Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
Anim Microbiome ; 6(1): 7, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383422

ABSTRACT

BACKGROUND: Age-associated changes in the gastrointestinal microbiome of young pigs have been robustly described; however, the temporal dynamics of the fecal microbiome of the female pig from early life to first parity are not well understood. Our objective was to describe microbiome and antimicrobial resistance dynamics of the fecal microbiome of breeding sows from early life through estrus, parturition and weaning of the first litter of piglets (i.e., from 3 to 53 weeks of age). RESULTS: Our analysis revealed that fecal bacterial populations in developing gilts undergo changes consistent with major maturation milestones. As the pigs progressed towards first estrus, the fecal bacteriome shifted from Rikenellaceae RC9 gut group- and UCG-002-dominated enterotypes to Treponema- and Clostridium sensu stricto 1-dominated enterotypes. After first estrus, the fecal bacteriome stabilized, with minimal changes in enterotype transition and associated microbial diversity from estrus to parturition and subsequent weaning of first litter piglets. Unlike bacterial communities, fecal fungal communities exhibited low diversity with high inter- and intra-pig variability and an increased relative abundance of certain taxa at parturition, including Candida spp. Counts of resistant fecal bacteria also fluctuated over time, and were highest in early life and subsequently abated as the pigs progressed to adulthood. CONCLUSIONS: This study provides insights into how the fecal microbial community and antimicrobial resistance in female pigs change from three weeks of age throughout their first breeding lifetime. The fecal bacteriome enterotypes and diversity are found to be age-driven and established by the time of first estrus, with minimal changes observed during subsequent physiological stages, such as parturition and lactation, when compared to the earlier age-related shifts. The use of pigs as a model for humans is well-established, however, further studies are needed to understand how our results compare to the human microbiome dynamics. Our findings suggest that the fecal microbiome exhibited consistent changes across individual pigs and became more diverse with age, which is a beneficial characteristic for an animal model system.

2.
Transl Anim Sci ; 7(1): txad093, 2023.
Article in English | MEDLINE | ID: mdl-37649650

ABSTRACT

A total of 34,749 pigs were used in two experiments to evaluate the effects of a postbiotic dried fermentation product (DFP) administered through drinking water on nursery pig growth performance, antibiotic injection frequency, morbidity, mortality, fecal consistency, and characterization of fecal Escherichia coli. The DFP is composed of bioactive molecules derived from Lactococcus lactis. In Exp. 1, 350 barrows (DNA Line 200 × 400; initial body weight [BW] 6.1 ± 0.01 kg) were used in a 42-d study with five pigs per pen and 35 pens per treatment. The DFP was supplied for 14 d at a target dosage of 24 mg/kg BW using a water medicator at a 1:128 dilution. On days 7 and 14, fecal samples were collected for dry matter (DM) and to determine, by a multiplex polymerase chain reaction (PCR) assay, prevalence of 11 virulence genes characteristic of E. coli pathotypes. There was no evidence (P > 0.10) for differences for growth, incidence of diarrhea, number of antibiotic injections, removals, or fecal DM. On both fecal collection days, E. coli virulence genes were present with day 7 samples positive for genes that encode for hemolysins (hlyA, exhA), intimin (eae), and enteroaggregative heat-stable enterotoxin (astA). Prevalence of enterotoxin genes (elt, estA, estB, astA) increased on day 14, but DFP had no effects on the prevalence of any of the virulence genes. A total of 32 out of 72 E. coli isolates were identified as enterotoxigenic pathotype and all except one were from day 14 fecal samples. Fourteen isolates were positive for F4 fimbria and one isolate was positive for F4 and F18 fimbriae. In Exp. 2, 34,399 nursery pigs (initially 5.6 kg) were used in 20 nursery barns with 10 barns per treatment (control or DFP). The target dosage of the DFP for the first 14 d was 35 mg/kg BW. Following the 14-d supplementation period, pigs continued to be monitored for approximately 31 d. There was no evidence (P > 0.05) for the DFP to influence the overall percentage of pigs that died or growth performance. From days 0 to 14, providing the DFP reduced (P < 0.05) the percentage of pigs that were euthanized. However, providing the DFP increased (P < 0.05) the overall percentage of pigs that were euthanized and total mortality. For the number of antibiotic injections (treatment interventions), providing the DFP reduced the number of injections for the common period (P < 0.001) and overall (P = 0.002). These results indicate that the DFP did not influence growth performance but providing the DFP in Exp. 2 led to increased total nursery pig mortality.

3.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-37480360

ABSTRACT

Holstein steers (n = 40; initial body weight [BW] = 96.0 ± 10.5 kg) were individually housed in a climate-controlled barn to evaluate potential models for the genesis of liver abscesses (LA). In this 2 × 2 factorial, steers were balanced by BW and randomly assigned to one of two treatments: 1) intravenous saline injection followed by intraruminal bacterial inoculation with Fusobacterium necrophorum subsp. necrophorum (1 × 109 colony forming unit [CFU]/mL) and Salmonella enterica serovar Lubbock (1 × 106 CFU/mL; CON; n = 20 steers); or 2) intravenous injection with 0.25 µg/kg BW of lipopolysaccharide (LPS; Escherichia coli O111:B4) followed by intraruminal bacterial inoculation of F. necrophorum subsp. necrophorum (1 × 109 CFU/mL) and S. enterica serovar Lubbock (1 × 106 CFU/mL; LBI; n = 20 steers) and 1 of 2 harvest dates (3 or 10 d post LPS infusion). Body weights were recorded on days -4, -1, 3, and 10, and blood was collected for hematology on days -4, 3, and 10, relative to LPS infusion on day 0. Intraruminal bacterial inoculation occurred on day 1. Steers from each treatment group were harvested at two different time points on day 3 or 10 to perform gross pathological examination of the lung, rumen, liver, LA (if present), and colon. Feed disappearance was less for LBI than CON (P < 0.01); however, BW did not differ (P = 0.33) between treatments. Neither treatment nor time differed for hematology (P ≥ 0.13), and no gross pathological differences were noted in the lung, liver, LA, or colon (P ≥ 0.25). A treatment × harvest date interaction was noted for ruminal pathology in which LBI had an increased percentage of abnormal rumen scores on day 3 (P < 0.01). These results suggest that an LPS challenge in combination with intraruminal bacterial inoculation of pathogens commonly isolated from LA was not sufficient to induce LA in steers within 3 or 10 d (P = 0.95) when compared to CON. Further evaluation is needed to produce a viable model to investigate the genesis and prevention of LA in cattle.


Liver abscesses in feedlot cattle can cause a decrease in feed intake, average daily gain, feed efficiency, and hot carcass weight. At harvest, liver abscesses result in liver condemnations, carcass trimming, and a potential decrease in quality grade, with an estimated economic cost to packers of $41.6 million annually. Our objective was to evaluate an intravenous endotoxin challenge followed by intraruminal inoculation of bacteria commonly isolated from liver abscesses over a 10-d period as a potential model to understand the genesis and etiology of liver abscesses in cattle and evaluate possible preventative interventions. Results suggest that an endotoxin challenge in combination with intraruminal bacterial inoculation is not a viable model to induce liver abscesses in steers, and bacterial inoculation alone was insufficient to induce liver abscesses. The length of time necessary to induce liver abscesses is also unknown. Based on our results, more research is needed to develop a noninvasive model to induce liver abscesses in cattle.


Subject(s)
Cattle Diseases , Liver Abscess , Cattle , Animals , Endotoxins , Lipopolysaccharides/toxicity , Liver Abscess/prevention & control , Liver Abscess/veterinary , Fusobacterium , Body Weight , Animal Feed/analysis , Diet/veterinary , Cattle Diseases/prevention & control
4.
Anim Microbiome ; 5(1): 2, 2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36624546

ABSTRACT

BACKGROUND: The pig gastrointestinal tract hosts a diverse microbiome, which can serve to select and maintain a reservoir of antimicrobial resistance genes (ARG). Studies suggest that the types and quantities of antimicrobial resistance (AMR) in fecal bacteria change as the animal host ages, yet the temporal dynamics of AMR within communities of bacteria in pigs during a full production cycle remains largely unstudied. RESULTS: A longitudinal study was performed to evaluate the dynamics of fecal microbiome and AMR in a cohort of pigs during a production cycle; from birth to market age. Our data showed that piglet fecal microbial communities assemble rapidly after birth and become more diverse with age. Individual piglet fecal microbiomes progressed along similar trajectories with age-specific community types/enterotypes and showed a clear shift from E. coli/Shigella-, Fusobacteria-, Bacteroides-dominant enterotypes to Prevotella-, Megaspheara-, and Lactobacillus-dominated enterotypes with aging. Even when the fecal microbiome was the least diverse, the richness of ARGs, quantities of AMR gene copies, and counts of AMR fecal bacteria were highest in piglets at 2 days of age; subsequently, these declined over time, likely due to age-related competitive changes in the underlying microbiome. ARGs conferring resistance to metals and multi-compound/biocides were detected predominately at the earliest sampled ages. CONCLUSIONS: The fecal microbiome and resistome-along with evaluated descriptors of phenotypic antimicrobial susceptibility of fecal bacteria-among a cohort of pigs, demonstrated opposing trajectories in diversity primarily driven by the aging of pigs.

5.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-36588460

ABSTRACT

Liver abscesses in feedlot cattle are a polymicrobial infection with Fusobacterium necrophorum and Trueperella pyogenes as the primary and secondary etiologic agents, respectively. Cattle with liver abscesses do not exhibit clinical signs and the abscesses are detected only at slaughter. The objective was to conduct metabolomics analysis of purulent materials of liver abscesses to identify biochemicals. Liver abscesses from crossbred cattle (n = 24) and Holstein steers (n = 24), each fed high-grain finishing diet with tylosin (n = 12) or no tylosin (n = 12), were included in the study. Abscess purulent materials were analyzed by ultrahigh-performance liquid chromatography-tandem mass spectroscopy. A total of 759 biochemicals were identified and were broadly categorized into carbohydrates, energy metabolism pathways intermediates, peptides, amino acids and their metabolites, lipids and their metabolites, nucleotides, vitamins and cofactors, xenobiotics, and partially characterized molecules. The top 50 biochemicals identified included amino acids, lipids, nucleotides, xenobiotics, peptides, and carbohydrates and their metabolites. Among the 15 amino acid metabolites in the top 50 biochemicals, four were tryptophan metabolites, indoleacrylate, indolepropionate, tryptamine, and anthranilate. The 3-phenylpropionate, a product of phenylalanine metabolism, was the predominant metabolite in purulent materials. Between the four treatment groups, a two-way ANOVA analysis identified biochemicals that exhibited significant main effects for cattle type and in-feed tylosin use and their interactions. A total of 59 and 85 biochemicals were different (P < 0.05) between the cattle type (crossbred vs. Holstein steers) and in-feed tylosin use (tylosin vs. no tylosin), respectively. Succinate, an intermediate of lactate fermentation by some bacterial species, was one of the top 30 biochemicals that differentiated the four treatment groups. A number of lysophospholipids, indicative of bacterial and host cell membrane lyses, were identified in the purulent materials. In conclusion, to our knowledge this is the first report on the metabolome of liver abscess purulent materials and several biochemicals identified were related to metabolic activities of the bacterial community, particularly F. necrophorum and T. pyogenes. Biochemicals unique to liver abscesses that appear in the blood may serve as biomarkers and be of diagnostic value to detect liver abscesses of cattle before slaughter.


Liver abscesses in feedlot cattle, a consequence of feeding a diet of high-grain and low-roughage, are a mixed bacterial infection with Fusobacterium necrophorum, a ruminal bacterium as the primary causative agent. Cattle with liver abscesses do not exhibit clinical signs and the abscesses are detected only at slaughter. The study analyzed purulent materials of liver abscesses of feedlot cattle collected at slaughter for biochemical molecules. A total of 759 biochemicals were identified and a majority belonged to biochemical classes of lipids and amino acids and their metabolites. Biochemicals unique to liver abscesses that enter blood circulation may have the potential to be used as biomarkers in cattle with liver abscess before slaughter.


Subject(s)
Cattle Diseases , Liver Abscess , Cattle , Animals , Tylosin , Anti-Bacterial Agents , Liver Abscess/veterinary , Diet/veterinary , Bacteria , Metabolome , Carbohydrates , Lipids , Animal Feed/analysis , Cattle Diseases/microbiology
6.
Vet Clin North Am Food Anim Pract ; 38(3): 335-346, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36243456

ABSTRACT

Liver abscesses are a bacterial infection, which occurs because of entry, via portal vein, of pyogenic bacteria into the hepatic parenchyma. Liver abscesses are a polymicrobial infection; however, Fusobacterium necrophorum, a ruminal bacterium, is the primary etiologic agent. Ruminal acidosis disrupts the protective barrier function of the ruminal epithelium and facilitates entry and colonization of F. necrophorum in the ruminal wall and subsequent entry into the portal circulation. Virulence factors of F. necrophorum contribute to the evasion of host defense mechanisms and cause tissue damage to set up an infection in the liver. The potential role of the hindgut in pathogenesis remains to be investigated.


Subject(s)
Cattle Diseases , Fusobacterium Infections , Liver Abscess , Animals , Cattle , Cattle Diseases/etiology , Fusobacterium Infections/microbiology , Fusobacterium Infections/veterinary , Fusobacterium necrophorum , Liver Abscess/etiology , Liver Abscess/microbiology , Liver Abscess/veterinary , Rumen/microbiology , Virulence Factors
7.
J Food Prot ; 85(8): 1221-1231, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35653626

ABSTRACT

ABSTRACT: This study was conducted to evaluate the association between a therapeutic dose of tulathromycin for bovine respiratory disease in beef steers and the antimicrobial and multidrug resistance profiles of the gastrointestinal tract commensals Escherichia coli and Enterococcus spp. and the foodborne pathogens Salmonella enterica and Campylobacter spp. isolated from fecal samples. Individual fecal samples were collected on days 0, 14, and 28 from 70 beef steers that were housed in a single pen and had been treated or not treated with tulathromycin. Samples were cultured for bacterial isolation, and isolates were tested for antimicrobial susceptibility with the broth microdilution method to determine the MICs of clinically relevant antimicrobials used in both human and veterinary medicine. Generalized linear mixed effects models were fitted to estimate the prevalence of the bacterial species and the prevalence of resistant isolates over time and between treated and nontreated cattle and of multidrug-resistant isolates. Model-adjusted mean prevalences of E. coli, Enterococcus spp., S. enterica, and Campylobacter spp. were 99.5, 85.9, 1.5, and 17.7%, respectively. The prevalence of erythromycin-resistant Enterococcus spp. was significantly higher on day 14 (59.7%) than on day 28 (22.2%). A higher prevalence of erythromycin-resistant Enterococcus spp. was found in samples from treated (59.3%) than in samples from nontreated (27.6%) animals. Multidrug resistance (three or more antimicrobial classes) was observed in 8.4% of E. coli isolates and 62.7% of Enterococcus isolates. The administration of tulathromycin was significantly associated with an increased prevalence of erythromycin-resistant Enterococcus spp. isolates.


Subject(s)
Anti-Infective Agents , Cattle Diseases , Salmonella enterica , Animals , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Cattle , Cattle Diseases/microbiology , Disaccharides , Drug Resistance, Bacterial , Enterococcus , Erythromycin/pharmacology , Erythromycin/therapeutic use , Escherichia coli , Feces/microbiology , Heterocyclic Compounds , Humans , Microbial Sensitivity Tests
8.
J Anim Sci ; 100(9)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35700748

ABSTRACT

Age and diet are among the factors that influence the community composition of the fecal microbiome. Additionally, antimicrobial use can alter the composition of bacterial communities. An 86-d study with finisher pigs aimed to evaluate age-related dynamics (day 98 to 177 of age), effects of types and levels of dietary fiber, and injectable antimicrobials on the fecal microbiome and antimicrobial resistance (AMR) was conducted. A total of 287 pigs, housed in 36 pens, with 7 to 8 pigs per pen, fed a corn grain and soybean meal-based basal diet, formulated to contain 8.7% neutral detergent fiber (NDF), were randomly assigned to one of three treatments: 1) basal diet with no supplement, 2) basal diet supplemented with 20% distillers dried grains with solubles (DDGS) formulated to contain 13.6% NDF, or 3) basal diet supplemented with 14.5% sugar beet pulp (SBP) formulated to contain 13.6% NDF. Five finisher pigs from each treatment group were selected randomly, and fecal samples were collected on days 98, 110, 144, and 177 of age. In addition, fecal samples were collected from pigs that were injected intramuscularly ceftiofur hydrochloride or penicillin G on days 1 and 3 along with pen-mate-untreated controls on day 1. Fecal samples were subjected to 16S rRNA amplicon-based microbiome analysis and culture methods to quantify the abundance of total AMR coliforms and enterococci populations. The alpha-diversity, such as species richness, increased with age, and the overall bacterial composition changed with age (P =0.001) and diet (P = 0.001). Diet-associated shifts in the specific bacterial taxa were observed. The richness, diversity, and evenness of bacterial taxa did not differ between pigs that were injected with ceftiofur vs. their untreated pen mates or by dietary treatments but differed in pigs that received penicillin G injection. Both antimicrobial treatments contributed to changes in the overall fecal bacterial composition at the genus level. Collectively, the data demonstrate that both age and the diet (control vs. DDGS-, control vs. SBP-, or DDGS- vs. SBP-based diets) were associated with the overall bacterial community composition, and the impact of age on variations in fecal microbiome composition was greater than the diet. Antibiotic treatment had minimal effect on bacterial diversity and relative abundance of taxa. Furthermore, diets and antimicrobial treatment had minimal impact on the overall counts of AMR coliforms and enterococci populations in feces.


Bacterial communities in the gut and the feces are strongly influenced by a number of factors, particularly the age of the animal and the diet. In addition, antibiotic administration routinely used to treat bacterial diseases can also affect the community composition. A study with finisher pigs was conducted to evaluate age-related changes, effects of types­distiller's dried grains with solubles (DGGS) or sugar beet pulp (SBP)­and levels of dietary fiber, and injectable antibiotics on the fecal bacterial composition and antibiotic resistance in fecal bacteria. Fecal samples were collected from five pigs in each of the three dietary treatment groups, control diet with no supplement or supplemented with DDGS or SBP, on days 98, 110, 144, and 177 of age and on days 1 and 3 after the first injection of antibiotics, ceftiofur or penicillin G. Samples were analyzed to identify the bacterial community composition and prevalence of antibiotic resistance in fecal bacteria. Data generated suggested that the overall bacterial composition changed with age and diet, and age appeared to have a greater impact than diet. Antibiotics had only a modest impact on the bacterial community and had minimum impact on antibiotic resistance of fecal bacteria.


Subject(s)
Animal Nutritional Physiological Phenomena , Microbiota , Animal Feed/analysis , Animals , Anti-Bacterial Agents/pharmacology , Detergents , Dietary Fiber/analysis , Drug Resistance, Bacterial , Feces/chemistry , RNA, Ribosomal, 16S , Sugars , Swine , Zea mays
9.
J Anim Sci ; 100(3)2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35150575

ABSTRACT

Enterococcus faecium is one of the more commonly used bacterial species as a probiotic in animals. The organism, a common inhabitant of the gut of animals and humans, is a major nosocomial pathogen responsible for a variety infections in humans and sporadic infections in animals. In swine and cattle, E. faecium-based probiotic products are used for growth promotion and gut functional and health benefits. The objective of this study was to utilize whole genome sequence-based analysis to assess virulence potential, detect antimicrobial resistance genes, and analyze phylogenetic relationships of E. faecium strains from commercial swine and cattle probiotics. Genomic DNA extracted from E. faecium strains, isolated from commercial probiotic products of swine (n = 9) and cattle (n = 13), were sequenced in an Illumina MiSeq platform and analyzed. Seven of the nine swine strains and seven of the 13 cattle strains were identified as Enterococcus lactis, and not as E. faecium. None of the 22 probiotic strains carried major virulence genes required to initiate infections, but many carried genes involved in adhesion to host cells, which may benefit the probiotic strains to colonize and persist in the gut. Strains also carried genes encoding resistance to a few medically important antibiotics, which included aminoglycosides [aac(6')-Ii, aph(3')-III, ant(6)-Ia], macrolide, lincosamide and streptogramin B (msrC), tetracyclines [tet(L) and tet(M)], and phenicols [cat-(pc194)]. The comparison of the genotypic to phentypic AMR data showed presence of both related and unrelated genes in the probiotic strains. Swine and cattle probiotic E. faecium strains belonged to diverse sequence types. Phylogenetic analysis of the probiotic strains, and strains of human (n = 29), swine (n = 4), and cattle (n = 4) origin, downloaded from GenBank, indicated close clustering of strains belonging to the same species and source, but a few swine and cattle probiotic strains clustered closely with other cattle and human fecal strains. In conclusion, the absence of major virulence genes characteristic of the clinical E. faecium strains suggests that these probiotic strains are unlikely to initiate opportunistic infection. However, the carriage of AMR genes to medically important antibiotics and close clustering of the probiotic strains with other human and cattle fecal strains suggests that probiotic strains may pose risk to serve as a source of transmitting AMR genes to other gut bacteria.


Probiotics, also called direct-fed microbials, are widely used in swine and cattle production systems, as an alternative for antibiotics. The benefits of feeding probiotic products include growth promotion and gut functional benefits. One of the more common bacterial species used in swine and cattle commercial probiotic products is Enterococcus faecium. The species is also a member of the normal flora of hindgut of humans and animals. In recent years, the species has emerged as a major hospital-acquired infection in humans, mainly because of the propensity to become resistant to antibiotics. In the United States, the species is considered as generally recognized as safe. In this study, the virulence and antimicrobial resistance genes profiles of 9 and 13 E. faecium strains isolated from commercial swine and cattle probiotics, respectively, were assessed by sequencing the whole genome DNA. The analysis indicated that 14 of 22 strains were Enterococcus lactis, and not E. faecium. The absence of major virulence genes characteristic of the clinical E. faecium strains suggests that the strains are unlikely to initiate opportunistic infection. However, the carriage of genes that confer resistance to medically important antibiotics suggests that probiotic strains may pose risk as a source of antimicrobial resistance genes to other bacteria.


Subject(s)
Anti-Infective Agents , Enterococcus faecium , Probiotics , Animals , Anti-Bacterial Agents/pharmacology , Cattle , Enterococcus faecium/genetics , Microbial Sensitivity Tests/veterinary , Phylogeny , Probiotics/pharmacology , Sequence Analysis/veterinary , Swine , Virulence/genetics
10.
J Food Prot ; 85(4): 701-705, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35076692

ABSTRACT

ABSTRACT: Following removal of hides and viscera during beef processing, carcasses are inspected for tissue adhesions that can affect meat quality or harbor bacteria. Carcasses with pleural or abdominal adhesions may be diverted from the production line for manual excision and then returned to the line. No published data indicate whether adhesion excision is associated with bacterial contamination. Therefore, our objective was to determine the presence and concentration of generic Escherichia coli and non-E. coli coliforms from the internal and external surfaces of carcasses that were, or were not, diverted for adhesion excision. During 9 processing days over a 4-month period in a large commercial beef processing facility, 1,738 carcass sponge samples from 2,730 cm2 areas on both the internal and the external surfaces of carcasses with and without tissue adhesions were collected. Coliforms and E. coli were cultured and enumerated using Petrifilm procedures, and data were analyzed with mixed models. Coliforms were present at higher concentrations than E. coli, and prevalence and mean log concentrations of both coliforms and E. coli were significantly higher for samples from the external than from the internal surfaces of carcasses. However, differences in prevalence and concentration of coliforms between external and internal surfaces varied significantly based on whether carcasses had adhesions excised. The difference was greatest for coliforms present on the external (2.06 log CFU/100 cm2) versus the internal (0.93 log CFU/100 cm2) carcass surfaces without adhesions, whereas the difference in concentrations from the external (1.80 log CFU/100 cm2) and the internal (1.31 log CFU/100 cm2) surfaces of carcasses with adhesions was not as large. These results indicate that surveillance of carcass bacteria may be affected by whether the external versus the internal surfaces are sampled and whether carcasses are diverted for excision of adhesions.


Subject(s)
Escherichia coli , Meat , Abattoirs , Animals , Bacteria , Cattle , Colony Count, Microbial , Food Contamination , Food Handling/methods , Food Microbiology , Meat/microbiology , Tissue Adhesions
11.
Transl Anim Sci ; 6(4): txac154, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36601062

ABSTRACT

Monensin and virginiamycin are included in beef cattle finishing diets as prophylaxis to minimize the incidence of ruminal acidosis and liver abscesses. Due to different and probably complementary modes of action, this study aimed to determine the effects of a combination of monensin and virginiamycin, both included in the diet at recommended doses, on ruminal health, the occurrence of liver abscesses, and growth performance of feedlot-finished cattle. One hundred and forty-four steers (6 animals/pen) were fed 1 of 3 corn-based finishing diets containing 30 mg of monensin (MN), 25 mg of virginiamycin (VM), or 30 and 25 mg of monensin and virginiamycin (MN + VM), respectively, per kilogram of dry matter. Ruminal pH probes were inserted into two animals per pen and set to record pH every 10 min. On d 100, animals were slaughtered, and rumens and livers were recovered, on which occurrence and degree of ruminal damage, prevalence and number of liver abscesses, and liver scores (A-: livers with no more than two small abscesses; A+: livers with at least one large abscess or more than four medium abscesses; A: any other abscessed liver) were determined. Simultaneous inclusion of monensin and virginiamycin resulted in a 4.3% decrease (P < 0.04) in dry matter intake (DMI; 8.8, 9.2, and 9.2 ± 0.19 kg/d for MN + VM, MN, and VM-fed animals, respectively) and similar (P > 0.13) average daily body weight gain (ADG; 1.49 ± 0.021 kg/d) and hot carcass weight (HCW; 269 ± 1.7 kg), compared with feeding diets containing one additive or the other. Therefore, in terms of ADG, a 9.4% improvement (P < 0.01) in feed efficiency was observed in MN + VM-fed animals. Backfat thickness (5.6 ± 0.08 mm) and ribeye area (69.9 ± 0.53 cm2) remained unaffected (P ≥ 0.74), as well as the minimum (4.98 ± 0.047), mean (6.11 ± 0.037), and maximum ruminal pH (7.23 ± 0.033) values and the time (125 ± 22.3 min/d), area (57.67 ± 12.383 pH × h), and episodes (22 ± 3.8 bouts) of pH below 5.6 (P ≥ 0.12). Overall, prevalence (24 ± 3.4%) and the number of liver abscesses (1.6 ± 0.14 abscesses/abscessed liver), liver scores (20 ± 3.1% of A- and 4 ± 1.8% of A livers), and prevalence (67 ± 3.5%) and degree of damage to the ruminal epithelium (2.5 ± 0.22% affected surface) were similar (P ≥ 0.18) across treatments; however, the occurrence of ruminal lesions tended (P ≤ 0.07) to be associated with that of liver abscesses and reduced ADG when feeding monensin alone.

12.
Food Microbiol ; 99: 103821, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34119106

ABSTRACT

Shiga toxin-producing E. coli (STEC) are major foodborne pathogens. While many studies have focused on the "top-7 STEC", little is known for minor serogroups. A total of 284 non-top-7 STEC strains isolated from cattle feces were subjected to whole-genome sequencing (WGS) to determine the serotypes, the presence of virulence genes and antimicrobial resistance (AMR) determinants. Nineteen typeable and three non-typeable serotypes with novel O-antigen loci were identified. Twenty-one AMR genes and point mutations in another six genes that conferred resistance to 10 antimicrobial classes were detected, as well as 46 virulence genes. The distribution of 33 virulence genes and 15 AMR determinants exhibited significant differences among serotypes (p < 0.05). Among all strains, 81.7% (n = 232) and 14.1% (n = 40) carried stx2 and stx1 only, respectively; only 4.2% (n = 12) carried both. Subtypes stx1a, stx1c, stx2a, stx2c, stx2d, and stx2g were identified. Forty-six strains carried eae and stx2a and therefore had the potential cause severe diseases; 47 strains were genetically related to human clinical strains inferred from a pan-genome phylogenetic tree. We were able to demonstrate the utility of WGS as a surveillance tool to characterize the novel serotypes, as well as AMR and virulence profiles of uncommon STEC that could potentially cause human illness.


Subject(s)
Cattle Diseases/microbiology , Drug Resistance, Bacterial , Escherichia coli Infections/veterinary , Shiga-Toxigenic Escherichia coli/isolation & purification , Animals , Anti-Bacterial Agents/pharmacology , Cattle , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Feces/microbiology , Phylogeny , Serogroup , Shiga-Toxigenic Escherichia coli/drug effects , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/pathogenicity , Virulence , Whole Genome Sequencing
14.
Sci Rep ; 11(1): 8601, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33883564

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) are foodborne pathogens that cause illnesses in humans ranging from mild to hemorrhagic enteritis with complications of hemolytic uremic syndrome and even death. Cattle are a major reservoir of STEC, which reside in the hindgut and are shed in the feces, a major source of food and water contaminations. Seven serogroups, O26, O45, O103, O111, O121, O145 and O157, called 'top-7', are responsible for the majority of human STEC infections in North America. Additionally, 151 serogroups of E. coli are known to carry Shiga toxin genes (stx). Not much is known about fecal shedding and prevalence and virulence potential of STEC other than the top-7. Our primary objectives were to identify serogroups of STEC strains, other than the top-7, isolated from cattle feces and subtype stx genes to assess their virulence potential. Additional objective was to develop and validate a novel multiplex PCR assay to detect and determine prevalence of six serogroups, O2, O74, O109, O131, O168, and O171, in cattle feces. A total of 351 strains, positive for stx gene and negative for the top-7 serogroups, isolated from feedlot cattle feces were used in the study. Of the 351 strains, 291 belonged to 16 serogroups and 60 could not be serogrouped. Among the 351 strains, 63 (17.9%) carried stx1 gene and 300 (82.1%) carried stx2, including 12 strains positive for both. The majority of the stx1 and stx2 were of stx1a (47/63; 74.6%) and stx2a subtypes (234/300; 78%), respectively, which are often associated with human infections. A novel multiplex PCR assay developed and validated to detect six serogroups, O2, O74, O109, O131, O168, and O171, which accounted for 86.9% of the STEC strains identified, was utilized to determine their prevalence in fecal samples (n = 576) collected from a commercial feedlot. Four serogroups, O2, O109, O168, and O171 were identified as the dominant serogroups prevalent in cattle feces. In conclusion, cattle shed in the feces a number of STEC serogroups, other than the top-7, and the majority of the strains isolated possessed stx2, particularly of the subtype 2a, suggesting their potential risk to cause human infections.


Subject(s)
Escherichia coli Infections/epidemiology , Escherichia coli Infections/veterinary , Escherichia coli Proteins/genetics , Shiga Toxin/genetics , Shiga-Toxigenic Escherichia coli/genetics , Animals , Cattle , Escherichia coli Infections/microbiology , Feces/microbiology , North America , Prevalence , Serogroup , Virulence/genetics
15.
J Anim Sci ; 99(4)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33693672

ABSTRACT

Liver abscesses in feedlot cattle are polymicrobial infections. Culture-based studies have identified Fusobacterium necrophorum as the primary causative agent, but a number of other bacterial species are frequently isolated. The incidence of liver abscesses is highly variable and is affected by a number of factors, including cattle type. Holstein steers raised for beef production have a higher incidence than crossbred feedlot cattle. Tylosin is the commonly used antimicrobial feed additive to reduce the incidence of liver abscesses. The objective of this study was to utilize 16S ribosomal RNA amplicon sequence analyses to analyze the bacterial community composition of purulent material of liver abscesses of crossbred cattle (n = 24) and Holstein steers (n = 24), each fed finishing diet with or without tylosin. DNA was extracted and the V3 and V4 regions of the 16S rRNA gene were amplified, sequenced, and analyzed. The minimum, mean, and maximum sequence reads per sample were 996, 177,070, and 877,770, respectively, across all the liver abscess samples. Sequence analyses identified 5 phyla, 14 families, 98 genera, and 102 amplicon sequence variants (ASV) in the 4 treatment groups. The dominant phyla identified were Fusobacteria (52% of total reads) and Proteobacteria (33%). Of the top 25 genera identified, 17 genera were Gram negative and 8 were Gram positive. The top 3 genera, which accounted for 75% of the total reads, in the order of abundance, were Fusobacterium, Pseudomonas, and Bacteroides. The relative abundance, expressed as percent of total reads, of phyla, family, and genera did not differ (P > 0.05) between the 4 treatment groups. Generic richness and evenness, determined by Shannon-Weiner and Simpson's diversity indices, respectively, did not differ between the groups. The UniFrac distance matrices data revealed no clustering of the ASV indicating variance between the samples within each treatment group. Co-occurrence network analysis at the genus level indicated a strong association of Fusobacterium with 15 other genera, and not all of them have been previously isolated from liver abscesses. In conclusion, the culture-independent method identified the bacterial composition of liver abscesses as predominantly Gram negative and Fusobacterium as the dominant genus, followed by Pseudomonas. The bacterial community composition did not differ between crossbred and Holstein steers fed finishing diets with or without tylosin.


Subject(s)
Cattle Diseases , Liver Abscess , Animal Feed/analysis , Animals , Cattle , Diet/veterinary , Liver Abscess/veterinary , RNA, Ribosomal, 16S/genetics , Tylosin
16.
Anaerobe ; 69: 102344, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33588043

ABSTRACT

Fusobacterium necrophorum, a Gram-negative anaerobe, is the primary etiologic agent of liver abscesses of beef cattle. The bacterium, a member of the microbial community of the rumen, travels to the liver via portal circulation to cause abscesses. The severity of liver abscesses vary from mild with one or two small abscesses to severe with medium to large multiple abscesses. Leukotoxin, a secreted protein, is the critical virulence factor involved in the infection. Our objective was to compare leukotoxin production between strains of F. necrophorum isolated from mild and severe liver abscesses collected from slaughtered cattle. The quantification of leukotoxin was based on assays to measure cytotoxicity and protein antigen concentration. One-hundred strains, 50 from mild and 50 from severe abscesses, were utilized in the study. Cell-free supernatants were prepared from cultures grown in anaerobic broth at 9 and 24 h incubations. The leukotoxic activity was quantified by measuring cytotoxicity based on the release of lactic dehydrogenase from bovine lymphocyte cells, BL3, treated with the culture supernatant. Leukotoxin protein concentration was quantified by a sandwich ELISA assay with a leukotoxin-specific monoclonal antibody as the capture antibody. The leukotoxin activity and concentration were highly variable among the strains within each severity of liver abscesses. Although the leukotoxic activity was unaffected by incubation time, leukotoxin protein concentration was consistently higher at 24 h compared to 9 h incubation. Strains from severe liver abscesses had significantly higher leukotoxic activity and higher protein concentration compared to strains from mild liver abscesses (P < 0.0001) at both 9 and 24 h culture supernatants. Across all strains, the correlation coefficients between leukotoxic activity and leukotoxin concentration at 9 and 24 h were 0.14 (P = 0.17) and 0.47 (P < 0.0001), respectively. In conclusion, strains isolated from severe liver abscesses had significantly higher leukotoxic activities and leukotoxin protein concentrations compared to strains isolated from mild liver abscesses.


Subject(s)
Exotoxins/biosynthesis , Fusobacterium Infections/microbiology , Fusobacterium Infections/physiopathology , Fusobacterium necrophorum/isolation & purification , Fusobacterium necrophorum/metabolism , Liver Abscess/microbiology , Liver Abscess/physiopathology , Animals , Cattle , Cattle Diseases/microbiology , Cattle Diseases/physiopathology , Fusobacterium necrophorum/genetics , Genetic Variation , Genotype , Severity of Illness Index
17.
J Food Prot ; 84(1): 169-180, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33411931

ABSTRACT

ABSTRACT: Shiga toxin-producing Escherichia coli (STEC) are major foodborne human pathogens that cause mild to hemorrhagic colitis, which could lead to complications of hemolytic uremic syndrome. Seven serogroups, O26, O45, O103, O111, O121, O145, and O157, account for the majority of the STEC illnesses in the United States. Shiga toxins 1 and 2, encoded by stx1 and stx2, respectively, and intimin, encoded by eae gene, are major virulence factors. Cattle are a major reservoir of STEC, but swine also harbor them in the hindgut and shed STEC in the feces. Our objectives were to use a culture method to isolate and identify major and minor serogroups of STEC in finisher pig feces. Shiga toxin genes were subtyped to assess public health implications of STEC. Fecal samples (n = 598) from finisher pigs, collected from 10 pig flows, were enriched in E. coli broth and tested for stx1, stx2, and eae by a multiplex PCR (mPCR) assay. Samples positive for stx1 or stx2 gene were subjected to culture methods, with or without immunomagnetic separation and plating on selective or nonselective media, for isolation and identification of stx-positive isolates. The culture method yielded a total of 178 isolates belonging to 23 serogroups. The three predominant serogroups were O8, O86, and O121. The 178 STEC strains included 26 strains with stx1a and 152 strains with stx2e subtypes. Strains with stx1a, particularly in association with eae (O26 and O103), have the potential to cause severe human infections. All stx2-positive isolates carried the subtype stx2e, a subtype that causes edema disease in swine, but is rarely involved in human infections. Several strains were also positive for genes that encode for enterotoxins, which are involved in neonatal and postweaning diarrhea in swine. In conclusion, our study showed that healthy finisher pigs harbored and shed several serogroups of E. coli carrying virulence genes involved in neonatal diarrhea, postweaning diarrhea, and edema disease, but prevalence of STEC of public health importance was low.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Shiga-Toxigenic Escherichia coli , Animals , Cattle , Escherichia coli Infections/veterinary , Escherichia coli Proteins/genetics , Feces , Public Health , Serogroup , Shiga Toxin/genetics , Shiga-Toxigenic Escherichia coli/genetics , Swine
18.
Foodborne Pathog Dis ; 18(1): 16-23, 2021 01.
Article in English | MEDLINE | ID: mdl-32898446

ABSTRACT

The objective of this study was to evaluate the effectiveness of a direct-fed microbial (DFM) product in reducing fecal shedding of Escherichia coli O157:H7 in finishing commercial feedlot cattle in Kansas (KS) and Nebraska (NE). Utilizing a randomized complete block design within the feedlot (KS, n = 1; NE, n = 1), cattle were randomly allocated to 20 pens, grouped in blocks of two based on allocation date, and then, within the block, randomly assigned to a treatment group (DFM or negative control). The DFM product was included in the diet at a targeted daily dose of 1 × 109 colony-forming units (CFU) of the Lactobacillus acidophilus and Lactobacillus casei combination per animal for at least 60 d before sampling. Feedlots were sampled for four consecutive weeks; weekly sampling consisted of collecting 20 pen floor fecal samples per pen. Fecal samples were subjected to culture-based methods for detection and isolation of E. coli O157, and positive samples were quantified using real-time polymerase chain reaction. Primary outcomes of interest were fecal prevalence of E. coli O157:H7 and E. coli O157 supershedding (≥104 CFU/g of feces) prevalence. Data for each feedlot were analyzed at the pen level using mixed models accounting for the study design features. Model-adjusted mean E. coli O157:H7 fecal prevalence estimates (standard error of the mean [SEM]) for DFM and control groups were 8.2% (SEM = 2.2%) and 9.9% (SEM = 2.5%) in KS and 14.6% (SEM = 2.8%) versus 14.3% (SEM = 2.6%) in NE; prevalence did not differ significantly between treatment groups at either site (KS, p = 0.51; NE, p = 0.92). Mean E. coli O157 supershedding prevalence estimates for DFM and control groups were 2.2% (SEM = 0.7%) versus 1.8% (SEM = 0.7%) in KS (p = 0.66) and 6.7% (SEM = 1.5%) versus 3.2% (SEM = 1.0%) in NE (p = 0.04). In conclusion, administering the DFM product in the finishing diet of feedlot cattle did not significantly reduce E. coli O157:H7 fecal prevalence or supershedding prevalence in study pens at either commercial feedlot.


Subject(s)
Cattle Diseases/prevention & control , Escherichia coli Infections/prevention & control , Escherichia coli O157 , Lacticaseibacillus casei , Lactobacillus acidophilus , Probiotics/administration & dosage , Animal Feed/microbiology , Animals , Bacterial Shedding , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/microbiology , Escherichia coli Infections/epidemiology , Escherichia coli Infections/veterinary , Feces/microbiology , Kansas/epidemiology , Nebraska/epidemiology
20.
Article in English | MEDLINE | ID: mdl-32850480

ABSTRACT

Escherichia coli carrying prophage with genes that encode for Shiga toxins are categorized as Shiga toxin-producing E. coli (STEC) pathotype. Illnesses caused by STEC in humans, which are often foodborne, range from mild to bloody diarrhea with life-threatening complications of renal failure and hemolytic uremic syndrome and even death, particularly in children. As many as 158 of the total 187 serogroups of E. coli are known to carry Shiga toxin genes, which makes STEC a major pathotype of E. coli. Seven STEC serogroups, called top-7, which include O26, O45, O103, O111, O121, O145, and O157, are responsible for the majority of the STEC-associated human illnesses. The STEC serogroups, other than the top-7, called "non-top-7" have also been associated with human illnesses, more often as sporadic infections. Ruminants, particularly cattle, are principal reservoirs of STEC and harbor the organisms in the hindgut and shed in the feces, which serves as a major source of food and water contaminations. A number of studies have reported on the fecal prevalence of top-7 STEC in cattle feces. However, there is paucity of data on the prevalence of non-top-7 STEC serogroups in cattle feces, generally because of lack of validated detection methods. The objective of our study was to develop and validate 14 sets of multiplex PCR (mPCR) assays targeting serogroup-specific genes to detect 137 non-top-7 STEC serogroups previously reported to be present in cattle feces. Each assay included 7-12 serogroups and primers were designed to amplify the target genes with distinct amplicon sizes for each serogroup that can be readily identified within each assay. The assays were validated with 460 strains of known serogroups. The multiplex PCR assays designed in our study can be readily adapted by most laboratories for rapid identification of strains belonging to the non-top-7 STEC serogroups associated with cattle.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Shiga-Toxigenic Escherichia coli , Animals , Cattle , Escherichia coli Infections/diagnosis , Escherichia coli Infections/veterinary , Escherichia coli Proteins/genetics , Feces , Multiplex Polymerase Chain Reaction , Serogroup , Shiga Toxin/genetics , Shiga-Toxigenic Escherichia coli/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...