary , Fatty Acids, Volatile/analysis , Gastrointestinal Microbiome , Intestinal Diseases/veterinary , Animals , Case-Control Studies , Dogs , Feces/chemistry , Intestinal Diseases/microbiology , Prospective Studies
6.
J Vet Intern Med ; 32(6): 1903-1910, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30353569

ABSTRACT

BACKGROUND: Dietary interventions are thought to modify gut microbial communities in healthy individuals. In dogs with chronic enteropathies, resolution of dysbiosis, along with remission of clinical signs, is expected with treatment. HYPOTHESIS/OBJECTIVE: To evaluate changes in the fecal microbiota in dogs with food-responsive chronic enteropathy (FRE) and in healthy control (HC) dogs before and after an elimination dietary trial with an animal protein-free diet (APFD). ANIMALS: Dogs with FRE (n = 10) and HC (n = 14). METHODS: Dogs were fed the APFD for 60 days. Fecal microbiota was analyzed by Illumina 16S rRNA sequencing and quantitative polymerase chain reaction (PCR). RESULTS: A significantly lower bacterial alpha-diversity was observed in dogs with FRE compared with HC dogs at baseline, and compared with FRE dogs after the trial. Distinct microbial communities were observed in dogs with FRE at baseline compared with HC dogs at baseline and compared with dogs with FRE after the trial. Microbial communities still were different in FRE dogs after the trial compared with HC dogs at baseline. In HC dogs, the fecal microbiota did not show a significant modification after administration of the APFD. CONCLUSION AND CLINICAL IMPORTANCE: Our results suggest that, in FRE dogs, treatment with the APFD led to a partial recovery of the fecal microbiota by significantly increasing microbiota richness, which was significantly closer to a healthy microbiota after the treatment. In contrast, no changes were detected in the fecal microbiota of HC dogs fed the same APFD.


Subject(s)
Diet, Protein-Restricted/veterinary , Dog Diseases/diet therapy , Gastrointestinal Diseases/veterinary , Gastrointestinal Microbiome , Animal Feed , Animals , Dog Diseases/microbiology , Dogs/microbiology , Feces/microbiology , Female , Gastrointestinal Diseases/diet therapy , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/genetics , Male , Polymerase Chain Reaction/veterinary
7.
J Nutr Biochem ; 43: 107-115, 2017 05.
Article in English | MEDLINE | ID: mdl-28282584

ABSTRACT

This study investigated the potential role of the p70S6K1/HIF1α axis in the anti-inflammatory activities of pomegranate (Punica granatum L.) polyphenolics in dextran sodium sulfate (DSS)-induced colitis in Sprague-Dawley rats and in lipopolysaccharide (LPS)-treated CCD-18Co colon-myofibroblastic cells. Rats were administered either control (CT) or pomegranate beverage (PG), containing ellagic acid and ellagitannins, then exposed to three cycles of 3% DSS followed by a 2-week recovery period. PG protected against DSS-induced colon inflammation and ulceration (50% and 66.7%, P=.05 and .045, respectively), and decreased the Ki-67 proliferative index in the central and basal regions compared to the control. PG also significantly reduced the expression of proinflammatory cytokines (TNF-α and IL-1ß), COX-2, and iNOS at mRNA and protein levels. In addition, the expression of p70S6K1 and HIF1α was reduced, while the tumor suppressor miR-145 was induced by PG. The intestinal microbiota of rats treated with PG showed a significant increase in Ruminococcaceae that include several butyrate producing bacteria (P=.03). In vitro, PG reduced the expression of p70S6K1 and HIF1α and induced miR-145 in a dose-dependent manner. The involvement of miR-145/p70S6K1 was confirmed by treating LPS-treated CCD-18Co cells with miR-145 antagomiR, where the pomegranate polyphenolics reversed the effects of the antagomiR for p70S6K1 mRNA and protein levels. These results suggest that pomegranate polyphenols attenuated DSS-induced colitis by modulating the miR-145/p70S6K/HIF1α axis, indicating potential use in therapeutic treatment of ulcerative colitis.


Subject(s)
Colitis, Ulcerative/diet therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lythraceae/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Cell Proliferation/drug effects , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Fruit and Vegetable Juices , Gastrointestinal Microbiome/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/metabolism , Polyphenols/pharmacology , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/genetics
8.
Gut Microbes ; 6(1): 33-47, 2015.
Article in English | MEDLINE | ID: mdl-25531678

ABSTRACT

Idiopathic inflammatory bowel disease (IBD) is a common cause of chronic gastrointestinal (GI) disease in dogs. The combination of an underlying host genetic susceptibility, an intestinal dysbiosis, and dietary/environmental factors are suspected as main contributing factors in the pathogenesis of canine IBD. However, actual mechanisms of the host-microbe interactions remain elusive. The aim of this study was to compare the fecal microbiota and serum metabolite profiles between healthy dogs (n = 10) and dogs with IBD before and after 3 weeks of medical therapy (n = 12). Fecal microbiota and metabolite profiles were characterized by 454-pyrosequencing of 16 S rRNA genes and by an untargeted metabolomics approach, respectively. Significantly lower bacterial diversity and distinct microbial communities were observed in dogs with IBD compared to the healthy control dogs. While Gammaproteobacteria were overrepresented, Erysipelotrichia, Clostridia, and Bacteroidia were underrepresented in dogs with IBD. The functional gene content was predicted from the 16 S rRNA gene data using PICRUSt, and revealed overrepresented bacterial secretion system and transcription factors, and underrepresented amino acid metabolism in dogs with IBD. The serum metabolites 3-hydroxybutyrate, hexuronic acid, ribose, and gluconic acid lactone were significantly more abundant in dogs with IBD. Although a clinical improvement was observed after medical therapy in all dogs with IBD, this was not accompanied by significant changes in the fecal microbiota or in serum metabolite profiles. These results suggest the presence of oxidative stress and a functional alteration of the GI microbiota in dogs with IBD, which persisted even in the face of a clinical response to medical therapy.


Subject(s)
Biota , Dog Diseases/pathology , Feces/microbiology , Inflammatory Bowel Diseases/veterinary , Serum/chemistry , Animals , Dog Diseases/therapy , Dogs , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Metabolome , Metabolomics , Metagenomics , Sequence Analysis, DNA
9.
World J Gastroenterol ; 20(44): 16489-97, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25469017

ABSTRACT

The intestinal microbiota is the collection of the living microorganisms (bacteria, fungi, protozoa, and viruses) inhabiting the gastrointestinal tract. Novel bacterial identification approaches have revealed that the gastrointestinal microbiota of dogs and cats is, similarly to humans, a highly complex ecosystem. Studies in dogs and cats have demonstrated that acute and chronic gastrointestinal diseases, including inflammatory bowel disease (IBD), are associated with alterations in the small intestinal and fecal microbial communities. Of interest is that these alterations are generally similar to the dysbiosis observed in humans with IBD or animal models of intestinal inflammation, suggesting that microbial responses to inflammatory conditions of the gut are conserved across mammalian host types. Studies have also revealed possible underlying susceptibilities in the innate immune system of dogs and cats with IBD, which further demonstrate the intricate relationship between gut microbiota and host health. Commonly identified microbiome changes in IBD are decreases in bacterial groups within the phyla Firmicutes and Bacteroidetes, and increases within Proteobacteia. Furthermore, a reduction in the diversity of Clostridium clusters XIVa and IV (i.e., Lachnospiraceae and Clostridium coccoides subgroups) are associated with IBD, suggesting that these bacterial groups may play an important role in maintenance of gastrointestinal health. Future studies are warranted to evaluate the functional changes associated with intestinal dysbiosis in dogs and cats.


Subject(s)
Bacteria/pathogenicity , Cat Diseases/microbiology , Dog Diseases/microbiology , Inflammatory Bowel Diseases/veterinary , Intestines/microbiology , Microbiota , Acute Disease , Animals , Bacteria/classification , Bacteria/genetics , Cat Diseases/diagnosis , Cat Diseases/immunology , Cats , Chronic Disease , Dog Diseases/diagnosis , Dog Diseases/immunology , Dogs , Fungi/pathogenicity , Host-Pathogen Interactions , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestines/immunology , Ribotyping
10.
Vet Microbiol ; 174(3-4): 463-473, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25458422

ABSTRACT

Clostridium perfringens has been suspected as an enteropathogen in dogs. However, its exact role in gastrointestinal (GI) disorders in dogs remains unknown. Recent studies suggest the importance of an altered intestinal microbiota in the activation of virulence factors of enteropathogens. The aim of this study was to evaluate the relationship between diarrhea, dysbiosis, and the presence of C. perfringens and its enterotoxin (CPE). Fecal samples were collected prospectively from 95 healthy control dogs and 104 dogs with GI disease and assessed for bacterial abundances and the presence of CPE using quantitative PCR and ELISA, respectively. C. perfringens was detected in all dogs. Potentially enterotoxigenic C. perfringens were detected in 33.7% (32/95) of healthy control dogs and 48.1% (50/104) diseased dogs, respectively. CPE was detected by ELISA in 1.0% (1/95) of control dogs and 16.3% (17/104) of diseased dogs. Abundances of Fusobacteria, Ruminococcaceae, Blautia, and Faecalibacterium were significantly decreased in diseased dogs, while abundances of Bifidobacterium, Lactobacillus, and Escherichia coli were significantly increased compared to control dogs. The microbial dysbiosis was independent of the presence of the enterotoxigenic C. perfringens or CPE. In conclusion, the presence of CPE as well as fecal dysbiosis was associated with GI disease. However, the presence of C. perfringens was not indicative of GI disease in all cases of diarrhea, and the observed increased abundance of enterotoxigenic C. perfringens may be part of intestinal dysbiosis occurring in GI disease. The significance of an intestinal dysbiosis in dogs with GI disease deserves further attention.


Subject(s)
Clostridium Infections/veterinary , Clostridium perfringens/isolation & purification , Dysbiosis/veterinary , Enterotoxins/analysis , Gastrointestinal Diseases/veterinary , Animals , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridium perfringens/genetics , Diarrhea/epidemiology , Diarrhea/microbiology , Diarrhea/veterinary , Dogs , Dysbiosis/epidemiology , Dysbiosis/microbiology , Enterotoxins/isolation & purification , Enzyme-Linked Immunosorbent Assay/veterinary , Feces/microbiology , Female , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/microbiology , Male , Microbiota , Polymerase Chain Reaction/veterinary , Prevalence
11.
PLoS One ; 9(7): e101723, 2014.
Article in English | MEDLINE | ID: mdl-25007331

ABSTRACT

BACKGROUND: Growing evidence shows the potential of nutritional interventions to treat obesity but most investigations have utilized non-digestible carbohydrates only. Peach and plum contain high amounts of polyphenols, compounds with demonstrated anti-obesity effects. The underlying process of successfully treating obesity using polyphenols may involve an alteration of the intestinal microbiota. However, this phenomenon is not well understood. METHODOLOGY/PRINCIPAL FINDINGS: Obese Zucker rats were assigned to three groups (peach, plum, and control, n = 10 each), wild-type group was named lean (n = 10). Carbohydrates in the fruit juices were eliminated using enzymatic hydrolysis. Fecal samples were obtained after 11 weeks of fruit or control juice administration. Real-time PCR and 454-pyrosequencing were used to evaluate changes in fecal microbiota. Over 1,500 different Operational Taxonomic Units at 97% similarity were detected in all rats. Several bacterial groups (e.g. Lactobacillus and members of Ruminococcacea) were found to be more abundant in the peach but especially in the plum group (plum juice contained 3 times more total polyphenolics compared to peach juice). Principal coordinate analysis based on Unifrac-based unweighted distance matrices revealed a distinct separation between the microbiota of control and treatment groups. These changes in fecal microbiota occurred simultaneously with differences in fecal short-chain acids concentrations between the control and treatment groups as well as a significant decrease in body weight in the plum group. CONCLUSIONS: This study suggests that consumption of carbohydrate-free peach and plum juice has the potential to modify fecal microbial ecology in an obese animal model. The separate contribution of polyphenols and non-polyphenols compounds (vitamins and minerals) to the observed changes is unknown.


Subject(s)
Anti-Obesity Agents/administration & dosage , Beverages , Microbiota/drug effects , Obesity/drug therapy , Plant Extracts/administration & dosage , Prunus/chemistry , Animals , Dietary Carbohydrates/administration & dosage , Fatty Acids/metabolism , Feces/microbiology , Male , Microbiota/genetics , Obesity/microbiology , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Rats, Zucker
12.
Anim Health Res Rev ; 13(1): 64-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22853923

ABSTRACT

The close relationship between gastrointestinal (GI) microbiota and its host has an impact on the health status of an animal that reaches beyond the GI tract. A balanced microbiome stimulates the immune system, aids in the competitive exclusion of transient pathogens and provides nutritional benefits to the host. With recent rapid advances in high-throughput sequencing technology, molecular approaches have become the routinely used tools for ecological studies of the feline microbiome, and have revealed a highly diverse and complex intestinal ecosystem in the feline GI tract. The major bacterial groups are similar to those found in other mammals, with Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria constituting more than 99% of intestinal microbiota. Several nutritional studies have demonstrated that the feline microbiota can be modulated by the amount of soluble fibers (i.e., prebiotics) and macronutrients (i.e., protein content) in the diet. Initial clinical studies have suggested the presence of a dysbiosis in feline inflammatory bowel disease (IBD). Recently, metagenomic approaches have attempted to characterize the microbial gene pool. However, more studies are needed to describe the phylogenetic and functional changes in the intestinal microbiome in disease states and in response to environmental and dietary modulations. This paper reviews recent studies cataloging the microbial phylotypes in the GI tract of cats.


Subject(s)
Bacteria/classification , Cats/microbiology , Gastrointestinal Tract/microbiology , Metagenome , Animals , Bacteria/genetics , Bacteria/metabolism , Cat Diseases/microbiology , High-Throughput Nucleotide Sequencing/veterinary , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/veterinary , Intestines/microbiology , Metagenomics , Phylogeny
13.
Anim Health Res Rev ; 13(1): 78-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22647637

ABSTRACT

Gastrointestinal (GI) microbes have important roles in the nutritional, immunological, and physiologic processes of the host. Traditional cultivation techniques have revealed bacterial density ranges from 10(4) to 10(5) colony forming units (CFU)/g in the stomach, from 10(5) to 10(7) CFU/g in the small intestine, and from 10(9) to 10(11) CFU/g in the colon of healthy dogs. As a small number of bacterial species can be grown and studied in culture, however, progress was limited until the recent emergence of DNA-based techniques. In recent years, DNA sequencing technology and bioinformatics have allowed for better phylogenetic and functional/metabolic characterization of the canine gut microbiome. Predominant phyla include Firmicutes, Bacteroidetes, Fusobacteria, Proteobacteria, and Actinobacteria. Studies using 16S ribosomal RNA (rRNA) gene pyrosequencing have demonstrated spatial differences along the GI tract and among microbes adhered to the GI mucosa compared to those in intestinal contents or feces. Similar to humans, GI microbiome dysbiosis is common in canine GI diseases such as chronic diarrhea and inflammatory bowel diseases. DNA-based assays have also identified key pathogens contributing to such conditions, including various Clostridium, Campylobacter, Salmonella, and Escherichia spp. Moreover, nutritionists have applied DNA-based techniques to study the effects of dietary interventions such as dietary fiber, prebiotics, and probiotics on the canine GI microbiome and associated health indices. Despite recent advances in the field, the canine GI microbiome is far from being fully characterized and a deeper characterization of the phylogenetic and functional/metabolic capacity of the GI microbiome in health and disease is needed. This paper provides an overview of recent studies performed to characterize the canine GI microbiome.


Subject(s)
Bacteria/classification , Dog Diseases/microbiology , Dogs/microbiology , Gastrointestinal Tract/microbiology , Metagenome , Animals , Bacteria/genetics , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/veterinary , High-Throughput Nucleotide Sequencing/veterinary , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/veterinary , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/veterinary
14.
FEMS Microbiol Ecol ; 80(3): 624-36, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22324305

ABSTRACT

The effect of a proton pump inhibitor on gastrointestinal (GI) microbiota was evaluated. Eight healthy 9-month-old dogs (four males and four females) received omeprazole (1.1 mg kg(-1) ) orally twice a day for 15 days. Fecal samples and endoscopic biopsies from the stomach and duodenum were obtained on days 30 and 15 before omeprazole administration, on day 15 (last day of administration), and 15 days after administration. The microbiota was evaluated using 16S rRNA gene 454-pyrosequencing, fluorescence in situ hybridization, and qPCR. In the stomach, pyrosequencing revealed a decrease in Helicobacter spp. during omeprazole (median 92% of sequences during administration compared to > 98% before and after administration; P = 0.0336), which was accompanied by higher proportions of Firmicutes and Fusobacteria. FISH confirmed this decrease in gastric Helicobacter (P < 0.0001) and showed an increase in total bacteria in the duodenum (P = 0.0033) during omeprazole. However, Unifrac analysis showed that omeprazole administration did not significantly alter the overall phylogenetic composition of the gastric and duodenal microbiota. In feces, qPCR showed an increase in Lactobacillus spp. during omeprazole (P < 0.0001), which was accompanied by a lower abundance of Faecalibacterium spp. and Bacteroides-Prevotella-Porphyromonas in the male dogs. This study suggests that omeprazole administration leads to quantitative changes in GI microbiota of healthy dogs.


Subject(s)
Bacteria/drug effects , Duodenum/microbiology , Metagenome , Omeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Stomach/microbiology , Animals , Bacteria/classification , Bacteria/genetics , DNA, Bacterial/genetics , Dogs , Feces/microbiology , Female , In Situ Hybridization, Fluorescence , Male , Phylogeny , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
15.
PLoS One ; 7(12): e51907, 2012.
Article in English | MEDLINE | ID: mdl-23300577

ABSTRACT

BACKGROUND: Recent molecular studies have revealed a highly complex bacterial assembly in the canine intestinal tract. There is mounting evidence that microbes play an important role in the pathogenesis of acute and chronic enteropathies of dogs, including idiopathic inflammatory bowel disease (IBD). The aim of this study was to characterize the bacterial microbiota in dogs with various gastrointestinal disorders. METHODOLOGY/PRINCIPAL FINDINGS: Fecal samples from healthy dogs (n = 32), dogs with acute non-hemorrhagic diarrhea (NHD; n = 12), dogs with acute hemorrhagic diarrhea (AHD; n = 13), and dogs with active (n = 9) and therapeutically controlled idiopathic IBD (n = 10) were analyzed by 454-pyrosequencing of the 16S rRNA gene and qPCR assays. Dogs with acute diarrhea, especially those with AHD, had the most profound alterations in their microbiome, as significant separations were observed on PCoA plots of unweighted Unifrac distances. Dogs with AHD had significant decreases in Blautia, Ruminococcaceae including Faecalibacterium, and Turicibacter spp., and significant increases in genus Sutterella and Clostridium perfringens when compared to healthy dogs. No significant separation on PCoA plots was observed for the dogs with IBD. Faecalibacterium spp. and Fusobacteria were, however, decreased in the dogs with clinically active IBD, but increased during time periods of clinically insignificant IBD, as defined by a clinical IBD activity index (CIBDAI). CONCLUSIONS: Results of this study revealed a bacterial dysbiosis in fecal samples of dogs with various GI disorders. The observed changes in the microbiome differed between acute and chronic disease states. The bacterial groups that were commonly decreased during diarrhea are considered to be important short-chain fatty acid producers and may be important for canine intestinal health. Future studies should correlate these observed phylogenetic differences with functional changes in the intestinal microbiome of dogs with defined disease phenotypes.


Subject(s)
Biomarkers/metabolism , Diarrhea/genetics , Dog Diseases/genetics , Feces/microbiology , Inflammatory Bowel Diseases/genetics , Metagenome/physiology , Animals , Case-Control Studies , Diarrhea/microbiology , Diarrhea/veterinary , Dog Diseases/microbiology , Dogs , Feces/chemistry , Female , Gene Expression Profiling , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/veterinary , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...